1
|
Peno C, Jagne YJ, Clerc M, Balcazar Lopez C, Armitage EP, Sallah H, Drammeh S, Senghore E, Goderski G, van Tol S, Meijer A, Ruiz-Rodriguez A, de Steenhuijsen Piters W, de Koff E, Jarju S, Lindsey BB, Camara J, Bah S, Mohammed NI, Kampmann B, Clarke E, Dockrell DH, de Silva TI, Bogaert D. Interactions between live attenuated influenza vaccine and nasopharyngeal microbiota among children aged 24-59 months in The Gambia: a phase 4, open-label, randomised controlled trial. THE LANCET. MICROBE 2025; 6:100971. [PMID: 39832517 DOI: 10.1016/j.lanmic.2024.100971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 05/13/2024] [Accepted: 08/02/2024] [Indexed: 01/22/2025]
Abstract
BACKGROUND Live attenuated influenza vaccines (LAIVs) alter nasopharyngeal microbiota in adults. It is poorly understood why LAIV immunogenicity varies across populations, but it could be linked to the microbiome. We aimed to investigate the interactions between intranasal immunisation with LAIV and nasopharyngeal microbiota composition in children from The Gambia. METHODS We conducted a phase 4, open-label, randomised controlled trial in Sukuta, The Gambia. Children aged 24-59 months with no underlying illness or history of respiratory illness for at least 14 days before recruitment were eligible. Participants were randomly assigned (2:1) by use of a computer-generated sequence in permuted blocks of 15, stratified by sex, to receive trivalent LAIV either on day 0 (intervention group) or after active follow-up at day 21 (control group). The investigator team was initially masked to block size and randomisation sequence; however, group allocation was later revealed to the team. Microbiome profiles were characterised from nasopharyngeal samples collected from all participants on days 0, 7, and 21 by use of 16S rRNA sequencing. The primary outcomes were the effect of LAIV on nasopharyngeal microbiome profiles on day 7 and day 21, and the association between the nasopharyngeal microbiome at baseline and LAIV-induced mucosal IgA responses at day 21, assessed with permutational ANOVA tests. Asymptomatic respiratory viral co-infection at baseline and year of recruitment (2017 or 2018) were included as covariates. This trial is registered with ClinicalTrials.gov (NCT02972957) and is closed. FINDINGS Between Feb 8 and April 12, 2017, and Jan 15 and March 28, 2018, 343 children were screened for eligibility, of whom 220 (64%) children were randomly assigned to the intervention group and 110 (32%) to the control group. 213 (97%) children in the intervention group and 108 (98%) in the control group completed the study and were included in the final analysis. Although we did not observe an independent effect of LAIV on microbial community composition at days 7 or 21, we found that LAIV had an effect dependent on the year of recruitment. LAIV affected microbial community composition in 2018 (R2 1·97% [95% CI 0·85-5·94]; p=0·037), but not in 2017 (1·23% [0·49-4·46]; p=0·091). We also found that viral co-infection at baseline had an effect on microbial composition at day 7, regardless of recruitment year (R2 1·01% [95% CI 0·28-3·01]; p=0·026). Nasopharyngeal microbial community composition at baseline had no effect on mucosal IgA responses to LAIV administration (R2 0·51% [95% CI 0·23-2·49]; p=0·46). INTERPRETATION Our findings suggest that the effect of LAIVs on nasopharyngeal microbiota composition in children is modest and temporary; therefore, LAIVs could be used as an intervention to curb influenza in children from low-income and middle-income countries, without causing long-lasting perturbations in nasopharyngeal microbiota. However, nasopharyngeal microbiota at the time of vaccination might not explain the variability observed between individuals in LAIV-induced IgA responses. FUNDING The Wellcome Trust, UK National Institute for Health Research, and Chief Scientist Office Scotland.
Collapse
Affiliation(s)
- Chikondi Peno
- Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK; Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Ya Jankey Jagne
- Vaccines and Immunity Theme, Medical Research Council Unit The Gambia at London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Melanie Clerc
- Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Carlos Balcazar Lopez
- Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Edwin P Armitage
- Vaccines and Immunity Theme, Medical Research Council Unit The Gambia at London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Hadijatou Sallah
- Vaccines and Immunity Theme, Medical Research Council Unit The Gambia at London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Sainabou Drammeh
- Vaccines and Immunity Theme, Medical Research Council Unit The Gambia at London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Elina Senghore
- Vaccines and Immunity Theme, Medical Research Council Unit The Gambia at London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Gabriel Goderski
- Centre for Infectious Disease Research, Diagnostics and Laboratory Surveillance, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Sophie van Tol
- Centre for Infectious Disease Research, Diagnostics and Laboratory Surveillance, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Adam Meijer
- Centre for Infectious Disease Research, Diagnostics and Laboratory Surveillance, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Alicia Ruiz-Rodriguez
- Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Wouter de Steenhuijsen Piters
- Department of Paediatric Immunology and Infectious Diseases, Wilhelmina Children's Hospital-University Medical Center Utrecht, Utrecht, Netherlands
| | - Emma de Koff
- Department of Paediatric Immunology and Infectious Diseases, Wilhelmina Children's Hospital-University Medical Center Utrecht, Utrecht, Netherlands
| | - Sheikh Jarju
- Vaccines and Immunity Theme, Medical Research Council Unit The Gambia at London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Benjamin B Lindsey
- Vaccines and Immunity Theme, Medical Research Council Unit The Gambia at London School of Hygiene & Tropical Medicine, Banjul, The Gambia; Centre for International Child Health, Section of Paediatrics, Department of Medicine, Imperial College London, London, UK
| | - Janko Camara
- Vaccines and Immunity Theme, Medical Research Council Unit The Gambia at London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Sulayman Bah
- Vaccines and Immunity Theme, Medical Research Council Unit The Gambia at London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Nuredin I Mohammed
- Vaccines and Immunity Theme, Medical Research Council Unit The Gambia at London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Beate Kampmann
- Vaccines and Immunity Theme, Medical Research Council Unit The Gambia at London School of Hygiene & Tropical Medicine, Banjul, The Gambia; The Vaccine Centre, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK
| | - Ed Clarke
- Vaccines and Immunity Theme, Medical Research Council Unit The Gambia at London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - David H Dockrell
- Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Thushan I de Silva
- Vaccines and Immunity Theme, Medical Research Council Unit The Gambia at London School of Hygiene & Tropical Medicine, Banjul, The Gambia; Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, UK
| | - Debby Bogaert
- Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK; Department of Paediatric Immunology and Infectious Diseases, Wilhelmina Children's Hospital-University Medical Center Utrecht, Utrecht, Netherlands.
| |
Collapse
|
2
|
Hao S, Tomic I, Lindsey BB, Jagne YJ, Hoschler K, Meijer A, Quiroz JMC, Meade P, Sano K, Peno C, Costa-Martins AG, Bogaert D, Kampmann B, Nakaya H, Krammer F, de Silva TI, Tomic A. Integrative Mapping of Pre-existing Immune Landscapes for Vaccine Response Prediction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.22.634302. [PMID: 39896552 PMCID: PMC11785181 DOI: 10.1101/2025.01.22.634302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Predicting individual vaccine responses remains a significant challenge due to the complexity and variability of immune processes. To address this gap, we developed immunaut, an open-source, data-driven framework implemented as an R package specifically designed for all systems vaccinologists seeking to analyze and predict immunological outcomes across diverse vaccination settings. Leveraging one of the most comprehensive live attenuated influenza vaccine (LAIV) datasets to date - 244 Gambian children enrolled in a phase 4 immunogenicity study - immunaut integrates humoral, mucosal, cellular, transcriptomic, and microbiological parameters collected before and after vaccination, providing an unprecedentedly holistic view of LAIV-induced immunity. Through advanced dimensionality reduction, clustering, and predictive modeling, immunaut identifies distinct immunophenotypic responder profiles and their underlying baseline determinants. In this study, immunaut delineated three immunophenotypes: (1) CD8 T-cell responders, marked by strong baseline mucosal immunity and extensive prior influenza virus exposure that boosts memory CD8 T-cell responses, without generating influenza virus-specific antibody responses; (2) Mucosal responders, characterized by pre-existing systemic influenza A virus immunity (specifically to H3N2) and stable epithelial integrity, leading to potent mucosal IgA expansions and subsequent seroconversion to influenza B virus; and (3) Systemic, broad influenza A virus responders, who start with relatively naive immunity and leverage greater initial viral replication to drive broad systemic antibody responses against multiple influenza A virus variants beyond those included in the LAIV vaccine. By integrating pathway-level analysis, model-derived contribution scores, and hierarchical decision rules, immunaut elucidates how distinct immunological landscapes shape each response trajectory and how key baseline features, including pre-existing immunity, mucosal preparedness, and cellular support, dictate vaccine outcomes. Collectively, these findings emphasize the power of integrative, predictive frameworks to advance precision vaccinology, and highlight immunaut as a versatile, community-available resource for optimizing immunization strategies across diverse populations and vaccine platforms.
Collapse
Affiliation(s)
- Stephanie Hao
- Atomic lab, The National Emerging Infectious Diseases Laboratories (NEIDL), Boston University; Boston, MA, US
| | - Ivan Tomic
- Atomic lab, The National Emerging Infectious Diseases Laboratories (NEIDL), Boston University; Boston, MA, US
| | - Benjamin B Lindsey
- The Florey Institute of Infection and NIHR Sheffield Biomedical Research Centre, University of Sheffield; Sheffield, UK
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Beech Hill Road; Sheffield, UK
| | - Ya Jankey Jagne
- Vaccines and Immunity Theme, Medical Research Council Unit The Gambia at the London School of Hygiene and Tropical Medicine; Fajara, The Gambia
| | - Katja Hoschler
- Respiratory Virus Unit, UK Health Security Agency; London, UK
| | - Adam Meijer
- National Institute for Public Health and the Environment; Bilthoven, The Netherlands
| | - Juan Manuel Carreño Quiroz
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, US
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, US
| | - Philip Meade
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, US
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, US
| | - Kaori Sano
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, US
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, US
| | - Chikondi Peno
- Centre for Inflammation Research, University of Edinburgh; Edinburgh, UK
| | - André G Costa-Martins
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo; São Paulo, Brazil
- Micromanufacturing Laboratory, Institute for Technological Research, São Paulo, Brazil
| | - Debby Bogaert
- Centre for Inflammation Research, University of Edinburgh; Edinburgh, UK
| | - Beate Kampmann
- Vaccines and Immunity Theme, London School of Hygiene & Tropical Medicine; London, UK
- Charité Centre for Global Health; Berlin, Germany
| | - Helder Nakaya
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo; São Paulo, Brazil
- Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, US
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, US
- Ignaz Semmelweis Institute, Interuniversity Institute for Infection Research, Medical University of Vienna, Vienna, Austria
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, US
| | - Thushan I de Silva
- The Florey Institute of Infection and NIHR Sheffield Biomedical Research Centre, University of Sheffield; Sheffield, UK
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Beech Hill Road; Sheffield, UK
- Vaccines and Immunity Theme, London School of Hygiene & Tropical Medicine; London, UK
| | - Adriana Tomic
- Atomic lab, The National Emerging Infectious Diseases Laboratories (NEIDL), Boston University; Boston, MA, US
- Department of Virology, Immunology & Microbiology, Boston University Medical School; Boston, MA, US
- Biomedical Engineering, Boston University, College of Engineering; Boston, MA, US
| |
Collapse
|
3
|
Gubbins S, Paudyal B, Dema B, Vats A, Ulaszewska M, Vatzia E, Tchilian E, Gilbert SC. Predicting airway immune responses and protection from immune parameters in blood following immunization in a pig influenza model. Front Immunol 2024; 15:1506224. [PMID: 39749329 PMCID: PMC11693722 DOI: 10.3389/fimmu.2024.1506224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 11/29/2024] [Indexed: 01/04/2025] Open
Abstract
Whereas the intranasally delivered influenza vaccines used in children affect transmission of influenza virus in the community as well as reducing illness, inactivated influenza vaccines administered by intramuscular injection do not prevent transmission and have a variable, sometimes low rate of vaccine effectiveness. Although mucosally administered vaccines have the potential to induce more protective immune response at the site of viral infection, quantitating such immune responses in large scale clinical trials and developing correlates of protection is challenging. Here we show that by using mathematical models immune responses measured in the blood after delivery of vaccine to the lungs by aerosol can predict immune responses in the respiratory tract in pigs. Additionally, these models can predict protection from influenza virus challenge despite lower levels of blood responses following aerosol immunization. However, the inclusion of immune responses measured in nasal swab eluates did not improve the predictive power of the model. Our models are an important first step, providing proof of principle that it is feasible to predict immune responses and protection in pigs. This approach now provides a path to develop correlates of protection for mucosally delivered vaccines in samples that are easily accessed in clinical trials.
Collapse
Affiliation(s)
| | | | - Barbara Dema
- Nuffield Department of Medicine, Pandemic Sciences Institute, University of Oxford, Oxford, United Kingdom
| | | | - Marta Ulaszewska
- Nuffield Department of Medicine, Pandemic Sciences Institute, University of Oxford, Oxford, United Kingdom
| | - Eleni Vatzia
- The Pirbright Institute, Pirbright, United Kingdom
| | | | - Sarah C. Gilbert
- Nuffield Department of Medicine, Pandemic Sciences Institute, University of Oxford, Oxford, United Kingdom
- Chinese Academy of Medical Science (CAMS), Oxford Institute (COI), University of Oxford, Oxford, United Kingdom
| |
Collapse
|
4
|
Zhang X, Zhang J, Chen S, He Q, Bai Y, Liu J, Wang Z, Liang Z, Chen L, Mao Q, Xu M. Progress and challenges in the clinical evaluation of immune responses to respiratory mucosal vaccines. Expert Rev Vaccines 2024; 23:362-370. [PMID: 38444382 DOI: 10.1080/14760584.2024.2326094] [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: 01/25/2024] [Accepted: 02/28/2024] [Indexed: 03/07/2024]
Abstract
INTRODUCTION Following the coronavirus disease pandemic, respiratory mucosal vaccines that elicit both mucosal and systemic immune responses have garnered increasing attention. However, human physiological characteristics pose significant challenges in the evaluation of mucosal immunity, which directly impedes the development and application of respiratory mucosal vaccines. AREAS COVERED This study summarizes the characteristics of immune responses in the respiratory mucosa and reviews the current status and challenges in evaluating immune response to respiratory mucosal vaccines. EXPERT OPINION Secretory Immunoglobulin A (S-IgA) is a major effector molecule at mucosal sites and a commonly used indicator for evaluating respiratory mucosal vaccines. However, the unique physiological structure of the respiratory tract pose significant challenges for the clinical collection and detection of S-IgA. Therefore, it is imperative to develop a sampling method with high collection efficiency and acceptance, a sensitive detection method, reference materials for mucosal antibodies, and to establish a threshold for S-IgA that correlates with clinical protection. Sample collection is even more challenging when evaluating mucosal cell immunity. Therefore, a mucosal cell sampling method with high operability and high tolerance should be established. Targets of the circulatory system capable of reflecting mucosal cellular immunity should also be explored.
Collapse
Affiliation(s)
- Xuanxuan Zhang
- State Key Laboratory of Drug Regulatory Science, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, China
| | - Jialu Zhang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Si Chen
- Drug and Vaccine Research Center, Guangzhou National Laboratory, Guangzhou, China
| | - Qian He
- State Key Laboratory of Drug Regulatory Science, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, China
| | - Yu Bai
- State Key Laboratory of Drug Regulatory Science, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, China
| | - Jianyang Liu
- State Key Laboratory of Drug Regulatory Science, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, China
| | - Zhongfang Wang
- Drug and Vaccine Research Center, Guangzhou National Laboratory, Guangzhou, China
| | - Zhenglun Liang
- State Key Laboratory of Drug Regulatory Science, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, China
| | - Ling Chen
- Drug and Vaccine Research Center, Guangzhou National Laboratory, Guangzhou, China
- State Key Laboratory of Respiratory Disease, Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Qunying Mao
- State Key Laboratory of Drug Regulatory Science, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, China
| | - Miao Xu
- State Key Laboratory of Drug Regulatory Science, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, China
| |
Collapse
|
5
|
Thwaites RS, Uruchurtu ASS, Negri VA, Cole ME, Singh N, Poshai N, Jackson D, Hoschler K, Baker T, Scott IC, Ros XR, Cohen ES, Zambon M, Pollock KM, Hansel TT, Openshaw PJM. Early mucosal events promote distinct mucosal and systemic antibody responses to live attenuated influenza vaccine. Nat Commun 2023; 14:8053. [PMID: 38052824 PMCID: PMC10697962 DOI: 10.1038/s41467-023-43842-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 11/22/2023] [Indexed: 12/07/2023] Open
Abstract
Compared to intramuscular vaccines, nasally administered vaccines have the advantage of inducing local mucosal immune responses that may block infection and interrupt transmission of respiratory pathogens. Live attenuated influenza vaccine (LAIV) is effective in preventing influenza in children, but a correlate of protection for LAIV remains unclear. Studying young adult volunteers, we observe that LAIV induces distinct, compartmentalized, antibody responses in the mucosa and blood. Seeking immunologic correlates of these distinct antibody responses we find associations with mucosal IL-33 release in the first 8 hours post-inoculation and divergent CD8+ and circulating T follicular helper (cTfh) T cell responses 7 days post-inoculation. Mucosal antibodies are induced separately from blood antibodies, are associated with distinct immune responses early post-inoculation, and may provide a correlate of protection for mucosal vaccination. This study was registered as NCT04110366 and reports primary (mucosal antibody) and secondary (blood antibody, and nasal viral load and cytokine) endpoint data.
Collapse
Affiliation(s)
- Ryan S Thwaites
- National Heart and Lung Institute, Imperial College London, London, UK.
| | | | - Victor Augusti Negri
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Megan E Cole
- Department of Infectious Disease, Imperial College London, London, UK
| | - Nehmat Singh
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Nelisa Poshai
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | | | - Tina Baker
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Ian C Scott
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Xavier Romero Ros
- Bioscience Asthma and Skin Immunity, Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Emma Suzanne Cohen
- Bioscience Asthma and Skin Immunity, Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Maria Zambon
- United Kingdom Health Security Agency, London, UK
| | - Katrina M Pollock
- Department of Infectious Disease, Imperial College London, London, UK
| | - Trevor T Hansel
- National Heart and Lung Institute, Imperial College London, London, UK
| | | |
Collapse
|
6
|
Hitchings MDT, Borgert BA, Shir A, Yang B, Grantz KH, Ball J, Moreno CA, Rand K, Small PA, Fowke KR, Cummings DAT. Dynamics of Anti-influenza Mucosal IgA Over a Season in a Cohort of Individuals Living or Working in a Long-term Care Facility. J Infect Dis 2023; 228:383-390. [PMID: 36740584 PMCID: PMC10428196 DOI: 10.1093/infdis/jiad029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 01/25/2023] [Accepted: 02/02/2023] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Serological surveys are used to ascertain influenza infection and immunity, but evidence for the utility of mucosal immunoglobulin A (IgA) as a correlate of infection or protection is limited. METHODS We performed influenza-like illness (ILI) surveillance on 220 individuals living or working in a retirement community in Gainesville, Florida from January to May 2018, and took pre- and postseason nasal samples of 11 individuals with polymerase chain reaction (PCR)-confirmed influenza infection and 60 randomly selected controls. Mucosal IgA against 10 strains of influenza was measured from nasal samples. RESULTS Overall, 28.2% and 11.3% of individuals experienced a 2-fold and 4-fold rise, respectively, in mucosal IgA to at least 1 influenza strain. Individuals with PCR-confirmed influenza A had significantly lower levels of preseason IgA to influenza A. Influenza-associated respiratory illness was associated with a higher rise in mucosal IgA to influenza strains of the same subtype, and H3N2-associated respiratory illness was associated with a higher rise in mucosal IgA to other influenza A strains. CONCLUSIONS By comparing individuals with and without influenza illness, we demonstrated that mucosal IgA is a correlate of influenza infection. There was evidence for cross-reactivity in mucosal IgA across influenza A subtypes.
Collapse
Affiliation(s)
- Matt D T Hitchings
- Department of Biostatistics, College of Public Health and Health Professions, University of Florida, Gainesville, Florida, USA
| | - Brooke A Borgert
- Department of Biology, University of Florida, Gainesville, Florida, USA
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida, USA
| | - Adam Shir
- Department of Biology, University of Florida, Gainesville, Florida, USA
| | - Bingyi Yang
- Department of Biology, University of Florida, Gainesville, Florida, USA
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida, USA
| | - Kyra H Grantz
- Department of Biology, University of Florida, Gainesville, Florida, USA
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida, USA
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Jacob Ball
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida, USA
- Department of Epidemiology, College of Public Health and Health Professions, University of Florida, Gainesville, Florida, USA
| | - Carlos A Moreno
- Department of Biology, University of Florida, Gainesville, Florida, USA
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida, USA
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Kenneth Rand
- Department of Pathology, University of Florida, Gainesville, Florida, USA
| | - Parker A Small
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida, USA
| | - Keith R Fowke
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Derek A T Cummings
- Department of Biology, University of Florida, Gainesville, Florida, USA
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
7
|
Aksyuk AA, Bansal H, Wilkins D, Stanley AM, Sproule S, Maaske J, Sanikommui S, Hartman WR, Sobieszczyk ME, Falsey AR, Kelly EJ. AZD1222-induced nasal antibody responses are shaped by prior SARS-CoV-2 infection and correlate with virologic outcomes in breakthrough infection. Cell Rep Med 2023; 4:100882. [PMID: 36610390 PMCID: PMC9750884 DOI: 10.1016/j.xcrm.2022.100882] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 11/11/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022]
Abstract
The nasal mucosa is an important initial site of host defense against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. However, intramuscularly administered vaccines typically do not achieve high antibody titers in the nasal mucosa. We measure anti-SARS-CoV-2 spike immunoglobulin G (IgG) and IgA in nasal epithelial lining fluid (NELF) following intramuscular vaccination of 3,058 participants from the immunogenicity substudy of a phase 3, double-blind, placebo-controlled study of AZD1222 vaccination (ClinicalTrials.gov: NCT04516746). IgG is detected in NELF collected 14 days following the first AZD1222 vaccination. IgG levels increase with a second vaccination and exceed pre-existing levels in baseline-SARS-CoV-2-seropositive participants. Nasal IgG responses are durable and display strong correlations with serum IgG, suggesting serum-to-NELF transudation. AZD1222 induces short-lived increases to pre-existing nasal IgA levels in baseline-seropositive vaccinees. Vaccinees display a robust recall IgG response upon breakthrough infection, with overall magnitudes unaffected by time between vaccination and illness. Mucosal responses correlate with reduced viral loads and shorter durations of viral shedding in saliva.
Collapse
Affiliation(s)
- Anastasia A Aksyuk
- Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878, USA
| | - Himanshu Bansal
- Biometrics, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878, USA
| | - Deidre Wilkins
- Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878, USA
| | - Ann Marie Stanley
- Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878, USA
| | - Stephanie Sproule
- Biometrics, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878, USA
| | - Jill Maaske
- Clinical Development, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878, USA
| | - Satya Sanikommui
- Biometrics, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878, USA
| | - William R Hartman
- Department of Anesthesiology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53726, USA
| | - Magdalena E Sobieszczyk
- Division of Infectious Diseases, Department of Medicine, Vagelos College of Physicians and Surgeons, New York Presbyterian/Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Ann R Falsey
- University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA; Rochester Regional Health, Rochester, NY 14621, USA.
| | - Elizabeth J Kelly
- Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878, USA.
| |
Collapse
|
8
|
Mu R, Huang Y, Bouquet J, Yuan J, Kubiak RJ, Ma E, Naser S, Mylott WR, Ismaiel OA, Wheeler AM, Burkart R, Cortes DF, Bruton J, Arends RH, Liang M, Rosenbaum AI. Multiplex Hybrid Antigen-Capture LC-MRM Quantification in Sera and Nasal Lining Fluid of AZD7442, a SARS-CoV-2-Targeting Antibody Combination. Anal Chem 2022; 94:14835-14845. [PMID: 36269894 DOI: 10.1021/acs.analchem.2c01320] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
AZD7442 (tixagevimab [AZD8895]/cilgavimab [AZD1061]) is a monoclonal antibody (mAb) combination in development for the prevention and treatment of coronavirus disease 2019. Traditionally, bioanalysis of mAbs is performed using ligand binding assays (LBAs), which offer sensitivity, robustness, and ease of implementation. However, LBAs frequently require generation of critical reagents that typically take several months. Instead, we developed a highly sensitive (5 ng/mL limit of quantification) method using a hybrid LBA-liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS) approach for quantification of the two codosed antibodies in serum and nasal lining fluid (NLF), a rare matrix. The method was optimized by careful selection of multiple reaction monitoring, capture reagents, magnetic beads, chromatographic conditions, evaluations of selectivity, and matrix effect. The final assay used viral spike protein receptor-binding domain as capture reagent and signature proteotypic peptides from the complementarity-determining region of each mAb for detection. In contrast to other methods of similar/superior sensitivity, our approach did not require multidimensional separations and can be operated in an analytical flow regime, ensuring high throughput and robustness required for clinical analysis at scale. The sensitivity of this method significantly exceeds typical sensitivity of ∼100 ng/mL for analytical flow 1D LBA-LC-MS/MS methods for large macromolecules, such as antibodies. Furthermore, infection and vaccination status did not impact method performance, ensuring method robustness and applicability to a broad patient population. This report demonstrated the general applicability of the hybrid LBA-LC-MS/MS approach to platform quantification of antibodies with high sensitivity and reproducibility, with specialized extension to matrices of increasing interest, such as NLF.
Collapse
Affiliation(s)
- Ruipeng Mu
- Integrated Bioanalysis, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, South San Francisco, California 94080, United States
| | - Yue Huang
- Integrated Bioanalysis, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, South San Francisco, California 94080, United States
| | - Jerome Bouquet
- Integrated Bioanalysis, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, South San Francisco, California 94080, United States
| | - Jiaqi Yuan
- Integrated Bioanalysis, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, South San Francisco, California 94080, United States
| | - Robert J Kubiak
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gaithersburg, Maryland 20878, United States
| | - Eric Ma
- Research and Development Biologics by LC-MS/MS, PPD Laboratories, Richmond, Virginia 23230, United States
| | - Sami Naser
- Research and Development Biologics by LC-MS/MS, PPD Laboratories, Richmond, Virginia 23230, United States
| | - William R Mylott
- Research and Development Biologics by LC-MS/MS, PPD Laboratories, Richmond, Virginia 23230, United States
| | - Omnia A Ismaiel
- Research and Development Biologics by LC-MS/MS, PPD Laboratories, Richmond, Virginia 23230, United States.,Faculty of Pharmacy, Zagazig University, Zagazig 2, Zagazig, Egypt
| | - Aaron M Wheeler
- Research and Development Biologics by LC-MS/MS, PPD Laboratories, Richmond, Virginia 23230, United States
| | - Rebecca Burkart
- Research and Development Biologics by LC-MS/MS, PPD Laboratories, Richmond, Virginia 23230, United States
| | - Diego F Cortes
- Research and Development Biologics by LC-MS/MS, PPD Laboratories, Richmond, Virginia 23230, United States
| | - James Bruton
- Research and Development Biologics by LC-MS/MS, PPD Laboratories, Richmond, Virginia 23230, United States
| | - Rosalinda H Arends
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gaithersburg, Maryland 20878, United States
| | - Meina Liang
- Integrated Bioanalysis, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, South San Francisco, California 94080, United States
| | - Anton I Rosenbaum
- Integrated Bioanalysis, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, South San Francisco, California 94080, United States
| |
Collapse
|
9
|
Clephane K, Wilson MC, Heiman JR, Craig AN, Lorenz T. Sexual violence history predicts changes in vaginal immune parameters during sexual arousal. Brain Behav Immun 2022; 104:171-180. [PMID: 35697156 PMCID: PMC9734281 DOI: 10.1016/j.bbi.2022.06.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 05/24/2022] [Accepted: 06/05/2022] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE To examine the influence of sexual arousal on vaginal mucosal inflammatory cytokine and antibody production in healthy women with and without histories of childhood and/or adult sexual violence. METHODS Ninety-one premenopausal healthy women (ages 18-42) attended a single laboratory session in which they provided vaginal fluid samples before and after viewing one neutral and one erotic film. While viewing the films, participants' vaginal sexual arousal was recorded using vaginal photoplethysmography. RESULTS Of the 91 participants, 41 (45%) reported no history of sexual violence, 17 (19%) reported a history of childhood sexual abuse (CSA) only, 19 (21%) reported a history of adult sexual assault (ASA) only, and 10 (11%) reported a history of both CSA and ASA, with 4 participants choosing not to provide information on their sexual violence history. For women with a history of ASA but not CSA, there was a significant increase in vaginal IL-1β following arousal, while for women with a history of CSA (with or without ASA), there was a significant decrease. Women without CSA histories had a significant increase in vaginal IgA following sexual arousal, while women with CSA histories had a decrease. CONCLUSION Sexual arousal possibly plays a role in modifying vaginal immune responses in young, healthy women. Moreover, these effects may vary depending upon sexual assault histories, such that relative to women without assault histories, women with a history of early life sexual trauma showed significantly altered vaginal immune responses following sexual arousal. If replicated, these findings may help explain the increased risk for sexually transmitted infections observed among women with sexual assault histories.
Collapse
Affiliation(s)
- Kirstin Clephane
- Department of Psychology, University of Nebraska-Lincoln, United States; Center for Brain, Biology & Behavior, University of Nebraska-Lincoln, United States
| | - M Claire Wilson
- Department of Psychological and Brain Sciences, Indiana University, United States
| | - Julia R Heiman
- Department of Psychological and Brain Sciences, Indiana University, United States
| | - Amber N Craig
- Medical College of Wisconsin, Department of Psychiatry and Behavioral Medicine, United States
| | - Tierney Lorenz
- Department of Psychology, University of Nebraska-Lincoln, United States; Center for Brain, Biology & Behavior, University of Nebraska-Lincoln, United States.
| |
Collapse
|
10
|
Haeusler IL, Daniel O, Isitt C, Watts R, Cantrell L, Feng S, Cochet M, Salloum M, Ikram S, Hayter E, Lim S, Hall T, Athaide S, Cosgrove CA, Tregoning JS, Le Doare K. Group B Streptococcus (GBS) colonisation is dynamic over time, whilst GBS capsular polysaccharides-specific antibody remains stable. Clin Exp Immunol 2022; 209:188-200. [PMID: 35802786 PMCID: PMC9390841 DOI: 10.1093/cei/uxac066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 06/08/2022] [Accepted: 06/27/2022] [Indexed: 11/20/2022] Open
Abstract
Group B Streptococcus (GBS) is a leading cause of adverse pregnancy outcomes due to invasive infection. This study investigated longitudinal variation in GBS rectovaginal colonization, serum and vaginal GBS capsular polysaccharide (CPS)-specific antibody levels. Non-pregnant women were recruited in the UK and were sampled every 2 weeks over a 12-week period. GBS isolates were taken from recto-vaginal swabs and serotyped by polymerase chain reaction. Serum and vaginal immunoglobulin G (IgG) and nasal immunoglobulin A (IgA) specific to CPS were measured by Luminex, and total IgG/A by ELISA. Seventy women were enrolled, of median age 26. Out of the 66 participants who completed at least three visits: 14/47 (29.8%) women that were GBS negative at screening became positive in follow-up visits and 16/19 (84.2%) women who were GBS positive at screening became negative. There was 50% probability of becoming negative 36 days after the first positive swab. The rate of detectable GBS carriage fluctuated over time, although serum, vaginal, and nasal CPS-specific antibody levels remained constant. Levels of CPS-specific antibodies were higher in the serum of individuals colonized with GBS than in non-colonized, but similar in the vaginal and nasal mucosa. We found correlations between antibody levels in serum and the vaginal and nasal mucosa. Our study demonstrates the feasibility of elution methods to retrieve vaginal and nasal antibodies, and the optimization of immunoassays to measure GBS-CPS-specific antibodies. The difference between the dynamics of colonization and antibody response is interesting and further investigation is required for vaccine development.
Collapse
Affiliation(s)
- I L Haeusler
- St George's University of London, Paediatric Infectious Diseases Research Group, London, United Kingdom
| | - O Daniel
- St George's University of London, Paediatric Infectious Diseases Research Group, London, United Kingdom
| | - C Isitt
- St George's University of London, The Vaccine Institute, London, United Kingdom
| | - R Watts
- St George's University of London, Paediatric Infectious Diseases Research Group, London, United Kingdom
| | - L Cantrell
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford
| | - S Feng
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford
| | - M Cochet
- St George's University of London, Paediatric Infectious Diseases Research Group, London, United Kingdom
| | - M Salloum
- St George's University of London, Paediatric Infectious Diseases Research Group, London, United Kingdom.,UnivLyon, Claude Bernard University Lyon I, France
| | - S Ikram
- St George's University of London, The Vaccine Institute, London, United Kingdom
| | - E Hayter
- St George's University of London, The Vaccine Institute, London, United Kingdom
| | - S Lim
- St George's University of London, Paediatric Infectious Diseases Research Group, London, United Kingdom
| | - T Hall
- St George's University of London, Paediatric Infectious Diseases Research Group, London, United Kingdom
| | - S Athaide
- St George's University of London, The Vaccine Institute, London, United Kingdom
| | - C A Cosgrove
- St George's University of London, The Vaccine Institute, London, United Kingdom
| | - J S Tregoning
- Imperial College London, Department of Infectious Disease, London, United Kingdom
| | - K Le Doare
- St George's University of London, Paediatric Infectious Diseases Research Group, London, United Kingdom.,Makerere University John Hopkins Research Collaboration, Kampala, Uganda.,Pathogen Immunology Group, United Kingdom Health Security Agency, Porton Down, United Kingdom
| |
Collapse
|
11
|
Vandoorn E, Parys A, Chepkwony S, Chiers K, Van Reeth K. Efficacy of the NS1-truncated live attenuated influenza virus vaccine for swine against infection with viruses of major North American and European H3N2 lineages. Vaccine 2022; 40:2723-2732. [PMID: 35367071 DOI: 10.1016/j.vaccine.2022.03.051] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/03/2022] [Accepted: 03/21/2022] [Indexed: 11/16/2022]
Abstract
Control of swine influenza A virus (swIAV) in North America and Europe is complicated because multiple antigenically distinct swIAV strains co-circulate in the field, and no vaccine is available that can provide broad cross-protection against all these swIAVs. In 2017, the first live attenuated influenza vaccine (LAIV) for swine was licensed in the US. The non-structural protein 1 (NS1)-truncated cluster I H3N2 strain A/swine/Texas/4199-2/98 NS1del126 (TX98 LAIV) in this vaccine provides partial cross-protection against heterologous North American cluster II and IV H3N2 swIAV strains. Its efficacy against European or more recent North American H3N2 lineages remains to be investigated. In this study, we evaluated the level of cross-protection against heterologous IAVs representative of the major H3N2 swIAV lineages in Europe and North America. TX98 LAIV prevented both nasal shedding and replication in the lungs of a North American cluster IV H3N2 swIAV for 2/4 pigs, prevented considerable nasal shedding of a North American novel human-like H3N2 swIAV for 2/4 pigs, and reduced replication of a European H3N2 swIAV in the lower respiratory tract to minimal titers for 1/3 pigs. Although TX98 LAIV elicited neutralizing antibodies against the homologous virus in serum and to a lesser extent in nose and lungs, no significant cross-reactive antibody titers against the heterologous swIAVs were detected. Partial cross-protection therefore likely relies on cellular and mucosal immune responses against conserved parts of the swIAV proteins. Since TX98 LAIV can offer partial protection against a broad range of H3N2 swIAVs, it might be a suitable priming vaccine for use in a heterologous prime-boost vaccination strategy.
Collapse
Affiliation(s)
- Elien Vandoorn
- Laboratory of Virology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| | - Anna Parys
- Laboratory of Virology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| | - Sharon Chepkwony
- Laboratory of Virology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| | - Koen Chiers
- Laboratory of Veterinary Pathology, Department of Pathobiology, Pharmacology and Zoological Medicine, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| | - Kristien Van Reeth
- Laboratory of Virology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium.
| |
Collapse
|
12
|
Saso A, Kampmann B, Roetynck S. Vaccine-Induced Cellular Immunity against Bordetella pertussis: Harnessing Lessons from Animal and Human Studies to Improve Design and Testing of Novel Pertussis Vaccines. Vaccines (Basel) 2021; 9:877. [PMID: 34452002 PMCID: PMC8402596 DOI: 10.3390/vaccines9080877] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 07/28/2021] [Indexed: 12/14/2022] Open
Abstract
Pertussis ('whooping cough') is a severe respiratory tract infection that primarily affects young children and unimmunised infants. Despite widespread vaccine coverage, it remains one of the least well-controlled vaccine-preventable diseases, with a recent resurgence even in highly vaccinated populations. Although the exact underlying reasons are still not clear, emerging evidence suggests that a key factor is the replacement of the whole-cell (wP) by the acellular pertussis (aP) vaccine, which is less reactogenic but may induce suboptimal and waning immunity. Differences between vaccines are hypothesised to be cell-mediated, with polarisation of Th1/Th2/Th17 responses determined by the composition of the pertussis vaccine given in infancy. Moreover, aP vaccines elicit strong antibody responses but fail to protect against nasal colonisation and/or transmission, in animal models, thereby potentially leading to inadequate herd immunity. Our review summarises current knowledge on vaccine-induced cellular immune responses, based on mucosal and systemic data collected within experimental animal and human vaccine studies. In addition, we describe key factors that may influence cell-mediated immunity and how antigen-specific responses are measured quantitatively and qualitatively, at both cellular and molecular levels. Finally, we discuss how we can harness this emerging knowledge and novel tools to inform the design and testing of the next generation of improved infant pertussis vaccines.
Collapse
Affiliation(s)
- Anja Saso
- The Vaccine Centre, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London WC1 7HT, UK; (B.K.); (S.R.)
- Vaccines and Immunity Theme, MRC Unit, The Gambia at London School of Hygiene & Tropical Medicine, Banjul P.O. Box 273, The Gambia
| | - Beate Kampmann
- The Vaccine Centre, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London WC1 7HT, UK; (B.K.); (S.R.)
- Vaccines and Immunity Theme, MRC Unit, The Gambia at London School of Hygiene & Tropical Medicine, Banjul P.O. Box 273, The Gambia
| | - Sophie Roetynck
- The Vaccine Centre, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London WC1 7HT, UK; (B.K.); (S.R.)
- Vaccines and Immunity Theme, MRC Unit, The Gambia at London School of Hygiene & Tropical Medicine, Banjul P.O. Box 273, The Gambia
| |
Collapse
|
13
|
Verdijk P, van der Plas JL, van Brummelen EMJ, Jeeninga RE, de Haan CAM, Roestenberg M, Burggraaf J, Kamerling IMC. First-in-human administration of a live-attenuated RSV vaccine lacking the G-protein assessing safety, tolerability, shedding and immunogenicity: a randomized controlled trial. Vaccine 2020; 38:6088-6095. [PMID: 32718816 DOI: 10.1016/j.vaccine.2020.07.029] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 06/17/2020] [Accepted: 07/14/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND Human respiratory syncytial virus (RSV) is a major cause of lower respiratory tract infections in early infancy and in elderly. A pediatric vaccine against RSV would not only prevent morbidity and mortality amongst infants and young children but could also reduce transmission to elderly. The RSVΔG vaccine consists of a live-attenuated RSV that lacks the G attachment protein. RSVΔG is severely impaired in binding to host cells and exhibits reduced infectivity in preclinical studies. Intranasal immunization of cotton rats with RSVΔG vaccine protected against replication of wildtype RSV, without inducing enhanced disease. METHODS We performed a first-in-human trial with primary objective to evaluate safety and shedding of RSVΔG (6.5 log10 CCID50) after intranasal administration. Healthy adults aged between 18 and 50, with RSV neutralizing serum titers below 9.6 log2, received a single dose of either vaccine or placebo (n = 48, ratio 3:1). In addition to safety and tolerability, nasal viral load, and systemic and humoral immune responses were assessed at selected time points until 4 weeks after immunization. RESULTS Intranasal administration of RSVΔG was well tolerated with no findings of clinical concern. No infectious virus was detected in nasal wash samples. Similar to other live-attenuated RSV vaccines, neutralizing antibody response following inoculation was limited in seropositive adults. CONCLUSIONS A single dose of 6.5 log10 CCID50 of RSVΔG was safe and well-tolerated in seropositive healthy adults. RSVΔG was sufficiently attenuated but there were no signs of induction of antibodies. Safety and immunogenicity can now be explored in children and eventually in seronegative infants. Clinical trial register: NTR7173/EudraCT number 2016-002437-30.
Collapse
Affiliation(s)
- Pauline Verdijk
- Institute for Translational Vaccinology (Intravacc), Bilthoven, The Netherlands
| | - Johan L van der Plas
- Centre for Human Drug Research, Leiden, The Netherlands; Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | - Cornelis A M de Haan
- Virology Division, Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Meta Roestenberg
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands; Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jacobus Burggraaf
- Centre for Human Drug Research, Leiden, The Netherlands; Leiden University Medical Center, Leiden, The Netherlands
| | - Ingrid M C Kamerling
- Centre for Human Drug Research, Leiden, The Netherlands; Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
14
|
Turner PJ, Abdulla AF, Cole ME, Javan RR, Gould V, O'Driscoll ME, Southern J, Zambon M, Miller E, Andrews NJ, Höschler K, Tregoning JS. Differences in nasal immunoglobulin A responses to influenza vaccine strains after live attenuated influenza vaccine (LAIV) immunization in children. Clin Exp Immunol 2020; 199:109-118. [PMID: 31670841 PMCID: PMC6954673 DOI: 10.1111/cei.13395] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2019] [Indexed: 11/28/2022] Open
Abstract
Different vaccine strains included in the live attenuated influenza vaccine (LAIV) have variable efficacy. The reasons for this are not clear and may include differences in immunogenicity. We report a Phase IV open-label study on the immunogenicity of a single dose of quadrivalent LAIV (Fluenz™ Tetra) in children during the 2015/16 season, to investigate the antibody responses to different strains. Eligible children were enrolled to receive LAIV; nasal samples were collected before and approximately 4 weeks after immunization. There was a significant increase in nasal immunoglobulin (Ig)A to the H3N2, B/Victoria lineage (B/Brisbane) and B/Yamagata lineage (B/Phuket) components, but not to the H1N1 component. The fold change in nasal IgA response was inversely proportional to the baseline nasal IgA titre for H1N1, H3N2 and B/Brisbane. We investigated possible associations that may explain baseline nasal IgA, including age and prior vaccination status, but found different patterns for different antigens, suggesting that the response is multi-factorial. Overall, we observed differences in immune responses to different viral strains included in the vaccine; the reasons for this require further investigation.
Collapse
Affiliation(s)
- P. J. Turner
- National Heart and Lung InstituteImperial College LondonLondonUK
- Public Health England (Colindale)LondonUK
| | - A. F. Abdulla
- Department of Infectious DiseaseSt Mary's CampusImperial College LondonLondonUK
| | - M. E. Cole
- Department of Infectious DiseaseSt Mary's CampusImperial College LondonLondonUK
| | - R. R. Javan
- Department of Infectious DiseaseSt Mary's CampusImperial College LondonLondonUK
| | - V. Gould
- Department of Infectious DiseaseSt Mary's CampusImperial College LondonLondonUK
| | - M. E. O'Driscoll
- Infectious Diseases EpidemiologySt Mary's CampusImperial College LondonLondonUK
| | | | - M. Zambon
- Public Health England (Colindale)LondonUK
| | - E. Miller
- Public Health England (Colindale)LondonUK
| | | | | | - J. S. Tregoning
- Department of Infectious DiseaseSt Mary's CampusImperial College LondonLondonUK
| |
Collapse
|
15
|
Lindsey BB, Jagne YJ, Armitage EP, Singanayagam A, Sallah HJ, Drammeh S, Senghore E, Mohammed NI, Jeffries D, Höschler K, Tregoning JS, Meijer A, Clarke E, Dong T, Barclay W, Kampmann B, de Silva TI. Effect of a Russian-backbone live-attenuated influenza vaccine with an updated pandemic H1N1 strain on shedding and immunogenicity among children in The Gambia: an open-label, observational, phase 4 study. THE LANCET. RESPIRATORY MEDICINE 2019; 7:665-676. [PMID: 31235405 PMCID: PMC6650545 DOI: 10.1016/s2213-2600(19)30086-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 03/03/2019] [Accepted: 03/04/2019] [Indexed: 01/19/2023]
Abstract
BACKGROUND The efficacy and effectiveness of the pandemic H1N1 (pH1N1) component in live attenuated influenza vaccine (LAIV) is poor. The reasons for this paucity are unclear but could be due to impaired replicative fitness of pH1N1 A/California/07/2009-like (Cal09) strains. We assessed whether an updated pH1N1 strain in the Russian-backbone trivalent LAIV resulted in greater shedding and immunogenicity compared with LAIV with Cal09. METHODS We did an open-label, prospective, observational, phase 4 study in Sukuta, a periurban area in The Gambia. We enrolled children aged 24-59 months who were clinically well. Children received one dose of the WHO prequalified Russian-backbone trivalent LAIV containing either A/17/California/2009/38 (Cal09) or A/17/New York/15/5364 (NY15) based on their year of enrolment. Primary outcomes were the percentage of children with LAIV strain shedding at day 2 and day 7, haemagglutinin inhibition seroconversion, and an increase in influenza haemagglutinin-specific IgA and T-cell responses at day 21 after LAIV. This study is nested within a randomised controlled trial investigating LAIV-microbiome interactions (NCT02972957). FINDINGS Between Feb 8, 2017, and April 12, 2017, 118 children were enrolled and received one dose of the Cal09 LAIV from 2016-17. Between Jan 15, 2018, and March 28, 2018, a separate cohort of 135 children were enrolled and received one dose of the NY15 LAIV from 2017-18, of whom 126 children completed the study. Cal09 showed impaired pH1N1 nasopharyngeal shedding (16 of 118 children [14%, 95% CI 8·0-21·1] with shedding at day 2 after administration of LAIV) compared with H3N2 (54 of 118 [46%, 36·6-55·2]; p<0·0001) and influenza B (95 of 118 [81%, 72·2-87·2]; p<0·0001), along with suboptimal serum antibody (seroconversion in six of 118 [5%, 1·9-10·7]) and T-cell responses (CD4+ interferon γ-positive and/or CD4+ interleukin 2-positive responses in 45 of 111 [41%, 31·3-50·3]). After the switch to NY15, a significant increase in pH1N1 shedding was seen (80 of 126 children [63%, 95% CI 54·4-71·9]; p<0·0001 compared with Cal09), along with improvements in seroconversion (24 of 126 [19%, 13·2-26·8]; p=0·011) and influenza-specific CD4+ T-cell responses (73 of 111 [66%, 60·0-75·6; p=0·00028]). The improvement in pH1N1 seroconversion with NY15 was even greater in children who were seronegative at baseline (24 of 64 children [38%, 95% CI 26·7-49·8] vs six of 79 children with Cal09 [8%, 2·8-15·8]; p<0·0001). Persistent shedding to day 7 was independently associated with both seroconversion (odds ratio 12·69, 95% CI 4·1-43·6; p<0·0001) and CD4+ T-cell responses (odds ratio 7·83, 95% CI 2·99-23·5; p<0·0001) by multivariable logistic regression. INTERPRETATION The pH1N1 component switch that took place between 2016 and 2018 might have overcome the poor efficacy and effectiveness reported with previous LAIV formulations. LAIV effectiveness against pH1N1 should, therefore, improve in upcoming influenza seasons. Our data highlight the importance of assessing replicative fitness, in addition to antigenicity, when selecting annual LAIV components. FUNDING The Wellcome Trust.
Collapse
Affiliation(s)
- Benjamin B Lindsey
- Vaccines and Immunity Theme, Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia; Department of Medicine, Imperial College London, London, UK
| | - Ya Jankey Jagne
- Vaccines and Immunity Theme, Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Edwin P Armitage
- Vaccines and Immunity Theme, Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | | | - Hadijatou J Sallah
- Vaccines and Immunity Theme, Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Sainabou Drammeh
- Vaccines and Immunity Theme, Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Elina Senghore
- Vaccines and Immunity Theme, Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Nuredin I Mohammed
- Vaccines and Immunity Theme, Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - David Jeffries
- Vaccines and Immunity Theme, Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Katja Höschler
- Virus Reference Department, Reference Microbiology Services, Public Health England, London, UK
| | | | - Adam Meijer
- Centre for Infectious Disease Research, Diagnostics and Laboratory Surveillance, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Ed Clarke
- Vaccines and Immunity Theme, Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Tao Dong
- Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, and Chinese Academy of Medical Science-Oxford Institute, Nuffield Department of Medicine, Oxford University, Oxford, UK
| | - Wendy Barclay
- Department of Medicine, Imperial College London, London, UK
| | - Beate Kampmann
- Vaccines and Immunity Theme, Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia; The Vaccine Centre, London School of Hygiene & Tropical Medicine, Faculty of Infectious and Tropical Diseases, London, UK
| | - Thushan I de Silva
- Vaccines and Immunity Theme, Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia; Department of Medicine, Imperial College London, London, UK; The Florey Institute for Host-Pathogen Interactions and Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK.
| |
Collapse
|
16
|
Nelson SA, Sant AJ. Imprinting and Editing of the Human CD4 T Cell Response to Influenza Virus. Front Immunol 2019; 10:932. [PMID: 31134060 PMCID: PMC6514101 DOI: 10.3389/fimmu.2019.00932] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 04/11/2019] [Indexed: 12/22/2022] Open
Abstract
Immunity to influenza is unique among pathogens, in that immune memory is established both via intermittent lung localized infections with highly variable influenza virus strains and by intramuscular vaccinations with inactivated protein-based vaccines. Studies in the past decades have suggested that the B cell responses to influenza infection and vaccination are highly biased by an individual's early history of influenza infection. This reactivity likely reflects both the competitive advantage that memory B cells have in an immune response and the relatively limited diversity of epitopes in influenza hemagglutinin that are recognized by B cells. In contrast, CD4 T cells recognize a wide array of epitopes, with specificities that are heavily influenced by the diversity of influenza antigens available, and a multiplicity of functions that are determined by both priming events and subsequent confrontations with antigens. Here, we consider the events that prime and remodel the influenza-specific CD4 T cell response in humans that have highly diverse immune histories and how the CD4 repertoire may be edited in terms of functional potential and viral epitope specificity. We discuss the consequences that imprinting and remodeling may have on the potential of different human hosts to rapidly respond with protective cellular immunity to infection. Finally, these issues are discussed in the context of future avenues of investigation and vaccine strategies.
Collapse
Affiliation(s)
| | - Andrea J. Sant
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Rochester, NY, United States
| |
Collapse
|
17
|
Farne H, Groves HT, Gill SK, Stokes I, McCulloch S, Karoly E, Trujillo-Torralbo MB, Johnston SL, Mallia P, Tregoning JS. Comparative Metabolomic Sampling of Upper and Lower Airways by Four Different Methods to Identify Biochemicals That May Support Bacterial Growth. Front Cell Infect Microbiol 2018; 8:432. [PMID: 30619778 PMCID: PMC6305596 DOI: 10.3389/fcimb.2018.00432] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 11/30/2018] [Indexed: 12/16/2022] Open
Abstract
Bacteria need nutrients from the host environment to survive, yet we know little about which biochemicals are present in the airways (the metabolome), which of these biochemicals are essential for bacterial growth and how they change with airway disease. The aims of this pilot study were to develop and compare methodologies for sampling the upper and lower airway metabolomes and to identify biochemicals present in the airways that could potentially support bacterial growth. Eight healthy human volunteers were sampled by four methods: two standard approaches - nasal lavage and induced sputum, and two using a novel platform, synthetic adsorptive matrix (SAM) strips—nasosorption and bronchosorption. Collected samples were analyzed by Ultrahigh Performance Liquid Chromatography-Tandem Mass Spectroscopy (UPLC-MS/MS). Five hundred and eighty-one biochemicals were recovered from the airways belonging to a range of metabolomic super-pathways. We observed significant differences between the sampling approaches. Significantly more biochemicals were recovered when SAM strips were used, compared to standard sampling techniques. A range of biochemicals that could support bacterial growth were detected in the different samples. This work demonstrates for the first time that SAM strips are a highly effective method for sampling the airway metabolome. This work will assist further studies to understand how changes in the airway metabolome affect bacterial infection in patients with underlying airway disease.
Collapse
Affiliation(s)
- Hugo Farne
- COPD and Asthma, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Helen T Groves
- Mucosal Infection and Immunity, Section of Virology, Imperial College London, London, United Kingdom
| | - Simren K Gill
- Mucosal Infection and Immunity, Section of Virology, Imperial College London, London, United Kingdom
| | - Isobel Stokes
- School of Veterinary Medicine, Faculty of Health & Medical Sciences, University of Surrey, Guildford, United Kingdom
| | | | | | | | - Sebastian L Johnston
- COPD and Asthma, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Patrick Mallia
- COPD and Asthma, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - John S Tregoning
- Mucosal Infection and Immunity, Section of Virology, Imperial College London, London, United Kingdom
| |
Collapse
|
18
|
Thwaites RS, Jarvis HC, Singh N, Jha A, Pritchard A, Fan H, Tunstall T, Nanan J, Nadel S, Kon OM, Openshaw PJ, Hansel TT. Absorption of Nasal and Bronchial Fluids: Precision Sampling of the Human Respiratory Mucosa and Laboratory Processing of Samples. J Vis Exp 2018. [PMID: 29443104 PMCID: PMC5908664 DOI: 10.3791/56413] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The methods of nasal absorption (NA) and bronchial absorption (BA) use synthetic absorptive matrices (SAM) to absorb the mucosal lining fluid (MLF) of the human respiratory tract. NA is a non-invasive technique which absorbs fluid from the inferior turbinate, and causes minimal discomfort. NA has yielded reproducible results with the ability to frequently repeat sampling of the upper airway. By comparison, alternative methods of sampling the respiratory mucosa, such as nasopharyngeal aspiration (NPA) and conventional swabbing, are more invasive and may result in greater data variability. Other methods have limitations, for instance, biopsies and bronchial procedures are invasive, sputum contains many dead and dying cells and requires liquefaction, exhaled breath condensate (EBC) contains water and saliva, and lavage samples are dilute and variable. BA can be performed through the working channel of a bronchoscope in clinic. Sampling is well tolerated and can be conducted at multiple sites in the airway. BA results in MLF samples being less dilute than bronchoalveolar lavage (BAL) samples. This article demonstrates the techniques of NA and BA, as well as the laboratory processing of the resulting samples, which can be tailored to the desired downstream biomarker being measured. These absorption techniques are useful alternatives to the conventional sampling techniques used in clinical respiratory research.
Collapse
Affiliation(s)
- Ryan S Thwaites
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, St Mary's Hospital
| | - Hannah C Jarvis
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, St Mary's Hospital
| | - Nehmat Singh
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, St Mary's Hospital
| | - Akhilesh Jha
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, St Mary's Hospital
| | | | - Hailing Fan
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, St Mary's Hospital
| | - Tanushree Tunstall
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, St Mary's Hospital
| | - Joan Nanan
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, St Mary's Hospital
| | - Simon Nadel
- St Mary's Hospital, Imperial College Healthcare Trust
| | - Onn Min Kon
- St Mary's Hospital, Imperial College Healthcare Trust
| | - Peter J Openshaw
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, St Mary's Hospital
| | - Trevor T Hansel
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, St Mary's Hospital;
| |
Collapse
|