1
|
Osei I, Schmidt-Chanasit J, Licciardi PV, Secka O, D'Alessandro U, Salaudeen R, Sarwar G, Clarke E, Mohammed NI, Nguyen C, Greenwood B, Jansen S, Mackenzie GA. Immunogenicity of yellow fever vaccine co-administered with 13-valent pneumococcal conjugate vaccine in rural Gambia: A cluster-randomised trial. Vaccine 2025; 47:126712. [PMID: 39798436 PMCID: PMC11797555 DOI: 10.1016/j.vaccine.2025.126712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 01/04/2025] [Accepted: 01/05/2025] [Indexed: 01/15/2025]
Abstract
INTRODUCTION Because booster doses of pneumococcal conjugate vaccine (PCV) may be given at a similar time to yellow fever vaccine (YF), it is important to assess the immune response to YF when co-administered with PCV. This has been investigated during a reduced-dose PCV trial in The Gambia. METHODS In this phase 4, parallel-group, cluster-randomized trial, healthy infants aged 0-10 weeks were randomly allocated to receive either a two-dose schedule of PCV13 with a booster dose co-administered with YF vaccine at age 9 months (1 + 1 co-administration) or YF vaccine administered separately at age 10 months (1 + 1 separate) or the standard three early doses of PCV13 with YF vaccine at age 9 months (3 + 0 separate). Blood samples were collected 28-35 days post-vaccination and YF neutralizing antibody (NA) titres were measured. Proportions with seroprotective YF NA titres ≥ 1:8 were calculated with 95 % confidence intervals (CI). Non-inferiority was demonstrated if the lower limit of the CI for the difference in proportions between the co-administration and separate groups was greater than - 10 %. RESULTS Forty-eight, 66, and 98 participants enrolled in 3 + 0 separate, 1 + 1 co-administration, and 1 + 1 separate groups respectively had NA results. Per protocol analysis of the 3 + 0 separate, 1 + 1 co-administration, 1 + 1 separate, and the combined 1 + 1 separate and 3 + 0 separate groups found that 81 %, 85 %, 92 %, and 88 % of participants respectively had YF NA titres ≥1:8. Results were similar with analysis by intention-to-treat. The difference in proportions comparing 1 + 1 co-administration and 1 + 1 separate groups was -7 % (95 % CI, -18 % to 3 %). The difference between 1 + 1 co-administration and 3 + 0 separate groups was 4 % (95 % CI, -10 % to 15 %). There was no statistical difference in the YF seroresponse when the YF vaccine was co-administered with PCV or administered separately. CONCLUSIONS No evidence was found of the non-inferiority of the seroresponse to YF vaccine when co-administered with PCV13. The levels of YF NA attaining seroprotection (NT ≥1:8) were high in all groups. PCV13 co-administered with YF vaccine at 9 months does not affect seroresponse to YF vaccine. http://www.isrctn.org/ - ISRCTN72821613.
Collapse
Affiliation(s)
- Isaac Osei
- Medical Research Council Unit The Gambia at London School of Hygiene & Tropical Medicine, Banjul, the Gambia; Department of Disease Control, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK.
| | - Jonas Schmidt-Chanasit
- Bernhard Nocht Institute for Tropical Medicine, Department of Arbovirology and Entomology, Hamburg, Germany
| | - Paul V Licciardi
- Murdoch Children's Research Institute, Melbourne, Australia; Department of Paediatrics, University of Melbourne, Australia
| | - Ousman Secka
- Medical Research Council Unit The Gambia at London School of Hygiene & Tropical Medicine, Banjul, the Gambia
| | - Umberto D'Alessandro
- Medical Research Council Unit The Gambia at London School of Hygiene & Tropical Medicine, Banjul, the Gambia
| | - Rasheed Salaudeen
- Medical Research Council Unit The Gambia at London School of Hygiene & Tropical Medicine, Banjul, the Gambia
| | - Golam Sarwar
- Medical Research Council Unit The Gambia at London School of Hygiene & Tropical Medicine, Banjul, the Gambia
| | - Ed Clarke
- Medical Research Council Unit The Gambia at London School of Hygiene & Tropical Medicine, Banjul, the Gambia
| | - Nuredin I Mohammed
- Medical Research Council Unit The Gambia at London School of Hygiene & Tropical Medicine, Banjul, the Gambia
| | - Cattram Nguyen
- Murdoch Children's Research Institute, Melbourne, Australia; Department of Paediatrics, University of Melbourne, Australia
| | - Brian Greenwood
- Department of Disease Control, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK
| | - Stephanie Jansen
- Bernhard Nocht Institute for Tropical Medicine, Department of Arbovirology and Entomology, Hamburg, Germany; University of Hamburg, Faculty of Mathematics, Informatics and Natural Sciences, Hamburg, Germany
| | - Grant A Mackenzie
- Medical Research Council Unit The Gambia at London School of Hygiene & Tropical Medicine, Banjul, the Gambia; Department of Disease Control, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK; Murdoch Children's Research Institute, Melbourne, Australia; Department of Paediatrics, University of Melbourne, Australia
| |
Collapse
|
2
|
Ferrara P, Losa L, Mantovani LG, Ambrosioni J, Agüero F. Humoral immunogenicity of primary yellow fever vaccination in infants and children: a systematic review, meta-analysis and meta-regression. J Travel Med 2024; 31:taae039. [PMID: 38438165 DOI: 10.1093/jtm/taae039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 02/26/2024] [Accepted: 02/28/2024] [Indexed: 03/06/2024]
Abstract
BACKGROUND Vaccination plays a critical role in mitigating the burden associated with yellow fever (YF). However, there is a lack of comprehensive evidence on the humoral response to primary vaccination in the paediatric population, with several questions debated, including the response when the vaccine is administered at early ages, the effect of co-administration with other vaccines, the duration of immunity and the use of fractional doses, among others. This study summarizes the existing evidence regarding the humoral response to primary YF vaccination in infants and children. METHODS Studies on the humoral response to primary YF vaccination in children aged 12 years or younger were reviewed. The humoral vaccine response rate (VRR), i.e. the proportion of children who tested positive for vaccine-induced YF-specific neutralizing antibodies, was pooled through random-effects meta-analysis and categorized based on the time elapsed since vaccination. Subgroup, meta-regression and sensitivity analyses were performed. RESULTS A total of 33 articles met the inclusion criteria, with all but one conducted in countries where YF is endemic. A total of 14 028 infants and children entered this systematic review. Within three months following vaccination, the pooled VRR was 91.9% (95% CI 89.8-93.9). A lower VRR was observed with the 17DD vaccine at the meta-regression analysis. No significant differences in immunogenicity outcomes were observed based on age, administration route, co-administration with other vaccines, or fractional dosing. Results also indicate a decline in VRR over time. CONCLUSIONS Primary YF vaccination effectively provides humoral immunity in paediatric population. However, humoral response declines over time, and this decline is observable after the first 18 months following vaccination. A differential response according to the vaccine substrain was also observed. This research has valuable implications for stimulating further research on the primary YF vaccination in infants and children, as well as for informing future policies.
Collapse
Affiliation(s)
- Pietro Ferrara
- Center for Public Health Research (CESP), University of Milan-Bicocca, Monza, Italy
- Laboratory of Public Health, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Lorenzo Losa
- Center for Public Health Research (CESP), University of Milan-Bicocca, Monza, Italy
| | - Lorenzo G Mantovani
- Center for Public Health Research (CESP), University of Milan-Bicocca, Monza, Italy
- Laboratory of Public Health, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Juan Ambrosioni
- Infectious Disease Department, School of Medicine, University of Barcelona, Barcelona, Spain
- HIV Unit, Infectious Diseases Service, Hospital Clinic-Fundació de Recerca Clínic Barcelon-IDIBAPS, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - Fernando Agüero
- Unit of Preventive Medicine, Catalan Institute of Oncology, L'Hospitalet de Llobregat, Barcelona, Spain
| |
Collapse
|
3
|
Kling K, Domingo C, Bogdan C, Duffy S, Harder T, Howick J, Kleijnen J, McDermott K, Wichmann O, Wilder-Smith A, Wolff R. Duration of Protection After Vaccination Against Yellow Fever: A Systematic Review and Meta-Analysis. Clin Infect Dis 2022; 75:2266-2274. [PMID: 35856638 PMCID: PMC9761887 DOI: 10.1093/cid/ciac580] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 06/30/2022] [Accepted: 07/13/2022] [Indexed: 01/19/2023] Open
Abstract
The duration of protection after a single dose of yellow fever vaccine is a matter of debate. To summarize the current knowledge, we performed a systematic literature review and meta-analysis. Studies on the duration of protection after 1 and ≥2 vaccine doses were reviewed. Data were stratified by time since vaccination. In our meta-analysis, we used random-effects models. We identified 36 studies from 20 countries, comprising more than 17 000 participants aged 6 months to 85 years. Among healthy adults and children, pooled seroprotection rates after single vaccination dose were close to 100% by 3 months and remained high in adults for 5 to 10 years. In children vaccinated before age 2 years, the seroprotection rate was 52% within 5 years after primary vaccination. For immunodeficient persons, data indicate relevant waning. The extent of waning of seroprotection after yellow fever vaccination depends on age and immune status at primary vaccination.
Collapse
Affiliation(s)
- Kerstin Kling
- Immunization Unit, Department of Infectious Disease Epidemiology, Robert Koch Institute, Berlin, Germany
| | - Cristina Domingo
- Center for International Health Protection, Robert Koch Institute, Berlin, Germany
| | - Christian Bogdan
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Friedrich Alexander Universität (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Steven Duffy
- Kleijnen Systematic Reviews Ltd, York, United Kingdom
| | - Thomas Harder
- Immunization Unit, Department of Infectious Disease Epidemiology, Robert Koch Institute, Berlin, Germany
| | - Jeremy Howick
- Kleijnen Systematic Reviews Ltd, York, United Kingdom
| | - Jos Kleijnen
- Kleijnen Systematic Reviews Ltd, York, United Kingdom
| | | | - Ole Wichmann
- Immunization Unit, Department of Infectious Disease Epidemiology, Robert Koch Institute, Berlin, Germany
| | - Annelies Wilder-Smith
- Heidelberg Institute of Global Health, University of Heidelberg, Heidelberg, Germany.,Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
| | - Robert Wolff
- Kleijnen Systematic Reviews Ltd, York, United Kingdom
| |
Collapse
|
4
|
Rasulova M, Vercruysse T, Paulissen J, Coun C, Suin V, Heyndrickx L, Ma J, Geerts K, Timmermans J, Mishra N, Li LH, Kum DB, Coelmont L, Van Gucht S, Karimzadeh H, Thorn-Seshold J, Rothenfußer S, Ariën KK, Neyts J, Dallmeier K, Thibaut HJ. A High-Throughput Yellow Fever Neutralization Assay. Microbiol Spectr 2022; 10:e0254821. [PMID: 35670599 PMCID: PMC9241659 DOI: 10.1128/spectrum.02548-21] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 05/19/2022] [Indexed: 11/20/2022] Open
Abstract
Quick and accurate detection of neutralizing antibodies (nAbs) against yellow fever is essential in serodiagnosis during outbreaks for surveillance and to evaluate vaccine efficacy in population-wide studies. All of this requires serological assays that can process a large number of samples in a highly standardized format. Albeit being laborious, time-consuming, and limited in throughput, the classical plaque reduction neutralization test (PRNT) is still considered the gold standard for the detection and quantification of nAbs due to its sensitivity and specificity. Here, we report the development of an alternative fluorescence-based serological assay (SNTFLUO) with an equally high sensitivity and specificity that is fit for high-throughput testing with the potential for automation. Finally, our novel SNTFLUO was cross-validated in several reference laboratories and against international WHO standards, showing its potential to be implemented in clinical use. SNTFLUO assays with similar performance are available for the Japanese encephalitis, Zika, and dengue viruses amenable to differential diagnostics. IMPORTANCE Fast and accurate detection of neutralizing antibodies (nAbs) against yellow fever virus (YFV) is key in yellow fever serodiagnosis, outbreak surveillance, and monitoring of vaccine efficacy. Although classical PRNT remains the gold standard for measuring YFV nAbs, this methodology suffers from inherent limitations such as low throughput and overall high labor intensity. We present a novel fluorescence-based serum neutralization test (SNTFLUO) with equally high sensitivity and specificity that is fit for processing a large number of samples in a highly standardized manner and has the potential to be implemented for clinical use. In addition, we present SNTFLUO assays with similar performance for Japanese encephalitis, Zika, and dengue viruses, opening new avenues for differential diagnostics.
Collapse
Affiliation(s)
- Madina Rasulova
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Virology and Chemotherapy, Molecular Vaccinology & Vaccine Discovery, Leuven, Belgium
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Translational Platform Virology and Chemotherapy (TPVC), Leuven, Belgium
| | - Thomas Vercruysse
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Virology and Chemotherapy, Molecular Vaccinology & Vaccine Discovery, Leuven, Belgium
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Translational Platform Virology and Chemotherapy (TPVC), Leuven, Belgium
| | - Jasmine Paulissen
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Virology and Chemotherapy, Molecular Vaccinology & Vaccine Discovery, Leuven, Belgium
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Translational Platform Virology and Chemotherapy (TPVC), Leuven, Belgium
| | - Catherina Coun
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Virology and Chemotherapy, Molecular Vaccinology & Vaccine Discovery, Leuven, Belgium
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Translational Platform Virology and Chemotherapy (TPVC), Leuven, Belgium
| | - Vanessa Suin
- Sciensano, Viral Diseases Service, Scientific Directorate of Infectious Diseases in Humans, Brussels, Belgium
| | - Leo Heyndrickx
- Virology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine Antwerp, Antwerp, Belgium
| | - Ji Ma
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Virology and Chemotherapy, Molecular Vaccinology & Vaccine Discovery, Leuven, Belgium
- Global Virus Network (GVN), Baltimore, Maryland, USA
| | - Katrien Geerts
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Virology and Chemotherapy, Molecular Vaccinology & Vaccine Discovery, Leuven, Belgium
- Global Virus Network (GVN), Baltimore, Maryland, USA
| | - Jolien Timmermans
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Virology and Chemotherapy, Molecular Vaccinology & Vaccine Discovery, Leuven, Belgium
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Translational Platform Virology and Chemotherapy (TPVC), Leuven, Belgium
| | - Niraj Mishra
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Virology and Chemotherapy, Molecular Vaccinology & Vaccine Discovery, Leuven, Belgium
- Global Virus Network (GVN), Baltimore, Maryland, USA
| | - Li-Hsin Li
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Virology and Chemotherapy, Molecular Vaccinology & Vaccine Discovery, Leuven, Belgium
- Global Virus Network (GVN), Baltimore, Maryland, USA
| | - Dieudonné Buh Kum
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Virology and Chemotherapy, Molecular Vaccinology & Vaccine Discovery, Leuven, Belgium
- Global Virus Network (GVN), Baltimore, Maryland, USA
| | - Lotte Coelmont
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Virology and Chemotherapy, Molecular Vaccinology & Vaccine Discovery, Leuven, Belgium
- Global Virus Network (GVN), Baltimore, Maryland, USA
| | - Steven Van Gucht
- Sciensano, Viral Diseases Service, Scientific Directorate of Infectious Diseases in Humans, Brussels, Belgium
| | - Hadi Karimzadeh
- Division of Clinical Pharmacology, University Hospital, LMU Munich, Munich, Germany
- Unit Clinical Pharmacology (EKliP), Helmholtz Center for Environmental Health, Munich, Germany
| | - Julia Thorn-Seshold
- Division of Clinical Pharmacology, University Hospital, LMU Munich, Munich, Germany
- Unit Clinical Pharmacology (EKliP), Helmholtz Center for Environmental Health, Munich, Germany
| | - Simon Rothenfußer
- Division of Clinical Pharmacology, University Hospital, LMU Munich, Munich, Germany
- Unit Clinical Pharmacology (EKliP), Helmholtz Center for Environmental Health, Munich, Germany
| | - Kevin K. Ariën
- Virology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine Antwerp, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Johan Neyts
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Virology and Chemotherapy, Molecular Vaccinology & Vaccine Discovery, Leuven, Belgium
- Global Virus Network (GVN), Baltimore, Maryland, USA
| | - Kai Dallmeier
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Virology and Chemotherapy, Molecular Vaccinology & Vaccine Discovery, Leuven, Belgium
- Global Virus Network (GVN), Baltimore, Maryland, USA
| | - Hendrik Jan Thibaut
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Virology and Chemotherapy, Molecular Vaccinology & Vaccine Discovery, Leuven, Belgium
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Translational Platform Virology and Chemotherapy (TPVC), Leuven, Belgium
| |
Collapse
|
5
|
Guimarães MDCN, Freitas MNO, Sousa AWD, Cunha MACRD, Almada GL, Romano APM, Santos MGDP, Rodrigues GAP, Martins LC, Chiang JO, Casseb LMN. Serological Evidence of Arboviruses in Horses During West Nile Fever Monitoring Surveillance in Southeastern Brazil. FRONTIERS IN TROPICAL DISEASES 2022. [DOI: 10.3389/fitd.2022.881710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Many human arboviruses are also pathogenic for horses, and some of these have emerged recently. A descriptive cross-sectional observational study was conducted to assess the prevalence of West Nile virus (WNV) and other arboviruses among 77 horses on the rural properties of the Espirito Santo state, Brazil. Serum samples were screened for arbovirus-reactive antibodies using the hemagglutination inhibition technique and subsequently a plaque reduction neutralization test for the confirmation of exposure from sera was used to detect heterotypic immune reactions. Overall, the total antibodies against at least one arbovirus of Alphavirus, Flavivirus, and Orthobunyavirus genera were detected in 39 (50.6%) animals. The antibodies to Phlebovirus were not detected in any sample. When the 24 WNV hemagglutination inhibition (HI)-positive samples were tested by the plaque-reduction neutralization test 90%, 9 (32.1%) were positive for WNV antibodies and 14 (50%) for Saint Louis encephalitis virus. Our findings indicate that the region provides ideal conditions for the emergence of arboviruses, reinforcing the need for further surveillance of mosquito-transmitted diseases in domestic animals.
Collapse
|
6
|
Blyth DM, Liang Z, Williams M, Murray CK. Immune interference revisited: Impact of live-attenuated influenza vaccine prior to yellow fever vaccination. Vaccine 2022; 40:961-966. [PMID: 35031146 DOI: 10.1016/j.vaccine.2021.12.031] [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: 11/24/2020] [Revised: 11/12/2021] [Accepted: 12/12/2021] [Indexed: 11/25/2022]
Abstract
BACKGROUND During routine mass live-attenuated influenza vaccination (LAIV) for military personnel, emergent deployment for Ebola humanitarian assistance (OUA) required mass yellow fever vaccination (YF17D), often < 4-weeks recommended timing post-LAIV-triggering concerns for immune interference. We compared YF17D seroconversion rates in personnel who received YF17D as recommended (vaccinated by guidelines [VBG]) to those who received the vaccine outside the recommended timing following LAIV (not vaccinated by guidelines [NVBG]). METHODS OUA deploying personnel who received LAIV simultaneously or before YF17D and had pre- and post-vaccination archived serum were included. VBG was defined as YF17D given concurrently or ≥ 30 days post-LAIV and NVBG as YF17D given 1-29 days post-LAIV. YF17D seroresponse was determined by screening ELISA confirmed with plaque reduction neutralization testing (PRNT) on positive ELISA samples. Exclusion criteria were prior YF17D and pre-vaccination YF17D positive PRNT. RESULTS Of the 660 personnel included, 507 were VBG and 153 were NVBG. Median age was 25 years for both groups. Men accounted for 84% of those VBG and 79% NVBG (p = 0.194). Seroconversion rates were 97.8% for VBG and 95.4% for NVBG (p = 0.15). Multivariate logistic regression revealed that YF17D on days 7-21 post-LAIV (adjusted odds ratio [aOR] 0.304, p = 0.017; confidence interval [CI] 0.114-0.810) and female sex (aOR 0.330, p = 0.026; CI 0.124-0.879) were associated with decreased seroresponse. CONCLUSIONS In this healthy, young adult military population, there was high seroconversion following YF17D when administered simultaneously and at various time points after LAIV. Slight decreases in seroresponse were seen in women and those receiving YF17D 7-21 days following LAIV.
Collapse
Affiliation(s)
- Dana M Blyth
- Infectious Disease Service, Department of Medicine, Brooke Army Medical Center, 3551 Roger Brooke Dr, JBSA Ft Sam Houston, TX 78234, USA; Infectious Disease Service, Directorate of Medicine, Walter Reed National Military Medical Center, 8960 Brown Drive, Bethesda, MD 20889, USA; Uniformed Services University of the Health Sciences, Bethesda, MA 20889, USA.
| | - Zhaodong Liang
- Henry M. Jackson Foundation and Viral and Rickettsial Diseases Department, Naval Medical Research Center, 503 Robert Grant Ave, Silver Spring, MD, USA
| | - Maya Williams
- Henry M. Jackson Foundation and Viral and Rickettsial Diseases Department, Naval Medical Research Center, 503 Robert Grant Ave, Silver Spring, MD, USA; Infectious Diseases Directorate, Naval Medical Research Center, 503 Robert Grant Ave, Silver Spring, MD, USA
| | - Clinton K Murray
- Uniformed Services University of the Health Sciences, Bethesda, MA 20889, USA; Walter Reed Army Institute for Research, 503 Robert Grant Ave, Silver Spring, MD, USA
| |
Collapse
|
7
|
Zimmermann P, Pollard AJ, Curtis N. What time interval is needed between the administration of live attenuated vaccines? Arch Dis Child 2020; 105:1232-1235. [PMID: 32900760 DOI: 10.1136/archdischild-2020-320091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/28/2020] [Accepted: 07/29/2020] [Indexed: 11/03/2022]
Affiliation(s)
- Petra Zimmermann
- Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland .,Department of Paediatrics, Fribourg Hospital HFR, Fribourg, Switzerland.,Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia.,Infectious Diseases Research Group, Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Nigel Curtis
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia.,Infectious Diseases Research Group, Murdoch Children's Research Institute, Parkville, Victoria, Australia.,Infectious Diseases Unit, The Royal Children's Hospital Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
8
|
Domingo C, Fraissinet J, Ansah PO, Kelly C, Bhat N, Sow SO, Mejía JE. Long-term immunity against yellow fever in children vaccinated during infancy: a longitudinal cohort study. THE LANCET. INFECTIOUS DISEASES 2019; 19:1363-1370. [PMID: 31543249 PMCID: PMC6892259 DOI: 10.1016/s1473-3099(19)30323-8] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 05/24/2019] [Accepted: 06/07/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND A single dose of vaccine against yellow fever is routinely administered to infants aged 9-12 months under the Expanded Programme on Immunization, but the long-term outcome of vaccination in this age group is unknown. We aimed to evaluate the long-term persistence of neutralising antibodies to yellow fever virus following routine vaccination in infancy. METHODS We did a longitudinal cohort study, using a microneutralisation assay to measure protective antibodies against yellow fever in Malian and Ghanaian children vaccinated around age 9 months and followed up for 4·5 years (Mali), or 2·3 and 6·0 years (Ghana). Healthy children with available day-0 sera, a complete follow-up history, and no record of yellow fever revaccination were included; children seropositive for yellow fever at baseline were excluded. We standardised antibody concentrations with reference to the yellow fever WHO International Standard. FINDINGS We included 587 Malian and 436 Ghanaian children vaccinated between June 5, 2009, and Dec 26, 2012. In the Malian group, 296 (50·4%, 95% CI 46·4-54·5) were seropositive (antibody concentration ≥0·5 IU/mL) 4·5 years after vaccination. Among the Ghanaian children, 121 (27·8%, 23·5-32·0) were seropositive after 2·3 years. These results show a large decrease from the proportions of seropositive infants 28 days after vaccination, 96·7% in Mali and 72·7% in Ghana, reported by a previous study of both study populations. The number of seropositive children increased to 188 (43·1%, 95% CI 38·5-47·8) in the Ghanaian group 6·0 years after vaccination, but this result might be confounded by unrecorded revaccination or natural infection with wild yellow fever virus during a 2011-12 outbreak in northern Ghana. INTERPRETATION Rapid waning of immunity during the early years after vaccination of 9-month-old infants argues for a revision of the single-dose recommendation for this target population in endemic countries. The short duration of immunity in many vaccinees suggests that booster vaccination is necessary to meet the 80% population immunity threshold for prevention of yellow fever outbreaks. FUNDING Wellcome Trust.
Collapse
Affiliation(s)
- Cristina Domingo
- Robert Koch Institute, Highly Pathogenic Viruses (ZBS 1), Centre for Biological Threats and Special Pathogens, WHO Collaborating Centre for Emerging Infections and Biological Threats, Berlin, Germany.
| | - Juliane Fraissinet
- Robert Koch Institute, Highly Pathogenic Viruses (ZBS 1), Centre for Biological Threats and Special Pathogens, WHO Collaborating Centre for Emerging Infections and Biological Threats, Berlin, Germany
| | - Patrick O Ansah
- Navrongo Health Research Centre and Research Laboratory, Navrongo, Ghana
| | | | | | - Samba O Sow
- National Institute of Research on Public Health, Bamako, Mali
| | - José E Mejía
- Centre de Physiopathologie Toulouse-Purpan (CNRS, INSERM, Université Paul Sabatier), Centre Hospitalier Universitaire Purpan, Toulouse, France
| |
Collapse
|
9
|
Souza NCSE, Félix AC, de Paula AV, Levi JE, Pannuti CS, Romano CM. Evaluation of serological cross-reactivity between yellow fever and other flaviviruses. Int J Infect Dis 2019; 81:4-5. [PMID: 30660799 DOI: 10.1016/j.ijid.2019.01.023] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 01/07/2019] [Accepted: 01/10/2019] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVES This study was performed to determine whether neutralizing antibodies against yellow fever virus (YFV) generated by YFV vaccine could interfere in the specificity of dengue virus (DENV) and Zika virus (ZIKV) IgG ELISA tests. METHODS Seventy-nine pairs of serum samples (pre- and post-vaccination), collected during the years 1997-1998 from children with no history of yellow fever disease who had been vaccinated against YFV, were tested. The seroconversion post-vaccination was evaluated through plaque reduction neutralization test (PRNT), and four different commercial ELISA kits were used for the detection of DENV and ZIKV IgG antibodies. RESULTS A cross-reactivity rate of 3.9% with DENV IgG antibodies was found only with the Dengue Virus IgG Dx Select kit (Focus Diagnostics). CONCLUSIONS As several countries have local transmission of multiple arboviruses, the absence of cross-reactivity or minimum cross-reactivity of YFV neutralizing antibodies with DENV and ZIKV antigens is a relevant finding, since the interpretation of sero-epidemiological investigations would be seriously impacted in many regions where YFV vaccination is mandatory.
Collapse
Affiliation(s)
- Nathalia Caroline Santiago E Souza
- Instituto de Medicina Tropical de São Paulo, Universidade de São Paulo, São Paulo, Brazil; Departamento de Moléstias Infecciosas e Parasitárias, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Alvina Clara Félix
- Instituto de Medicina Tropical de São Paulo, Universidade de São Paulo, São Paulo, Brazil
| | | | - José Eduardo Levi
- Instituto de Medicina Tropical de São Paulo, Universidade de São Paulo, São Paulo, Brazil
| | - Claudio Sérgio Pannuti
- Instituto de Medicina Tropical de São Paulo, Universidade de São Paulo, São Paulo, Brazil
| | - Camila Malta Romano
- Instituto de Medicina Tropical de São Paulo, Universidade de São Paulo, São Paulo, Brazil; Hospital das Clinicas HCFMUSP (LIM52), Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil.
| |
Collapse
|
10
|
Willcox AC, Collins MH, Jadi R, Keeler C, Parr JB, Mumba D, Kashamuka M, Tshefu A, de Silva AM, Meshnick SR. Seroepidemiology of Dengue, Zika, and Yellow Fever Viruses among Children in the Democratic Republic of the Congo. Am J Trop Med Hyg 2018; 99:756-763. [PMID: 29988000 DOI: 10.4269/ajtmh.18-0156] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Flaviviruses such as Zika, dengue, and yellow fever cause epidemics throughout the tropics and account for substantial global morbidity and mortality. Although malaria and other vector-borne diseases have long been appreciated in Africa, flavivirus epidemiology is incompletely understood. Despite the existence of an effective vaccine, yellow fever continues to cause outbreaks and deaths, including at least 42 fatalities in the Democratic Republic of the Congo (DRC) in 2016. Here, we leveraged biospecimens collected as part of the nationally representative 2013-2014 Demographic and Health Survey in the DRC to examine serological evidence of flavivirus infection or vaccination in children aged 6 months to 5 years. Even in this young stratum of the Congolese population, we find evidence of infection by dengue and Zika viruses based on results from enzyme-linked immunosorbent assay and neutralization assay. Surprisingly, there was remarkable discordance between reported yellow fever vaccination status and results of serological assays. The estimated seroprevalences of neutralizing antibodies against each virus are yellow fever, 6.0% (95% confidence interval [CI] = 4.6-7.5%); dengue, 0.4% (0.1-0.9%); and Zika, 0.1% (0.0-0.5%). These results merit targeted, prospective studies to assess effectiveness of yellow fever vaccination programs, determine flavivirus seroprevalence across a broader age range, and investigate how these emerging diseases contribute to the burden of acute febrile illness in the DRC.
Collapse
Affiliation(s)
- Alexandra C Willcox
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, North Carolina
| | - Matthew H Collins
- Department of Medicine, Division of Infectious Diseases, Hope Clinic of the Emory Vaccine Center, Emory School of Medicine, Decatur, Georgia
| | - Ramesh Jadi
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Corinna Keeler
- Department of Geography, University of North Carolina, Chapel Hill, North Carolina
| | - Jonathan B Parr
- Department of Medicine, Division of Infectious Diseases, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Dieudonné Mumba
- Institut National de Recherche Biomédicale, Kinshasa, Democratic Republic of the Congo
| | - Melchior Kashamuka
- Ecole de Santé Publique, Faculté de Médecine, University of Kinshasa, Kinshasa, Democratic Republic of the Congo
| | - Antoinette Tshefu
- Ecole de Santé Publique, Faculté de Médecine, University of Kinshasa, Kinshasa, Democratic Republic of the Congo
| | - Aravinda M de Silva
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Steven R Meshnick
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, North Carolina
| |
Collapse
|
11
|
CHRONOVAC VOYAGEUR: A study of the immune response to yellow fever vaccine among infants previously immunized against measles. Vaccine 2017; 35:6166-6171. [DOI: 10.1016/j.vaccine.2017.09.047] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 09/12/2017] [Accepted: 09/14/2017] [Indexed: 11/20/2022]
|
12
|
Boikos C, Papenburg J, Martineau C, Joseph L, Scheifele D, Chilvers M, Lands LC, De Serres G, Quach C. Viral interference and the live-attenuated intranasal influenza vaccine: Results from a pediatric cohort with cystic fibrosis. Hum Vaccin Immunother 2017; 13:1-7. [PMID: 28273006 PMCID: PMC5489283 DOI: 10.1080/21645515.2017.1287641] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 01/03/2017] [Accepted: 01/24/2017] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND The objective of this study was to explore the effects of viral co-detection in individuals recently vaccinated with the live-attenuated intranasal influenza virus vaccine (LAIV) on the detection of influenza RNA. METHODS Before the 2013-2014 influenza season, nasal swabs were obtained from 59 pediatric participants with cystic fibrosis (CF) and 17 of their healthy siblings immediately before vaccination and 4 times during the week of follow-up. Real-time RT-PCR assays were used to detect influenza RNA. Co-detection of a non-influenza respiratory virus (NIRV) at the time of vaccination was determined by a multiplex RT-PCR assay. Differences in the proportions and rates of influenza detection and their 95% credible intervals (CrI) were estimated. RESULTS Influenza RNA was detected in 16% fewer participants (95% CrI: -7, 39%) throughout follow-up in the NIRV-positive group compared with the NIRV-negative group (59% vs. 75%). This was also observed in participants with CF alone (66% vs. 74%; RD = 8% 95% CrI: -16, 33%) as well as in healthy participants only (75% vs. 30%; RD = 45%, 95% CrI: -2, 81%). Influenza was detected in NIRV-negative subjects for 0.49 d more compared with NIRV-positive subjects (95% CrI: -0.37, 1.26). CONCLUSION The observed proportion of subjects in whom influenza RNA was detected and the duration of detection differed slightly between NIRV- positive and -negative subjects. However, wide credible intervals for the difference preclude definitive conclusions. If true, this observed association may be related to a recent viral respiratory infection, a phenomenon known as viral interference.
Collapse
Affiliation(s)
- Constantina Boikos
- Department of Epidemiology, Biostatistics & Occupational Health, McGill University, Montreal, QC, Canada
| | - Jesse Papenburg
- Department of Pediatrics, Division of Infectious Diseases, The Montreal Children's Hospital, McGill University, Montreal, QC, Canada
| | - Christine Martineau
- Laboratoire de santé publique du Québec, Institut national de santé publique du Québec, QC, Canada
| | - Lawrence Joseph
- Department of Epidemiology, Biostatistics & Occupational Health, McGill University, Montreal, QC, Canada
| | - David Scheifele
- Vaccine Evaluation Center, Child & Family Research Institute, University of British Columbia, BC, Canada
| | - Mark Chilvers
- Director, Cystic Fibrosis Clinic, University of British Columbia, BC, Canada
| | - Larry C. Lands
- Department of Pediatrics, Division of Respiratory Medicine, The Montreal Children's Hospital, McGill University, Montreal, QC, Canada
| | - Gaston De Serres
- Direction des risques biologiques et de la santé au travail, Institut national de santé publique du Québec, QC, Canada
| | - Caroline Quach
- Department of Epidemiology, Biostatistics & Occupational Health, McGill University, Montreal, QC, Canada
- Department of Pediatrics, Division of Infectious Diseases, The Montreal Children's Hospital, McGill University, Montreal, QC, Canada
- Direction des risques biologiques et de la santé au travail, Institut national de santé publique du Québec, QC, Canada
- McGill University Health Centre, Vaccine Study Centre, Research Institute of the MUHC, Montreal, QC, Canada
| |
Collapse
|
13
|
Nicoli F, Appay V. Immunological considerations regarding parental concerns on pediatric immunizations. Vaccine 2017; 35:3012-3019. [PMID: 28465096 DOI: 10.1016/j.vaccine.2017.04.030] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 03/31/2017] [Accepted: 04/12/2017] [Indexed: 01/10/2023]
Abstract
Despite the fundamental role of vaccines in the decline of infant mortality, parents may decide to decline vaccination for their own children. Many factors may influence this decision, such as the belief that the infant immune system is weakened by vaccines, and concerns have been raised about the number of vaccines and the early age at which they are administered. Studies focused on the infant immune system and its reaction to immunizations, summarized in this review, show that vaccines can overcome those suboptimal features of infant immune system that render them more at risk of infections and of their severe manifestations. In addition, many vaccines have been shown to improve heterologous innate and adaptive immunity resulting in lower mortality rates for fully vaccinated children. Thus, multiple vaccinations are necessary and not dangerous, as infants can respond to several antigens as well as when responding to single stimuli. Current immunization schedules have been developed and tested to avoid vaccine interference, improve benefits and reduce side effects compared to single administrations. The infant immune system is therefore capable, early after birth, of managing several antigenic challenges and exploits them to prompt its development.
Collapse
Affiliation(s)
- Francesco Nicoli
- Sorbonne Universités, UPMC Univ Paris 06, DHU FAST, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), F-75013 Paris, France; INSERM, U1135, CIMI-Paris, F-75013 Paris, France.
| | - Victor Appay
- Sorbonne Universités, UPMC Univ Paris 06, DHU FAST, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), F-75013 Paris, France; INSERM, U1135, CIMI-Paris, F-75013 Paris, France; International Research Center of Medical Sciences (IRCMS), Kumamoto University, Kumamoto 860-0811, Japan
| |
Collapse
|
14
|
Immunogenicity and Safety of Yellow Fever Vaccine (Stamaril) When Administered Concomitantly With a Tetravalent Dengue Vaccine Candidate in Healthy Toddlers at 12-13 Months of Age in Colombia and Peru: A Randomized Trial. Pediatr Infect Dis J 2016; 35:1140-7. [PMID: 27254034 DOI: 10.1097/inf.0000000000001250] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND Dengue and yellow fever (YF) viruses are closely related members of the Flaviviridae family. Given the inherent similarities between the YF vaccine and dengue vaccine (CYD-TDV) candidate, it is possible that the latter could interfere with the response to the licensed YF vaccine when coadministered. METHODS In this randomized, observer-blind, controlled, phase III trial, conducted in Colombia and Peru, 787 toddlers were administered YF vaccine concomitantly with CYD-TDV (group 1) or placebo (group 2), followed by CYD-TDV after 6 and 12 months. YF and dengue neutralizing antibody titers were determined using a 50% plaque reduction neutralization test. Noninferiority was demonstrated if the lower limit of the 2-sided 95% confidence interval of the difference in seroconversion rates [(YF + CYD-TDV) - YF alone] was greater than -10%. The safety of both vaccines was also assessed. RESULTS Concomitant administration of YF with either CYD-TDV or placebo yielded YF seroconversion rates of 100.0% and 99.7%, respectively. The difference in YF seroconversion rates between the 2 groups was 0.33% (95% confidence interval:0.98; 1.87), demonstrating that the immune response against YF administered concomitantly with CYD-TDV was noninferior to YF administered with placebo. After 2 injections of CYD-TDV, the percentage of participants with dengue titres ≥10 (1/dil) for the 4 dengue serotypes were 91.2%-100% for group 1 and 97.2%-100% in group 2. There were no safety concerns during the study period. CONCLUSIONS Concomitant administration of YF vaccine with CYD-TDV has no relevant impact on the immunogenicity or safety profile of the YF vaccine.
Collapse
|
15
|
Roy Chowdhury P, Meier C, Laraway H, Tang Y, Hodgson A, Sow SO, Enwere GC, Plikaytis BD, Kulkarni PS, Preziosi MP, Niedrig M. Immunogenicity of Yellow Fever Vaccine Coadministered With MenAfriVac in Healthy Infants in Ghana and Mali. Clin Infect Dis 2016; 61 Suppl 5:S586-93. [PMID: 26553692 PMCID: PMC4639505 DOI: 10.1093/cid/civ603] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Yellow fever (YF) is still a major public health problem in endemic regions of Africa and South America. In Africa, one of the main control strategies is routine vaccination within the Expanded Programme on Immunization (EPI). A new meningococcal A conjugate vaccine (PsA-TT) is about to be introduced in the EPI of countries in the African meningitis belt, and this study reports on the immunogenicity of the YF-17D vaccines in infants when administered concomitantly with measles vaccine and PsA-TT. METHODS Two clinical studies were conducted in Ghana and in Mali among infants who received PsA-TT concomitantly with measles and YF vaccines at 9 months of age. YF neutralizing antibody titers were measured using a microneutralization assay. RESULTS In both studies, the PsA-TT did not adversely affect the immune response to the concomitantly administered YF vaccine at the age of 9 months. The magnitude of the immune response was different between the 2 studies, with higher seroconversion and seroprotection rates found in Mali vs Ghana. CONCLUSIONS Immunogenicity to YF vaccine is unaffected when coadministered with PsA-TT at 9 months of age. Further studies are warranted to better understand the determinants of the immune response to YF vaccine in infancy. CLINICAL TRIALS REGISTRATION ISRCTN82484612 (PsA-TT-004); PACTR201110000328305 (PsA-TT-007).
Collapse
Affiliation(s)
- Panchali Roy Chowdhury
- Centre for Biological Threats and Special Pathogens, Robert Koch Institut, Berlin, Germany
| | - Christian Meier
- Centre for Biological Threats and Special Pathogens, Robert Koch Institut, Berlin, Germany
| | - Hewad Laraway
- Centre for Biological Threats and Special Pathogens, Robert Koch Institut, Berlin, Germany
| | - Yuxiao Tang
- Meningitis Vaccine Project, PATH, Seattle, Washington
| | - Abraham Hodgson
- Navrongo Health Research Centre, Ghana Health Service, Navrongo, Ghana
| | - Samba O Sow
- Centre pour le Développement des Vaccins, Ministère de la Santé, Bamako, Mali
| | | | | | | | - Marie-Pierre Preziosi
- Meningitis Vaccine Project, PATH, Ferney-Voltaire, France Meningitis Vaccine Project, Department of Immunization, Vaccines and Biologicals, World Health Organization, Geneva, Switzerland
| | - Matthias Niedrig
- Centre for Biological Threats and Special Pathogens, Robert Koch Institut, Berlin, Germany
| |
Collapse
|
16
|
A randomised double-blind clinical trial of two yellow fever vaccines prepared with substrains 17DD and 17D-213/77 in children nine-23 months old. Mem Inst Oswaldo Cruz 2015; 110:771-80. [PMID: 26517656 PMCID: PMC4667580 DOI: 10.1590/0074-02760150176] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 07/20/2015] [Indexed: 11/22/2022] Open
Abstract
This randomised, double-blind, multicentre study with children nine-23 months old evaluated the immunogenicity of yellow fever (YF) vaccines prepared with substrains 17DD and 17D-213/77. YF antibodies were titered before and 30 or more days after vaccination. Seropositivity and seroconversion were analysed according to the maternal serological status and the collaborating centre. A total of 1,966 children were randomised in the municipalities of the states of Mato Grosso do Sul, Minas Gerais and São Paulo and blood samples were collected from 1,714 mothers. Seropositivity was observed in 78.6% of mothers and 8.9% of children before vaccination. After vaccination, seropositivity rates of 81.9% and 83.2%, seroconversion rates of 84.8% and 85.8% and rates of a four-fold increase over the pre-vaccination titre of 77.6% and 81.8% were observed in the 17D-213/77 and 17DD subgroups, respectively. There was no association with maternal immunity. Among children aged 12 months or older, the seroconversion rates of 69% were associated with concomitant vaccination against measles, mumps and rubella. The data were not conclusive regarding the interference of maternal immunity in the immune response to the YF vaccine, but they suggest interference from other vaccines. The failures in seroconversion after vaccination support the recommendation of a booster dose in children within 10 years of the first dose.
Collapse
|
17
|
Observational study on immune response to yellow fever and measles vaccines in 9 to 15-month old children. Is it necessary to wait 4 weeks between two live attenuated vaccines? Vaccine 2015; 33:2301-6. [DOI: 10.1016/j.vaccine.2015.03.069] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2014] [Revised: 03/20/2015] [Accepted: 03/23/2015] [Indexed: 11/24/2022]
|
18
|
Maciel M, Cruz FDSP, Cordeiro MT, da Motta MA, Cassemiro KMSDM, Maia RDCC, de Figueiredo RCBQ, Galler R, Freire MDS, August JT, Marques ETA, Dhalia R. A DNA vaccine against yellow fever virus: development and evaluation. PLoS Negl Trop Dis 2015; 9:e0003693. [PMID: 25875109 PMCID: PMC4395287 DOI: 10.1371/journal.pntd.0003693] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 03/10/2015] [Indexed: 11/30/2022] Open
Abstract
Attenuated yellow fever (YF) virus 17D/17DD vaccines are the only available protection from YF infection, which remains a significant source of morbidity and mortality in the tropical areas of the world. The attenuated YF virus vaccine, which is used worldwide, generates both long-lasting neutralizing antibodies and strong T-cell responses. However, on rare occasions, this vaccine has toxic side effects that can be fatal. This study presents the design of two non-viral DNA-based antigen formulations and the characterization of their expression and immunological properties. The two antigen formulations consist of DNA encoding the full-length envelope protein (p/YFE) or the full-length envelope protein fused to the lysosomal-associated membrane protein signal, LAMP-1 (pL/YFE), aimed at diverting antigen processing/presentation through the major histocompatibility complex II precursor compartments. The immune responses triggered by these formulations were evaluated in H2b and H2d backgrounds, corresponding to the C57Bl/6 and BALB/c mice strains, respectively. Both DNA constructs were able to induce very strong T-cell responses of similar magnitude against almost all epitopes that are also generated by the YF 17DD vaccine. The pL/YFE formulation performed best overall. In addition to the T-cell response, it was also able to stimulate high titers of anti-YF neutralizing antibodies comparable to the levels elicited by the 17DD vaccine. More importantly, the pL/YFE vaccine conferred 100% protection against the YF virus in intracerebrally challenged mice. These results indicate that pL/YFE DNA is an excellent vaccine candidate and should be considered for further developmental studies.
Collapse
Affiliation(s)
- Milton Maciel
- Johns Hopkins University, School of Medicine, Department of Pharmacology & Molecular Sciences, Baltimore, Maryland, United States of America
| | - Fábia da Silva Pereira Cruz
- Oswaldo Cruz Foundation (FIOCRUZ), Aggeu Magalhães Research Centre, Department of Virology, Laboratório de Virologia e Terapia Experimental (LAVITE), Universidade Federal de Pernambuco (UFPE), University City, Recife, Pernambuco, Brazil
| | - Marli Tenório Cordeiro
- Oswaldo Cruz Foundation (FIOCRUZ), Aggeu Magalhães Research Centre, Department of Virology, Laboratório de Virologia e Terapia Experimental (LAVITE), Universidade Federal de Pernambuco (UFPE), University City, Recife, Pernambuco, Brazil
- Health Secretariat of the State of Pernambuco, Central Public Health Laboratory-LACEN, Boa Vista, Recife, Pernambuco, Brazil
| | - Márcia Archer da Motta
- Oswaldo Cruz Foundation (FIOCRUZ), Oswaldo Cruz Institute, Bio-Manguinhos, Laboratório de Tecnologia Virológica (LATEV), Manguinhos, Rio de Janeiro, Brazil
| | - Klécia Marília Soares de Melo Cassemiro
- Oswaldo Cruz Foundation (FIOCRUZ), Aggeu Magalhães Research Centre, Department of Virology, Laboratório de Virologia e Terapia Experimental (LAVITE), Universidade Federal de Pernambuco (UFPE), University City, Recife, Pernambuco, Brazil
| | - Rita de Cássia Carvalho Maia
- Federal Rural University of Pernambuco, Department of Veterinary Medicine, Dois Irmãos, Recife, Pernambuco, Brazil
| | - Regina Célia Bressan Queiroz de Figueiredo
- Oswaldo Cruz Foundation (FIOCRUZ), Aggeu Magalhães Research Centre, Department of Virology, Laboratório de Virologia e Terapia Experimental (LAVITE), Universidade Federal de Pernambuco (UFPE), University City, Recife, Pernambuco, Brazil
| | - Ricardo Galler
- Oswaldo Cruz Foundation (FIOCRUZ), Oswaldo Cruz Institute, Bio-Manguinhos, Laboratório de Tecnologia Virológica (LATEV), Manguinhos, Rio de Janeiro, Brazil
| | - Marcos da Silva Freire
- Oswaldo Cruz Foundation (FIOCRUZ), Oswaldo Cruz Institute, Bio-Manguinhos, Laboratório de Tecnologia Virológica (LATEV), Manguinhos, Rio de Janeiro, Brazil
| | - Joseph Thomas August
- Johns Hopkins University, School of Medicine, Department of Pharmacology & Molecular Sciences, Baltimore, Maryland, United States of America
| | - Ernesto T. A. Marques
- Johns Hopkins University, School of Medicine, Department of Pharmacology & Molecular Sciences, Baltimore, Maryland, United States of America
- Oswaldo Cruz Foundation (FIOCRUZ), Aggeu Magalhães Research Centre, Department of Virology, Laboratório de Virologia e Terapia Experimental (LAVITE), Universidade Federal de Pernambuco (UFPE), University City, Recife, Pernambuco, Brazil
- University of Pittsburgh, Center for Vaccine Research, Pittsburgh, Pennsylvania, United States of America
| | - Rafael Dhalia
- Oswaldo Cruz Foundation (FIOCRUZ), Aggeu Magalhães Research Centre, Department of Virology, Laboratório de Virologia e Terapia Experimental (LAVITE), Universidade Federal de Pernambuco (UFPE), University City, Recife, Pernambuco, Brazil
| |
Collapse
|
19
|
Martins RDM, Possas CDA, Homma A. Historical review of clinical vaccine studies at Oswaldo Cruz Institute and Oswaldo Cruz Foundation--technological development issues. Mem Inst Oswaldo Cruz 2015; 110:114-24. [PMID: 25742271 PMCID: PMC4371225 DOI: 10.1590/0074-02760140346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 12/05/2014] [Indexed: 12/03/2022] Open
Abstract
This paper presents, from the perspective of technological development and
production, the results of an investigation examining 61 clinical studies with
vaccines conducted in Brazil between 1938-2013, with the participation of the Oswaldo
Cruz Institute (IOC) and the Oswaldo Cruz Foundation (Fiocruz). These studies have
been identified and reviewed according to criteria, such as the kind of vaccine
(viral, bacterial, parasitic), their rationale, design and methodological strategies.
The results indicate that IOC and Fiocruz have accumulated along this time
significant knowledge and experience for the performance of studies in all clinical
phases and are prepared for the development of new vaccines products and processes.
We recommend national policy strategies to overcome existing regulatory and financing
constraints.
Collapse
Affiliation(s)
| | | | - Akira Homma
- Bio-Manguinhos-Fiocruz, Rio de Janeiro, RJ, Brasil
| |
Collapse
|
20
|
Campi-Azevedo AC, de Almeida Estevam P, Coelho-Dos-Reis JG, Peruhype-Magalhães V, Villela-Rezende G, Quaresma PF, Maia MDLS, Farias RHG, Camacho LAB, Freire MDS, Galler R, Yamamura AMY, Almeida LFC, Lima SMB, Nogueira RMR, Silva Sá GR, Hokama DA, de Carvalho R, Freire RAV, Filho EP, Leal MDLF, Homma A, Teixeira-Carvalho A, Martins RM, Martins-Filho OA. Subdoses of 17DD yellow fever vaccine elicit equivalent virological/immunological kinetics timeline. BMC Infect Dis 2014; 14:391. [PMID: 25022840 PMCID: PMC4223624 DOI: 10.1186/1471-2334-14-391] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Accepted: 07/03/2014] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND The live attenuated 17DD Yellow Fever vaccine is one of the most successful prophylactic interventions for controlling disease expansion ever designed and utilized in larger scale. However, increase on worldwide vaccine demands and manufacturing restrictions urge for more detailed dose sparing studies. The establishment of complementary biomarkers in addition to PRNT and Viremia could support a secure decision-making regarding the use of 17DD YF vaccine subdoses. The present work aimed at comparing the serum chemokine and cytokine kinetics triggered by five subdoses of 17DD YF Vaccine. METHODS Neutralizing antibody titers, viremia, cytokines and chemokines were tested on blood samples obtained from eligible primary vaccinees. RESULTS AND DISCUSSION The results demonstrated that a fifty-fold lower dose of 17DD-YF vaccine (587 IU) is able to trigger similar immunogenicity, as evidenced by significant titers of anti-YF PRNT. However, only subdoses as low as 3,013 IU elicit viremia kinetics with an early peak at five days after primary vaccination equivalent to the current dose (27,476 IU), while other subdoses show a distinct, lower in magnitude and later peak at day 6 post-vaccination. Although the subdose of 587 IU is able to trigger equivalent kinetics of IL-8/CXCL-8 and MCP-1/CCL-2, only the subdose of 3,013 IU is able to trigger similar kinetics of MIG/CXCL-9, pro-inflammatory (TNF, IFN-γ and IL-2) and modulatory cytokines (IL-5 and IL-10). CONCLUSIONS The analysis of serum biomarkers IFN-γ and IL-10, in association to PRNT and viremia, support the recommendation of use of a ten-fold lower subdose (3,013 IU) of 17DD-YF vaccine.
Collapse
|
21
|
de Santana MGV, Neves PCC, dos Santos JR, Lima NS, dos Santos AAC, Watkins DI, Galler R, Bonaldo MC. Improved genetic stability of recombinant yellow fever 17D virus expressing a lentiviral Gag gene fragment. Virology 2014; 452-453:202-11. [PMID: 24606697 DOI: 10.1016/j.virol.2014.01.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Revised: 11/11/2013] [Accepted: 01/20/2014] [Indexed: 10/25/2022]
Abstract
We have previously designed a method to construct viable recombinant Yellow Fever (YF) 17D viruses expressing heterologous polypeptides including part of the Simian Immunodeficiency Virus (SIV) Gag protein. However, the expressed region, encompassing amino acid residues from 45 to 269, was genetically unstable. In this study, we improved the genetic stability of this recombinant YF 17D virus by introducing mutations in the IRES element localized at the 5' end of the SIV gag gene. The new stable recombinant virus elicited adaptive immune responses similar to those induced by the original recombinant virus. It is, therefore, possible to increase recombinant stability by removing functional motifs from the insert that may have deleterious effects on recombinant YF viral fitness.
Collapse
Affiliation(s)
- Marlon G Veloso de Santana
- Laboratório de Biologia Molecular de Flavivírus, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil; Department of Pathology, University of Miami, Miller School of Medicine, United States of America
| | - Patrícia C C Neves
- Laboratório de Biologia Molecular de Flavivírus, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - Juliana Ribeiro dos Santos
- Laboratório de Biologia Molecular de Flavivírus, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - Noemia S Lima
- Laboratório de Biologia Molecular de Flavivírus, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - Alexandre A C dos Santos
- Laboratório de Biologia Molecular de Flavivírus, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - David I Watkins
- Department of Pathology, University of Miami, Miller School of Medicine, United States of America
| | - Ricardo Galler
- Instituto de Tecnologia em Imunobiológicos, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Myrna C Bonaldo
- Laboratório de Biologia Molecular de Flavivírus, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil.
| |
Collapse
|
22
|
Neves PCC, Santos JR, Tubarão LN, Bonaldo MC, Galler R. Early IFN-gamma production after YF 17D vaccine virus immunization in mice and its association with adaptive immune responses. PLoS One 2013; 8:e81953. [PMID: 24324734 PMCID: PMC3855709 DOI: 10.1371/journal.pone.0081953] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Accepted: 10/18/2013] [Indexed: 01/01/2023] Open
Abstract
Yellow Fever vaccine is one of the most efficacious human vaccines ever made. The vaccine (YF 17D) virus induces polyvalent immune responses, with a mixed TH1/TH2 CD4(+) cell profile, which results in robust T CD8(+) responses and high titers of neutralizing antibody. In recent years, it has been suggested that early events after yellow fever vaccination are crucial to the development of adequate acquired immunity. We have previously shown that primary immunization of humans and monkeys with YF 17D virus vaccine resulted in the early synthesis of IFN-γ. Herein we have demonstrated, for the first time that early IFN-γ production after yellow fever vaccination is a feature also of murine infection and is much more pronounced in the C57BL/6 strain compared to the BALB/c strain. Likewise, in C57BL/6 strain, we have observed the highest CD8(+) T cells responses as well as higher titers of neutralizing antibodies and total anti-YF IgG. Regardless of this intense IFN-γ response in mice, it was not possible to see higher titers of IgG2a in relation to IgG1 in both mice lineages. However, IgG2a titers were positively correlated to neutralizing antibodies levels, pointing to an important role of IFN-γ in eliciting high quality responses against YF 17D, therefore influencing the immunogenicity of this vaccine.
Collapse
Affiliation(s)
- Patrícia C. C. Neves
- Vice-diretoria de Desenvolvimento Tecnológico, Instituto de Tecnologia em Imunobiológicos, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Juliana R. Santos
- Laboratório de Biologia Molecular de Flavivírus, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Luciana N. Tubarão
- Laboratório de Biologia Molecular de Flavivírus, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Myrna C. Bonaldo
- Laboratório de Biologia Molecular de Flavivírus, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Ricardo Galler
- Vice-diretoria de Desenvolvimento Tecnológico, Instituto de Tecnologia em Imunobiológicos, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| |
Collapse
|
23
|
Gotuzzo E, Yactayo S, Córdova E. Efficacy and duration of immunity after yellow fever vaccination: systematic review on the need for a booster every 10 years. Am J Trop Med Hyg 2013; 89:434-44. [PMID: 24006295 DOI: 10.4269/ajtmh.13-0264] [Citation(s) in RCA: 167] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Abstract. Current regulations stipulate a yellow fever (YF) booster every 10 years. We conducted a systematic review of the protective efficacy and duration of immunity of YF vaccine in residents of disease-endemic areas and in travelers to assess the need for a booster in these two settings and in selected populations (human immunodeficiency virus-infected persons, infants, children, pregnant women, and severely malnourished persons). Thirty-six studies and 22 reports were included. We identified 12 studies of immunogenicity, 8 of duration of immunity, 8 of vaccine response in infants and children, 7 of human-immunodeficiency virus-infected persons, 2 of pregnant women, and 1 of severely malnourished children. Based on currently available data, a single dose of YF vaccine is highly immunogenic and confers sustained life-long protective immunity against YF. Therefore, a booster dose of YF vaccine is not needed. Special considerations for selected populations are detailed.
Collapse
Affiliation(s)
- Eduardo Gotuzzo
- Instituto de Medicina Tropical Alexander von Humbolt, Lima, Peru.
| | | | | |
Collapse
|
24
|
Machado VW, Vasconcelos PFDC, Silva EVP, Santos JB. Serologic assessment of yellow fever immunity in the rural population of a yellow fever-endemic area in Central Brazil. Rev Soc Bras Med Trop 2013; 46:166-71. [DOI: 10.1590/0037-8682-0007-2012] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Accepted: 03/26/2013] [Indexed: 11/22/2022] Open
|
25
|
|
26
|
|
27
|
|
28
|
Campi-Azevedo AC, de Araújo-Porto LP, Luiza-Silva M, Batista MA, Martins MA, Sathler-Avelar R, da Silveira-Lemos D, Camacho LAB, de Menezes Martins R, de Lourdes de Sousa Maia M, Farias RHG, da Silva Freire M, Galler R, Homma A, Ribeiro JGL, Lemos JAC, Auxiliadora-Martins M, Caldas IR, Elói-Santos SM, Teixeira-Carvalho A, Martins-Filho OA. 17DD and 17D-213/77 yellow fever substrains trigger a balanced cytokine profile in primary vaccinated children. PLoS One 2012; 7:e49828. [PMID: 23251351 PMCID: PMC3519464 DOI: 10.1371/journal.pone.0049828] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2012] [Accepted: 10/17/2012] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND This study aimed to compare the cytokine-mediated immune response in children submitted to primary vaccination with the YF-17D-213/77 or YF-17DD yellow fever (YF) substrains. METHODS A non-probabilistic sample of eighty healthy primary vaccinated (PV) children was selected on the basis of their previously known humoral immune response to the YF vaccines. The selected children were categorized according to their YF-neutralizing antibody titers (PRNT) and referred to as seroconverters (PV-PRNT(+)) or nonseroconverters (PV-PRNT(-)). Following revaccination with the YF-17DD, the PV-PRNT(-) children (YF-17D-213/77 and YF-17DD groups) seroconverted and were referred as RV-PRNT(+). The cytokine-mediated immune response was investigated after short-term in vitro cultures of whole blood samples. The results are expressed as frequency of high cytokine producers, taking the global median of the cytokine index (YF-Ag/control) as the cut-off. RESULTS The YF-17D-213/77 and the YF-17DD substrains triggered a balanced overall inflammatory/regulatory cytokine pattern in PV-PRNT(+), with a slight predominance of IL-12 in YF-17DD vaccinees and a modest prevalence of IL-10 in YF-17D-213/77. Prominent frequency of neutrophil-derived TNF-α and neutrophils and monocyte-producing IL-12 were the major features of PV-PRNT(+) in the YF-17DD, whereas relevant inflammatory response, mediated by IL-12(+)CD8(+) T cells, was the hallmark of the YF-17D-213/77 vaccinees. Both substrains were able to elicit particular but relevant inflammatory events, regardless of the anti-YF PRNT antibody levels. PV-PRNT(-) children belonging to the YF-17DD arm presented gaps in the inflammatory cytokine signature, especially in terms of the innate immunity, whereas in the YF-17D-213/77 arm the most relevant gap was the deficiency of IL-12-producing CD8(+)T cells. Revaccination with YF-17DD prompted a balanced cytokine profile in YF-17DD nonresponders and a robust inflammatory profile in YF-17D-213/77 nonresponders. CONCLUSION Our findings demonstrated that, just like the YF-17DD reference vaccine, the YF-17D-213/77 seed lot induced a mixed pattern of inflammatory and regulatory cytokines, supporting its universal use for immunization.
Collapse
Affiliation(s)
- Ana Carolina Campi-Azevedo
- Laboratório de Biomarcadores de Diagnóstico e Monitoração, Centro de Pesquisas René Rachou, FIOCRUZ, Belo Horizonte, Minas Gerais, Brasil
- * E-mail:
| | - Luiza Pacheco de Araújo-Porto
- Laboratório de Biomarcadores de Diagnóstico e Monitoração, Centro de Pesquisas René Rachou, FIOCRUZ, Belo Horizonte, Minas Gerais, Brasil
| | - Maria Luiza-Silva
- Laboratório de Biomarcadores de Diagnóstico e Monitoração, Centro de Pesquisas René Rachou, FIOCRUZ, Belo Horizonte, Minas Gerais, Brasil
| | - Maurício Azevedo Batista
- Núcleo de Pesquisas em Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brasil
| | - Marina Angela Martins
- Laboratório de Biomarcadores de Diagnóstico e Monitoração, Centro de Pesquisas René Rachou, FIOCRUZ, Belo Horizonte, Minas Gerais, Brasil
| | - Renato Sathler-Avelar
- Laboratório de Biomarcadores de Diagnóstico e Monitoração, Centro de Pesquisas René Rachou, FIOCRUZ, Belo Horizonte, Minas Gerais, Brasil
| | - Denise da Silveira-Lemos
- Núcleo de Pesquisas em Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brasil
| | | | - Reinaldo de Menezes Martins
- Instituto de Tecnologia em Imunobiológicos, Bio-Manguinhos - FIOCRUZ - Rio de Janeiro, Rio de Janeiro, Brasil
| | | | | | - Marcos da Silva Freire
- Instituto de Tecnologia em Imunobiológicos, Bio-Manguinhos - FIOCRUZ - Rio de Janeiro, Rio de Janeiro, Brasil
| | - Ricardo Galler
- Instituto de Tecnologia em Imunobiológicos, Bio-Manguinhos - FIOCRUZ - Rio de Janeiro, Rio de Janeiro, Brasil
| | - Akira Homma
- Instituto de Tecnologia em Imunobiológicos, Bio-Manguinhos - FIOCRUZ - Rio de Janeiro, Rio de Janeiro, Brasil
| | | | | | - Maria Auxiliadora-Martins
- Hospital das Clínicas, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo-USP, Ribeirão Preto, São Paulo, Brasil
| | | | - Silvana Maria Elói-Santos
- Departamento de Propedêutica Complementar, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
| | - Andréa Teixeira-Carvalho
- Laboratório de Biomarcadores de Diagnóstico e Monitoração, Centro de Pesquisas René Rachou, FIOCRUZ, Belo Horizonte, Minas Gerais, Brasil
| | - Olindo Assis Martins-Filho
- Laboratório de Biomarcadores de Diagnóstico e Monitoração, Centro de Pesquisas René Rachou, FIOCRUZ, Belo Horizonte, Minas Gerais, Brasil
| |
Collapse
|
29
|
Simões M, Camacho LAB, Yamamura AM, Miranda EH, Cajaraville ACR, da Silva Freire M. Evaluation of accuracy and reliability of the plaque reduction neutralization test (micro-PRNT) in detection of yellow fever virus antibodies. Biologicals 2012; 40:399-404. [DOI: 10.1016/j.biologicals.2012.09.005] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Revised: 08/24/2012] [Accepted: 09/07/2012] [Indexed: 12/01/2022] Open
|
30
|
Stier DM, Weber IB, Staples JE. Lack of interference by zoster vaccine with the immune response to yellow fever vaccine. J Travel Med 2012; 19:122-3. [PMID: 22414038 PMCID: PMC4669900 DOI: 10.1111/j.1708-8305.2011.00585.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Concerns exist about the serologic response to yellow fever (YF) vaccine when given within 28 days of another live virus vaccine. We report the case of a healthy adult who received 17D YF vaccine 21 days following administration of another live viral vaccine, and developed a protective level of immunity against YF virus.
Collapse
Affiliation(s)
- David M Stier
- Communicable Disease Control and Prevention Section, San Francisco Department of Public Health, San Francisco, CA 94102, USA.
| | | | | |
Collapse
|
31
|
Nascimento Silva JR, Camacho LAB, Siqueira MM, Freire MDS, Castro YP, Maia MDLS, Yamamura AMY, Martins RM, Leal MDLF. Mutual interference on the immune response to yellow fever vaccine and a combined vaccine against measles, mumps and rubella. Vaccine 2011; 29:6327-34. [DOI: 10.1016/j.vaccine.2011.05.019] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2010] [Revised: 05/05/2011] [Accepted: 05/09/2011] [Indexed: 12/01/2022]
|
32
|
Azevedo AS, Yamamura AMY, Freire MS, Trindade GF, Bonaldo M, Galler R, Alves AMB. DNA vaccines against dengue virus type 2 based on truncate envelope protein or its domain III. PLoS One 2011; 6:e20528. [PMID: 21779317 PMCID: PMC3136928 DOI: 10.1371/journal.pone.0020528] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2011] [Accepted: 05/03/2011] [Indexed: 01/18/2023] Open
Abstract
Two DNA vaccines were constructed encoding the ectodomain (domains I, II and III) of the DENV2 envelope protein (pE1D2) or only its domain III (pE2D2), fused to the human tissue plasminogen activator signal peptide (t-PA). The expression and secretion of recombinant proteins was confirmed in vitro in BHK cells transfected with the two plasmids, detected by immunofluorescence or immunoprecipitation of metabolically labeled gene products, using polyclonal and monoclonal antibodies against DENV2. Besides, results reveal that the ectodomain of the E protein can be efficiently expressed in vivo, in a mammalian system, without the prM protein that is hypothesized to act as a chaperonin during dengue infection. Balb/c mice were immunized with the DNA vaccines and challenged with a lethal dose of DENV2. All pE1D2-vaccinated mice survived challenge, while 45% of animals immunized with the pE2D2 died after infection. Furthermore, only 10% of pE1D2-immunized mice presented some clinical signs of infection after challenge, whereas most of animals inoculated with the pE2D2 showed effects of the disease with high morbidity degrees. Levels of neutralizing antibodies were significantly higher in pE1D2-vaccinated mice than in pE2D2-immunized animals, also suggesting that the pE1D2 vaccine was more protective than the pE2D2.
Collapse
Affiliation(s)
- Adriana S. Azevedo
- Laboratório de Biotecnologia e Fisiologia de Infecções Virais, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Anna M. Y. Yamamura
- Laboratório de Tecnologia Virológica, Instituto de Tecnologia em Imunobiológicos, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Marcos S. Freire
- Laboratório de Tecnologia Virológica, Instituto de Tecnologia em Imunobiológicos, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Gisela F. Trindade
- Laboratório de Biologia Molecular de Flavivirus, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Myrna Bonaldo
- Laboratório de Biologia Molecular de Flavivirus, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Ricardo Galler
- Laboratório de Tecnologia Virológica, Instituto de Tecnologia em Imunobiológicos, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Ada M. B. Alves
- Laboratório de Biotecnologia e Fisiologia de Infecções Virais, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
- * E-mail:
| |
Collapse
|
33
|
Veras MASM, Flannery B, de Moraes JC, da Silva Teixeira AM, Luna EJA. Yellow fever vaccination coverage among children in Brazilian capitals. Vaccine 2010; 28:6478-82. [PMID: 20674878 DOI: 10.1016/j.vaccine.2010.07.035] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2010] [Revised: 07/13/2010] [Accepted: 07/13/2010] [Indexed: 11/24/2022]
Abstract
Brazil recommends universal yellow fever (YF) vaccination for children who reside in or travel to endemic areas. We conducted a household survey to calculate YF vaccine coverage among children 18-30 months of age in 27 capital cities. A total of 9285 children were surveyed in the 15 cities with YF fever universal vaccination; 7290 (79%) had documented evidence of YF vaccination by 12 months of age, 7996 (86%) by 18 months of age, and 8479 (91%) prior to the survey. In 12 cities with selective YF vaccination coverage was only 1% by 18 months of age. YF fever vaccination can be improved to reach all children where vaccine is recommended.
Collapse
|
34
|
Gaspar LP, Mendes YS, Yamamura AMY, Almeida LFC, Caride E, Gonçalves RB, Silva JL, Oliveira AC, Galler R, Freire MS. Pressure-inactivated yellow fever 17DD virus: implications for vaccine development. J Virol Methods 2008; 150:57-62. [PMID: 18420285 DOI: 10.1016/j.jviromet.2008.03.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2007] [Revised: 03/03/2008] [Accepted: 03/06/2008] [Indexed: 10/22/2022]
Abstract
The successful Yellow Fever (YF) vaccine consists of the live attenuated 17D-204 or 17DD viruses. Despite its excellent record of efficacy and safety, serious adverse events have been recorded and influenced extensive vaccination in endemic areas. Therefore, alternative strategies should be considered, which may include inactivated whole virus. High hydrostatic pressure has been described as a method for viral inactivation and vaccine development. The present study evaluated whether high hydrostatic pressure would inactivate the YF 17DD virus. YF 17DD virus was grown in Vero cells in roller bottle cultures and subjected to 310MPa for 3h at 4 degrees C. This treatment abolished YF infectivity and eliminated the ability of the virus to cause disease in mice. Pressure-inactivated virus elicited low level of neutralizing antibody titers although exhibited complete protection against an otherwise lethal challenge with 17DD virus in the murine model. The data warrant further development of pressure-inactivated vaccine against YF.
Collapse
Affiliation(s)
- Luciane P Gaspar
- Programa de Vacinas Virais, Instituto de Tecnologia em Imunobiológicos, Fundação Oswaldo Cruz, Rio de Janeiro, RJ, Brazil.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Martins MÂ, Silva ML, Elói-Santos SM, Ribeiro JGL, Peruhype-Magalhães V, Marciano APV, Homma A, Kroon EG, Teixeira-Carvalho A, Martins-Filho OA. Innate immunity phenotypic features point toward simultaneous raise of activation and modulation events following 17DD live attenuated yellow fever first-time vaccination. Vaccine 2008; 26:1173-84. [DOI: 10.1016/j.vaccine.2007.12.035] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2007] [Revised: 12/11/2007] [Accepted: 12/19/2007] [Indexed: 11/16/2022]
|
36
|
General immunization practices. Vaccines (Basel) 2008. [DOI: 10.1016/b978-1-4160-3611-1.50011-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] Open
|
37
|
Barnett ED, Kozarsky PE, Steffen R. Vaccines for international travel. Vaccines (Basel) 2008. [DOI: 10.1016/b978-1-4160-3611-1.50069-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
38
|
|
39
|
Borges MBJ, Kato SEM, Damaso CRA, Moussatché N, da Silva Freire M, Lambert Passos SR, do Nascimento JP. Accuracy and repeatability of a micro plaque reduction neutralization test for vaccinia antibodies. Biologicals 2007; 36:105-10. [PMID: 17892944 DOI: 10.1016/j.biologicals.2007.07.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2006] [Revised: 06/22/2007] [Accepted: 07/02/2007] [Indexed: 10/22/2022] Open
Abstract
The detection of neutralizing antibodies against vaccinia virus is a valuable tool for the investigation of previous smallpox vaccination. Compulsory smallpox vaccination ended in Brazil during the early 1970s, although the vaccine was available until the late 1970s. The threat of smallpox as a biological weapon has called the attention of public health authorities to the need for an evaluation of the immune status of the population. Based on our previous experience with a micro plaque reduction neutralization test (PRNT) for the evaluation of yellow fever immunity, a similar test was developed for the detection and quantification of vaccinia neutralizing antibodies. A cross-sectional study to test the repeatability and validity of plaque reduction neutralization test (PRNT) for vaccinia antibodies was performed in 182 subjects divided into two categories: subjects above 31 years old and the other > or = 35 years old. Cases were subjects considered to have been vaccinated with vaccinia virus if they declared vaccination history or evidenced vaccination marks. The assay is carried out in 96-well plates, provides results within 30 h, is easily performed, has good sensitivity (92.7%) and specificity (90.8), excellent repeatability (ICC 0.89 (0.88; 0.92)) and is thus suitable for use in mass screening of a population's antibody levels.
Collapse
Affiliation(s)
- Maria Beatriz J Borges
- Vice-diretoria de Desenvolvimento Tecnológico, Bio-Manguinhos/FIOCRUZ, Avenida Brasil 4365, Manguinhos, 21040-900 Rio de Janeiro, RJ, Brazil.
| | | | | | | | | | | | | |
Collapse
|
40
|
Martins MA, Silva ML, Marciano APV, Peruhype-Magalhães V, Eloi-Santos SM, Ribeiro JGL, Correa-Oliveira R, Homma A, Kroon EG, Teixeira-Carvalho A, Martins-Filho OA. Activation/modulation of adaptive immunity emerges simultaneously after 17DD yellow fever first-time vaccination: is this the key to prevent severe adverse reactions following immunization? Clin Exp Immunol 2007; 148:90-100. [PMID: 17309541 PMCID: PMC1868854 DOI: 10.1111/j.1365-2249.2006.03317.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Over past decades the 17DD yellow fever vaccine has proved to be effective in controlling yellow fever and promises to be a vaccine vector for other diseases, but the cellular and molecular mechanisms by which it elicits such broad-based immunity are still unclear. In this study we describe a detailed phenotypic investigation of major and minor peripheral blood lymphocyte subpopulations aimed at characterizing the kinetics of the adaptive immune response following primary 17DD vaccination. Our major finding is a decreased frequency of circulating CD19+ cells at day 7 followed by emerging activation/modulation phenotypic features (CD19+interleukin(IL)10R+/CD19+CD32+) at day 15. Increased frequency of CD4+human leucocyte antigen D-related(HLA-DR+) at day 7 and CD8+HLA-DR+ at day 30 suggest distinct kinetics of T cell activation, with CD4+ T cells being activated early and CD8+ T cells representing a later event following 17DD vaccination. Up-regulation of modulatory features on CD4+ and CD8+ cells at day 15 seems to be the key event leading to lower frequency of CD38+ T cells at day 30. Taken together, our findings demonstrate the co-existence of phenotypic features associated with activation events and modulatory pathways. Positive correlations between CD4+HLA-DR+ cells and CD4+CD25high regulatory T cells and the association between the type 0 chemokine receptor CCR2 and the activation status of CD4+ and CD8+ cells further support this hypothesis. We hypothesize that this controlled microenviroment seems to be the key to prevent the development of serious adverse events, and even deaths, associated with the 17DD vaccine reported in the literature.
Collapse
Affiliation(s)
- M A Martins
- Laboratório de Doença de Chagas, Centro de Pesquisas René Rachou, Fundação Oswaldo Cruz, Brazil
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Tuboi SH, Costa ZGA, da Costa Vasconcelos PF, Hatch D. Clinical and epidemiological characteristics of yellow fever in Brazil: analysis of reported cases 1998-2002. Trans R Soc Trop Med Hyg 2006; 101:169-75. [PMID: 16814821 DOI: 10.1016/j.trstmh.2006.04.001] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2006] [Revised: 04/03/2006] [Accepted: 04/03/2006] [Indexed: 10/24/2022] Open
Abstract
Yellow fever (YF), an arboviral infection of major public health importance in Brazil, is associated with high mortality and high epidemic potential. We analysed confirmed YF cases from the National Surveillance System from 1998-2002 and assessed risk factors for death among hospitalised patients. Variables assessed included age, gender, clinical signs and laboratory findings. A logistic regression model was used to identify independent predictors of death among hospitalised patients. From 1998-2002, among 2117 suspected YF cases reported to Brazil's Ministry of Health, 251 (11.9%) had confirmed YF, of whom 217 (86.5%) were hospitalised and the case fatality rate was 44.2%. Factors associated with higher mortality in univariate analysis included male gender (relative risk (RR) 1.96, 95% CI 1.17-2.28), age >40 years (RR 2.61, 95% CI 1.25-5.45), jaundice (RR 2.66, 95% CI 2.12-3.35), serum aspartate aminotransferase (AST) >1200 IU/l (RR 1.84, 95% CI 1.23-2.74), alanine aminotransferase >1500 IU/l (RR 2.09, 95% CI 1.38-3.17), total bilirubin >7.0mg/dl (RR 2.33, 95% CI 1.44-3.78), direct bilirubin >5.0mg/dl (RR 2.29, 95% CI 1.33-3.94) and blood urea nitrogen >100mg/dl (RR 5.77, 95% CI 1.43-23.22). In multivariate analysis, elevated AST and jaundice remained independently associated with higher mortality. These findings suggest that selected clinical and laboratory indicators may help clinicians recognise potentially fatal cases of YF.
Collapse
Affiliation(s)
- Suely Hiromi Tuboi
- Secretaria de Vigilância em Saúde, Ministry of Health, Brasília, Distrito Federal, Brazil
| | | | | | | |
Collapse
|
42
|
Galler R, Marchevsky RS, Caride E, Almeida LFC, Yamamura AMY, Jabor AV, Motta MCA, Bonaldo MC, Coutinho ESF, Freire MS. Attenuation and immunogenicity of recombinant yellow fever 17D-dengue type 2 virus for rhesus monkeys. Braz J Med Biol Res 2005; 38:1835-46. [PMID: 16302098 DOI: 10.1590/s0100-879x2005001200012] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
A chimeric yellow fever (YF)-dengue serotype 2 (dengue 2) virus was constructed by replacing the premembrane and envelope genes of the YF 17D virus with those from dengue 2 virus strains of Southeast Asian genotype. The virus grew to high titers in Vero cells and, after passage 2, was used for immunogenicity and attenuation studies in rhesus monkeys. Subcutaneous immunization of naive rhesus monkeys with the 17D-D2 chimeric virus induced a neutralizing antibody response associated with the protection of 6 of 7 monkeys against viremia by wild-type dengue 2 virus. Neutralizing antibody titers to dengue 2 were significantly lower in YF-immune animals than in YF-naive monkeys and protection against challenge with wild-type dengue 2 virus was observed in only 2 of 11 YF-immune monkeys. An anamnestic response to dengue 2, indicated by a sharp increase of neutralizing antibody titers, was observed in the majority of the monkeys after challenge with wild-type virus. Virus attenuation was demonstrated using the standard monkey neurovirulence test. The 17D-D2 chimera caused significantly fewer histological lesions than the YF 17DD virus. The attenuated phenotype could also be inferred from the limited viremias compared to the YF 17DD vaccine. Overall, these results provide further support for the use of chimeric viruses for the development of a new live tetravalent dengue vaccine.
Collapse
Affiliation(s)
- R Galler
- Departamento de Bioquímica e Biologia Molecular, Instituto Oswaldo Cruz, FIOCRUZ, 21045-900 Rio de Janeiro, RJ, Brazil.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Freire MS, Mann GF, Marchevsky RS, Yamamura AMY, Almeida LFC, Jabor AV, Malachias JMN, Coutinho ESF, Galler R. Production of yellow fever 17DD vaccine virus in primary culture of chicken embryo fibroblasts: yields, thermo and genetic stability, attenuation and immunogenicity. Vaccine 2005; 23:2501-12. [PMID: 15752837 DOI: 10.1016/j.vaccine.2004.10.035] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2004] [Revised: 10/04/2004] [Accepted: 10/08/2004] [Indexed: 10/26/2022]
Abstract
While a good vaccine against yellow fever (YF) virus has been available for decades, the basic technology for the production of YF vaccine in chicken embryos has remained substantially unchanged since the 1940s. Here we describe the highly efficient and economic production of the 17DD strain of YF virus in chicken embryo fibroblast (CEF) cell cultures with viral titers ranging from 6.3 to 6.7 log10PFU/mL. Thermostability of two different formulations (5 and 50-dose vials) of the CEF vaccine virus was found to be as high as the current vaccines retaining the minimal titer required for YF 17D vaccines. The production passage in CEF did not lead to the selection of genetic variants as shown by nucleotide sequence analyses of the CEF-derived vaccine lots or the sequence of viruses recovered from monkeys experimentally inoculated with the CEF virus. YF 17DD virus produced in CEF was also indistinguishable from its seed lot virus parent in terms of plaque size and immunogenicity in mice and monkeys. Comparison of the CEF virus and the seed lot virus made in chicken embryo in the internationally accepted monkey neurovirulence test (MNVT) revealed a higher clinical score for the former. The differences in central nervous system (CNS) histological scores for monkeys inoculated with the chicken embryo and experimental CEF vaccines were at the borderline level of statistical significance. These data warrant further studies on the monkey attenuation of other batches of CEF-derived vaccines.
Collapse
Affiliation(s)
- Marcos S Freire
- Fundacao Oswaldo Cruz, Instituto de Tecnologia em Imunobiológicos, Avenida Brasil 4365, Manguinhos, Rio de Janeiro 21045-900, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Bonaldo MC, Garratt RC, Marchevsky RS, Coutinho ESF, Jabor AV, Almeida LFC, Yamamura AMY, Duarte AS, Oliveira PJ, Lizeu JOP, Camacho LAB, Freire MS, Galler R. Attenuation of recombinant yellow fever 17D viruses expressing foreign protein epitopes at the surface. J Virol 2005; 79:8602-13. [PMID: 15956601 PMCID: PMC1143750 DOI: 10.1128/jvi.79.13.8602-8613.2005] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The yellow fever (YF) 17D vaccine is a live attenuated virus. Three-dimensional (3D) homology modeling of the E protein structure from YF 17D virus and its comparison with that from tick-borne encephalitis virus revealed that it is possible to accommodate inserts of different sizes and amino acid compositions in the flavivirus E protein fg loop. This is consistent with the 3D structures of both the dimeric and trimeric forms in which the fg loop lies exposed to solvents. We demonstrate here that YF 17D viruses bearing foreign humoral (17D/8) and T-cell (17D/13) epitopes, which vary in sequence and length, displayed growth restriction. It is hypothesized that interference with the dimer-trimer transition and with the formation of a ring of such trimers in order to allow fusion compromises the capability of the E protein to induce fusion of viral and endosomal membranes, and a slower rate of fusion may delay the extent of virus production. This would account for the lower levels of replication in cultured cells and of viremia in monkeys, as well as for the more attenuated phenotype of the recombinant viruses in monkeys. Testing of both recombinant viruses (17D/8 and 17D/13) for monkey neurovirulence also suggests that insertion at the 17D E protein fg loop does not compromise the attenuated phenotype of YF 17D virus, further confirming the potential use of this site for the development of new live attenuated 17D virus-based vaccines.
Collapse
Affiliation(s)
- Myrna C Bonaldo
- Fundação Oswaldo Cruz, Instituto Oswaldo Cruz, Departamento de Bioquimica e Biologia Molecular, Rio de Janeiro, RJ, Brazil
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Santos APD, Bertho AL, Dias DC, Santos JR, Marcovistz R. Lymphocyte subset analyses in healthy adults vaccinated with yellow fever 17DD virus. Mem Inst Oswaldo Cruz 2005; 100:331-7. [PMID: 16113878 DOI: 10.1590/s0074-02762005000300021] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In this study the kinetics of humoral and cellular immune responses in first-time vaccinees and re-vaccinees with the yellow fever 17DD vaccine virus was analyzed. Flow cytometric analyses were used to determine percentual values of T and B cells in parallel to the yellow fever neutralizing antibody production. All lymphocyte subsets analyzed were augmented around the 30th post vaccination day, both for first-time vaccinees and re-vaccinees. CD3+ T cells increased from 30.8% (SE +/- 4%) to 61.15% (SE +/- 4.2%), CD4+ T cells from 22.4% (SE +/- 3.6%) to 39.17% (SE +/- 2%) with 43% of these cells corresponding to CD4+CD45RO+ T cells, CD8+ T cells from 15.2% (SE +/- 2.9%) to 27% (SE +/- 3%) with 70% corresponding to CD8+CD45RO+ T cells in first-time vaccinees. In re-vaccinees, the CD3+ T cells increased from 50.7% (SE +/- 3%) to 80% (SE +/- 2.3%), CD4+ T cells from 24.9% (SE +/- 1.4%) to 40% (SE +/- 3%) presenting a percentage of 95% CD4+CD45RO+ T cells, CD8+ T cells from 19.7% (SE +/- 1.8%) to 25% (SE +/- 2%). Among CD8+CD38+ T cells there could be observed an increase from 15 to 41.6% in first-time vaccinees and 20.7 to 62.6% in re-vaccinees. Regarding neutralizing antibodies, the re-vaccinees presented high titers even before re-vaccination. The levels of neutralizing antibodies of first-time vaccinees were similar to those presented by re-vaccinees at day 30 after vaccination, indicating the success of primary vaccination. Our data provide a basis for further studies on immunological behavior of the YF 17DD vaccine.
Collapse
Affiliation(s)
- Ana Paula dos Santos
- Laboratório de Tecnologia Imunológica, Bio-Manguinhos, Fiocruz, 21045-900 Rio de Janeiro, RJ, Brazil
| | | | | | | | | |
Collapse
|
46
|
Fletcher MA, Fabre P, Debois H, Saliou P. Vaccines administered simultaneously: directions for new combination vaccines based on an historical review of the literature. Int J Infect Dis 2004; 8:328-38. [PMID: 15494254 DOI: 10.1016/j.ijid.2004.03.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2003] [Revised: 03/03/2004] [Accepted: 03/09/2004] [Indexed: 11/20/2022] Open
Abstract
OBJECTIVES The recognized benefits of administering vaccines simultaneously has encouraged vaccine producers to develop combination vaccines. If contemporary research and development can realize vaccines that achieve the current standards for safety, immunogenicity, and efficacy, other specific vaccine associations may also merit reconsideration as combination vaccines. METHODS An historical review of the vaccine association literature reveals two important themes: first, the programs of mass vaccination, in particular, the eradication of smallpox, sessions where multiple vaccines (other than the smallpox vaccine) were given concurrently, and the Expanded Programme on Immunization (EPI); and, second, the domain of travel vaccines, including travellers to a disease-endemic country (such as migrants, tourists, military personnel, or expatriates) and WHO requirements for international travellers. RESULTS/CONCLUSIONS Based on this historical review, combination vaccines worth reconsideration could fill epidemiologic niches in the EPI with, for instance, a measles--yellow fever, a measles--Japanese encephalitis or a pertussis-based paediatric combination rabies vaccine. Furthermore, other combinations could broaden protection against the pathogens responsible for meningitis, pneumonia, or enteric diseases. Nevertheless, complex issues such as necessity, feasibility, or affordability will ultimately determine if any one of these becomes a combination vaccine.
Collapse
Affiliation(s)
- Mark A Fletcher
- Wyeth Vaccines Research, Coeur Défense-Tour A, 110 esplanade du Général de Gaulle, 92931 Paris la Défense, Cedex, France.
| | | | | | | |
Collapse
|
47
|
Camacho LAB, Freire MDS, Leal MDLF, Aguiar SGD, Nascimento JPD, Iguchi T, Lozana JDA, Farias RHG. Immunogenicity of WHO-17D and Brazilian 17DD yellow fever vaccines: a randomized trial. Rev Saude Publica 2004; 38:671-8. [PMID: 15499438 DOI: 10.1590/s0034-89102004000500009] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
OBJECTIVE: To compare the immunogenicity of three yellow fever vaccines from WHO-17D and Brazilian 17DD substrains (different seed-lots). METHODS: An equivalence trial was carried out involving 1,087 adults in Rio de Janeiro. Vaccines produced by Bio-Manguinhos, Fiocruz (Rio de Janeiro, Brazil) were administered following standardized procedures adapted to allow blocked randomized allocation of participants to coded vaccine types (double-blind). Neutralizing yellow fever antibody titters were compared in pre- and post-immunization serum samples. Equivalence was defined as a difference of no more than five percentage points in seroconversion rates, and ratio between Geometric Mean Titters (GMT) higher than 0.67. RESULTS: Seroconversion rates were 98% or higher among subjects previously seronegative, and 90% or more of the total cohort of vaccinees, including those previously seropositive. Differences in seroconversion ranged from -0.05% to -3.02%. The intensity of the immune response was also very similar across vaccines: 14.5 to 18.6 IU/mL. GMT ratios ranged from 0.78 to 0.93. Taking the placebo group into account, the vaccines explained 93% of seroconversion. Viremia was detected in 2.7% of vaccinated subjects from Day 3 to Day 7. CONCLUSIONS: The equivalent immunogenicity of yellow fever vaccines from the 17D and 17DD substrains was demonstrated for the first time in placebo-controlled double-blind randomized trial. The study completed the clinical validation process of a new vaccine seed-lot, provided evidence for use of alternative attenuated virus substrains in vaccine production for a major manufacturer, and for the utilization of the 17DD vaccine in other countries.
Collapse
|
48
|
Rabello A, Orsini M, Disch J, Marcial T, Leal Md MDLF, Freire Md MDS, Yamamura AMY, Viana A. Low frequency of side effects following an incidental 25 times concentrated dose of yellow fever vaccine. Rev Soc Bras Med Trop 2002; 35:177-80. [PMID: 12011927 DOI: 10.1590/s0037-86822002000200008] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In August/1999, a group of 14 adults from the staff of a private hospital in Contagem - Minas Gerais State, Brazil, received unintentionally a 25 times concentrated dose of the 17-DD yellow fever vaccine (Bio-Manguinhos), due to a mistake at the reconstitution step. All patients were clinically and laboratorially evaluated at days 5, 13 and 35 post vaccination. Frequency of side effects and clinical observations of this group of individuals were not different from the observed in recipients immunized with normal doses of the vaccine. At the second and third evaluation none of the subjects reported symptoms. None of the patients presented abnormalities at the physical examination at none of the time points and in all cases the blood examination was normal, except for a reduced number of platelets that was detected in one subject at the first and second evaluation and reverted to normal at third evaluation. At the first evaluation point, 8 subjects were serum negative and 6 serum positive for yellow fever at the plaque reduction neutralization test. In 5 subjects the observed titre was 10 times higher as the baseline of 2.36 Log10 mUI/ml. The samples collected at second and third evaluation (13th and 35th days) demonstrated that all subjects responded to the vaccination with the exception of one that did not present a positive result in any of the samples collected. This evaluation confirms the safety of the 17-DD yellow fever vaccine.
Collapse
Affiliation(s)
- Ana Rabello
- Laboratório de Pesquisas Clínicas, Centro de Pesquisas René Rachou, Belo Horizonte, MG, Brasil
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Bonaldo MC, Garratt RC, Caufour PS, Freire MS, Rodrigues MM, Nussenzweig RS, Galler R. Surface expression of an immunodominant malaria protein B cell epitope by yellow fever virus. J Mol Biol 2002; 315:873-85. [PMID: 11812154 DOI: 10.1006/jmbi.2001.5258] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The yellow fever 17D virus (YF17D) has several characteristics that are desirable for the development of new, live attenuated vaccines. We approached its development as a vector for heterologous antigens by studying the expression of a humoral epitope at the surface of the E protein based on the results of modelling its three-dimensional structure. This model indicated that the most promising insertion site is between beta-strands f and g, a site that is exposed at the external surface of the virus. The large deletion of six residues from the fg loop of the E protein from yellow fever virus, compared to tick-born encephalitis virus, leaves space at the dimer interface for a large insertion without creating steric hindrance. We have tested this hypothesis by inserting a model humoral epitope from the circumsporozoite protein of Plasmodium falciparum consisting of triple NANP repeats. Recombinant virus (17D/8) expressing this insertion flanked by two glycine residues at each end, is specifically neutralized by a monoclonal antibody to the model epitope. Furthermore, mouse antibodies raised to the recombinant virus recognize the parasite protein in an ELISA assay. Serial passage analysis confirmed the genetic stability of the insertion made in the viral genome and the resulting 17D/8 virus is significantly more attenuated in mouse neurovirulence tests than the 17DD vaccine. The fg loop belongs to the dimerization domain of the E protein and lies at the interface between monomers. This domain undergoes a low pH transition, which is related to the fusion of the viral envelope to the endosome membrane. It is conceivable that a slower rate of fusion, resulting from the insertion close to the dimer interface, may delay the onset of virus production and thereby lead to a milder infection of the host. This would account for the more attenuated phenotype of the recombinant virus in the mouse model and lower extent of replication in cultured cells. The vectorial capacity of the yellow fever virus is being further explored for the expression and presentation of other epitopes, including those mediating T-cell responses.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antibodies, Monoclonal/immunology
- Antigens, Protozoan/chemistry
- Antigens, Protozoan/genetics
- Antigens, Protozoan/immunology
- Cell Line
- Epitopes, B-Lymphocyte/chemistry
- Epitopes, B-Lymphocyte/genetics
- Epitopes, B-Lymphocyte/immunology
- Gene Expression
- Genome, Viral
- Hydrogen-Ion Concentration
- Immunodominant Epitopes/chemistry
- Immunodominant Epitopes/genetics
- Immunodominant Epitopes/immunology
- Malaria/immunology
- Malaria/parasitology
- Malaria Vaccines/administration & dosage
- Malaria Vaccines/adverse effects
- Malaria Vaccines/genetics
- Malaria Vaccines/immunology
- Mice
- Models, Molecular
- Molecular Sequence Data
- Neutralization Tests
- Plasmodium falciparum/chemistry
- Plasmodium falciparum/genetics
- Plasmodium falciparum/immunology
- Protein Structure, Tertiary
- Sequence Alignment
- Serial Passage
- Survival Rate
- Vaccines, Attenuated/adverse effects
- Vaccines, Attenuated/genetics
- Vaccines, Attenuated/immunology
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/adverse effects
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
- Viral Envelope Proteins/chemistry
- Viral Envelope Proteins/genetics
- Viral Envelope Proteins/immunology
- Yellow fever virus/genetics
- Yellow fever virus/isolation & purification
- Yellow fever virus/pathogenicity
- Yellow fever virus/physiology
Collapse
Affiliation(s)
- Myrna C Bonaldo
- Departamento de Bioquímica e Biologia Molecular, Fundação Oswaldo Cruz Instituto Oswaldo Cruz, Rio de Janeiro, RJ 21045-900, Brazil
| | | | | | | | | | | | | |
Collapse
|
50
|
Galler R, Pugachev KV, Santos CL, Ocran SW, Jabor AV, Rodrigues SG, Marchevsky RS, Freire MS, Almeida LF, Cruz AC, Yamamura AM, Rocco IM, da Rosa ES, Souza LT, Vasconcelos PF, Guirakhoo F, Monath TP. Phenotypic and molecular analyses of yellow fever 17DD vaccine viruses associated with serious adverse events in Brazil. Virology 2001; 290:309-19. [PMID: 11883195 DOI: 10.1006/viro.2001.1168] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The yellow fever (YF) 17D virus is one of the most successful vaccines developed to data. Its use has been estimated to be over 400 million doses with an excellent record of safety. In the past 3 years, yellow fever vaccination was intensified in Brazil in response to higher risk of urban outbreaks of the disease. Two fatal adverse events temporally associated with YF vaccination were reported. Both cases had features similar to yellow fever disease, including hepatitis and multiorgan failure. Two different lots of YF 17DD virus vaccine were administered to the affected patients and also to hundreds of thousands of other individuals without any other reported serious adverse events. The lots were prepared from the secondary seed, which has been in continuous use since 1984. Nucleotide sequencing revealed minor variations at some nucleotide positions between the secondary seed lot virus and the virus isolates from patients; these differences were not consistent across the isolates, represented differences in the relative amount of each nucleotide in a heterogeneous position, and did not result in amino acid substitutions. Inoculation of rhesus monkeys with the viruses isolated from the two patients by the intracerebral (ic) or intrahepatic (ih) route caused minimal viremia and no clinical signs of infection or alterations in laboratory markers. Central nervous system histological scores of rhesus monkeys inoculated ic were within the expected range, and there were no histopathological lesions in animals inoculated ih. Altogether, these results demonstrated the genetic stability and attenuated phenotype of the viruses that caused fatal illness in the two patients. Therefore, the fatal adverse events experienced by the vaccinees are related to individual, genetically determined host factors that regulate cellular susceptibility to yellow fever virus. Such increased susceptibility, resulting in clinically overt disease expression, appears to be extremely rare.
Collapse
Affiliation(s)
- R Galler
- Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, 21045-900, Rio de Janeiro, RJ, Brazil.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|