1
|
Emami S, Westerlund E, Rojas Converso T, Johansson-Lindbom B, Persson JJ. Protection acquired upon intraperitoneal group a Streptococcus immunization is independent of concurrent adaptive immune responses but relies on macrophages and IFN-γ. Virulence 2025; 16:2457957. [PMID: 39921669 PMCID: PMC11810095 DOI: 10.1080/21505594.2025.2457957] [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: 08/09/2024] [Revised: 11/09/2024] [Accepted: 01/20/2025] [Indexed: 02/10/2025] Open
Abstract
Group A Streptococcus (GAS; Streptococcus pyogenes) is an important bacterial pathogen causing over 700 million superficial infections and around 500.000 deaths due to invasive disease or severe post-infection sequelae yearly. In spite of this major impact on society, there is currently no vaccine available against this bacterium. GAS strains can be separated into >250 distinct emm (M)-types, and protective immunity against GAS is believed to in part be dependent on type-specific antibodies. Here, we analyse the nature of protective immunity generated against GAS in a model of intraperitoneal immunization in mice. We demonstrate that multiple immunizations are required for the ability to survive a subsequent lethal challenge, and although significant levels of GAS-specific antibodies are produced, these are redundant for protection. Instead, our data show that the immunization-dependent protection in this model is induced in the absence of B and T cells and is accompanied by the induction of an altered acute cytokine profile upon subsequent infection, noticeable e.g. by the absence of classical pro-inflammatory cytokines and increased IFN-γ production. Further, the ability of immunized mice to survive a lethal infection is dependent on macrophages and the macrophage-activating cytokine IFN-γ. To our knowledge these findings are the first to suggest that GAS may have the ability to induce forms of trained innate immunity. Taken together, the current study proposes a novel role for the innate immune system in response to GAS infections that potentially could be leveraged for future development of effective vaccines.
Collapse
Affiliation(s)
- Shiva Emami
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Elsa Westerlund
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | | | | | - Jenny J Persson
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| |
Collapse
|
2
|
Lehtinen M, van Damme P, Beddows S, Pinto LA, Mariz F, Gray P, Dillner J. Scientific approaches to defining HPV vaccine-induced protective immunity. Int J Cancer 2025; 156:1848-1857. [PMID: 39945620 PMCID: PMC11924311 DOI: 10.1002/ijc.35345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 11/06/2024] [Accepted: 12/16/2024] [Indexed: 03/21/2025]
Abstract
Seventeen years after the licensure of prophylactic human papillomavirus (HPV) L1 virus-like-particle vaccines, a defined antibody level that correlates with vaccine-induced protection against HPV infections and associated neoplasia is missing. In contrast, correlates of protection have been defined for many viral vaccines, including for the hepatitis B virus (HBV) vaccine. This review includes lessons learned from vaccination against HBV and the use of an established protective HBV surface antigen antibody level: 10 mIU/mL, an overview of HPV infection-induced and HPV vaccine-induced antibody responses, successful efforts to establish international standardization of serological reagents and associated tools, and 15-year vigilance of HPV vaccine-induced antibody levels in a vaccination cohort against breakthrough infections. This report identifies progress but also gaps on the journey toward the definition of a HPV vaccine-induced correlate of protection.
Collapse
Affiliation(s)
- Matti Lehtinen
- Department of Vaccines, Institute for Health and Welfare, Helsinki, Finland
- Center for Cervical Cancer Elimination, Department of Clinical Science, Intervention and Technology, Karolinska Institute, Stockholm, Sweden
| | - Pierre van Damme
- Centre for the Evaluation of Vaccination@Vaccinopolis, Universiteit Antwerp, Antwerp, Belgium
| | - Simon Beddows
- Virus Reference Department, Public Health Microbiology Division, UK Health Security Agency, London, UK
| | - Ligia A Pinto
- HPV Serology Laboratory, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Filipe Mariz
- Division of Infections and Cancer, Deutsches Krebsforschungszentrum, Heidelberg, Germany
| | - Penelope Gray
- Center for Cervical Cancer Elimination, Department of Clinical Science, Intervention and Technology, Karolinska Institute, Stockholm, Sweden
| | - Joakim Dillner
- Center for Cervical Cancer Elimination, Department of Clinical Science, Intervention and Technology, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
3
|
Borrow R, Tomasi Cont L, Toneatto D, Bambini S, Bobde S, Sohn WY, Biolchi A, Masignani V, Beernink PT, Lattanzi M. Methods to evaluate the performance of a multicomponent meningococcal serogroup B vaccine. mSphere 2025; 10:e0089824. [PMID: 40197090 PMCID: PMC12039234 DOI: 10.1128/msphere.00898-24] [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] [Indexed: 04/09/2025] Open
Abstract
Meningococcal serogroup B (MenB) vaccine licensure was based on the assessment of vaccine-induced immune responses by human serum bactericidal antibody (hSBA) assay against a small number of antigen-specific strains complemented by strain coverage predictions. However, the evaluation of vaccine strain coverage is challenging because of genotypic and phenotypic diversity in surface-exposed MenB strain antigens. This narrative review considers the principal methods applied to assess the performance of a multicomponent MenB vaccine at different stages of its development. Traditional hSBA assay against a limited panel of strains is useful at all stages, while predicted strain coverage methods, such as the meningococcal antigen typing system, are used independent of clinical trials. A new method, the endogenous complement hSBA assay, has been developed to evaluate a vaccine's ability to induce a bactericidal immune response in clinical trials, in conditions that approximate real-world settings through the use of each vaccinee's serum as a source of complement and by testing against a panel of 110 epidemiologically representative MenB strains. Each assay, therefore, has a different scope during the vaccine's development and all complement each other, enabling comprehensive evaluation of the performance of multicomponent MenB vaccines, in advance of real-world evidence of vaccine effectiveness and vaccine impact.
Collapse
Affiliation(s)
- Ray Borrow
- Meningococcal Reference Unit, UK Health Security Agency, Manchester Royal Infirmary, Manchester, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
4
|
Nielsen-Saines K, Kalbasi-Romero T, Duarte ACM, Almeida da Silva S, Adachi K, Damasceno L, Kerin T, Fuller T, Deville JG, Moreira ME, Vasconcelos Z, Zin A, Shin-Sim M, Barbosa de Lima SM, Brasil P. Development of Maternal Antibodies Post ZIKV in Pregnancy is Associated with Lower Risk of Microcephaly and Structural Brain Abnormalities in Exposed Infants. J Infect Dis 2025:jiaf146. [PMID: 40257773 DOI: 10.1093/infdis/jiaf146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Indexed: 04/22/2025] Open
Abstract
BACKGROUND We investigated the association between maternal neutralizing antibodies (nAb) to Zika virus (ZIKV) in pregnancy and neonatal outcomes. METHODS In pregnant participants with confirmed ZIKV infection, we determined trimester of infection, collected sera longitudinally, and measured nAbs via plaque reduction. In neonates, adverse outcomes included microcephaly (MC), structural brain abnormalities (SBA), hearing, and eye abnormalities. Associations between trimester of infection, nAbs, and neonatal outcomes were analyzed with Cox regression. RESULTS In total, 137 ZIKV-positive pregnant participants had neutralization assays performed during pregnancy and postdelivery. Infection rates were 29% in the first, 50% in the second, and 21% in the third trimester. Mean ZIKV nAb titer >2 weeks postinfection was 64 258 (SD 213 288). Ten percent of 90% plaque reduction neutralization assays (PRNT90) titers were <500, 10% 500-1000, 73% > 1000, and 7% did not have serologic follow-up; 15%. of infants had adverse findings. Protective factors against MC in 88 mothers with nAb titers available during pregnancy included infection later in gestation (adjusted hazard ratio [aHR], 0.06; P = .036) and adequate nAb titers (aHR, 0.17; P = .014). No SBA was associated with later infection in pregnancy (aHR, 0.16; P = .017) and adequate nAb titers (aHR, 0.34; P = .012). Adjusting for trimester, higher maternal nAb titers were associated with lower risk of MC and SBA. Seven of 137 participants (5.1%) had positive serum ZIKV polymerase chain reaction (PCR) results beyond 14 days (range, 35-269 days). Participants with ZIKV PCR positivity >60 days (n = 2) had infants with MC/SBA. CONCLUSIONS MC and SBA were less frequent in infants of mothers with higher ZIKV nAb titers during pregnancy.
Collapse
Affiliation(s)
- Karin Nielsen-Saines
- Division of Pediatric Infectious Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Tahmineh Kalbasi-Romero
- Division of General Internal Medicine and Health Services Research, University of California Los Angeles, Los Angeles, California, USA
| | | | | | - Kristina Adachi
- Division of Pediatric Infectious Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Luana Damasceno
- Acute Febrile Illness Laboratory, Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Tara Kerin
- Division of Pediatric Infectious Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Trevon Fuller
- Acute Febrile Illness Laboratory, Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- Institute of the Environment and Sustainability, University of California Los Angeles, Los Angeles, California, USA
| | - Jaime G Deville
- Division of Pediatric Infectious Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Maria Elisabeth Moreira
- Fernandes Figueira National Institute of Women's, Children's, and Adolescents' Health, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Zilton Vasconcelos
- Fernandes Figueira National Institute of Women's, Children's, and Adolescents' Health, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Andrea Zin
- Fernandes Figueira National Institute of Women's, Children's, and Adolescents' Health, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Myung Shin-Sim
- Division of General Internal Medicine and Health Services Research, University of California Los Angeles, Los Angeles, California, USA
| | | | - Patricia Brasil
- Acute Febrile Illness Laboratory, Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| |
Collapse
|
5
|
Rao VN, Coelho CH. Public antibodies: convergent signatures in human humoral immunity against pathogens. mBio 2025:e0224724. [PMID: 40237455 DOI: 10.1128/mbio.02247-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2025] Open
Abstract
The human humoral immune system has evolved to recognize a vast array of pathogenic threats. This ability is primarily driven by the immense diversity of antibodies generated by gene rearrangement during B cell development. However, different people often produce strikingly similar antibodies when exposed to the same antigen-known as public antibodies. Public antibodies not only reflect the immune system's ability to consistently select for optimal B cells but can also serve as signatures of the humoral responses triggered by infection and vaccination. In this Minireview, we examine and compare public antibody identification methods, including the identification criteria used based on V(D)J gene usage and similarity in the complementarity-determining region three sequences, and explore the molecular features of public antibodies elicited against common pathogens, including viruses, protozoa, and bacteria. Finally, we discuss the evolutionary significance and potential applications of public antibodies in informing the design of germline-targeting vaccines, predicting escape mutations in emerging viruses, and providing insights into the process of affinity maturation. The ongoing discovery of public antibodies in response to emerging pathogens holds the potential to improve pandemic preparedness, accelerate vaccine design efforts, and deepen our understanding of human B cell biology.
Collapse
Affiliation(s)
- Vishal N Rao
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, USA
- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Camila H Coelho
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, USA
- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, USA
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| |
Collapse
|
6
|
Brady C, Tipton T, Carnell O, Longet S, Gooch K, Hall Y, Salguero J, Tomic A, Carroll M. A systems biology approach to define SARS-CoV-2 correlates of protection. NPJ Vaccines 2025; 10:69. [PMID: 40229322 PMCID: PMC11997207 DOI: 10.1038/s41541-025-01103-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 03/10/2025] [Indexed: 04/16/2025] Open
Abstract
Correlates of protection (CoPs) for SARS-CoV-2 have yet to be sufficiently defined. This study uses the machine learning platform, SIMON, to accurately predict the immunological parameters that reduced clinical pathology or viral load following SARS-CoV-2 challenge in a cohort of 90 non-human primates. We found that anti-SARS-CoV-2 spike antibody and neutralising antibody titres were the best predictors of clinical protection and low viral load in the lung. Since antibodies to SARS-CoV-2 spike showed the greatest association with clinical protection and reduced viral load, we next used SIMON to investigate the immunological features that predict high antibody titres. It was found that a pre-immunisation response to seasonal beta-HCoVs and a high frequency of peripheral intermediate and non-classical monocytes predicted low SARS-CoV-2 spike IgG titres. In contrast, an elevated T cell response as measured by IFNγ ELISpot predicted high IgG titres. Additional predictors of clinical protection and low SARS-CoV-2 burden included a high abundance of peripheral T cells. In contrast, increased numbers of intermediate monocytes predicted clinical pathology and high viral burden in the throat. We also conclude that an immunisation strategy that minimises pathology post-challenge did not necessarily mediate viral control. This would be an important finding to take forward into the development of future vaccines aimed at limiting the transmission of SARS-CoV-2. These results contribute to SARS-CoV-2 CoP definition and shed light on the factors influencing the success of SARS-CoV-2 vaccination.
Collapse
Affiliation(s)
- Caolann Brady
- Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom.
- Pandemic Sciences Institute, University of Oxford, Oxford, United Kingdom.
| | - Tom Tipton
- Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Pandemic Sciences Institute, University of Oxford, Oxford, United Kingdom
| | - Oliver Carnell
- UK Health Security Agency; Porton Down, Salisbury, United Kingdom
| | - Stephanie Longet
- Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Pandemic Sciences Institute, University of Oxford, Oxford, United Kingdom
- International Center for Infectiology Research (CIRI), Team GIMAP, Claude Bernard Lyon 1 University, Saint-Etienne, France
| | - Karen Gooch
- UK Health Security Agency; Porton Down, Salisbury, United Kingdom
| | - Yper Hall
- UK Health Security Agency; Porton Down, Salisbury, United Kingdom
| | - Javier Salguero
- UK Health Security Agency; Porton Down, Salisbury, United Kingdom
| | - Adriana Tomic
- National Emerging Infectious Diseases Laboratories, Boston, MA, USA
- Department of Virology, Immunology & Microbiology, Boston University Medical School, Boston, MA, USA
- Biomedical Engineering, Boston University, College of Engineering, Boston, MA, USA
| | - Miles Carroll
- Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom.
- Pandemic Sciences Institute, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
7
|
Madden PJ, Marina-Zárate E, Rodrigues KA, Steichen JM, Shil M, Ni K, Michaels KK, Maiorino L, Upadhyay AA, Saha S, Pradhan A, Kalyuzhiny O, Liguori A, Lopez PG, Phung I, Flynn C, Zhou A, Melo MB, Lemnios A, Phelps N, Georgeson E, Alavi N, Kubitz M, Lu D, Eskandarzadeh S, Metz A, Rodriguez OL, Shields K, Schultze S, Smith ML, Healy BS, Lim D, Lewis VR, Ben-Akiva E, Pinney W, Gregory J, Xiao S, Carnathan DG, Pai Kasturi S, Watson CT, Bosinger SE, Silvestri G, Schief WR, Irvine DJ, Crotty S. Diverse priming outcomes under conditions of very rare precursor B cells. Immunity 2025; 58:997-1014.e11. [PMID: 40168992 DOI: 10.1016/j.immuni.2025.03.003] [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: 10/11/2024] [Revised: 02/18/2025] [Accepted: 03/04/2025] [Indexed: 04/03/2025]
Abstract
Rare naive B cells have special pathogen-recognition features that enable outsized contributions to protective immunity but infrequently participate in immune responses. We investigatee how germline-targeting vaccine delivery and adjuvant selection affect priming of exceptionally rare BG18-like HIV broadly neutralizing antibody-precursor B cells (<1-in-50 million) in non-human primates. Only escalating dose (ED) priming immunization using the saponin adjuvant SMNP elicited detectable BG18-like cells in germinal centers (GCs) compared with other conditions. All groups had strong GC responses, but only ED+SMNP and bolus+SMNP induced BG18-like memory B cells in >50% of animals. One group had vaccine-specific GC responses equivalent to ED+SMNP but scarce BG18-like B cells. Following homologous boosting, BG18-like memory B cells were present in a bolus priming group but with lower somatic hypermutation and affinities than ED+SMNP. This outcome inversely associated with post-prime antibody titers, suggesting antibody feedback significantly influences rare precursor B cell responses. Thus, antigen and inflammatory stimuli extensively impact priming and affinity maturation of rare B cells.
Collapse
Affiliation(s)
- Patrick J Madden
- La Jolla Institute for Immunology, La Jolla, CA, USA; Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA, USA
| | - Ester Marina-Zárate
- La Jolla Institute for Immunology, La Jolla, CA, USA; Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA, USA
| | - Kristen A Rodrigues
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jon M Steichen
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA, USA; Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Monolina Shil
- La Jolla Institute for Immunology, La Jolla, CA, USA; Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA, USA
| | - Kaiyuan Ni
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Laura Maiorino
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Amit A Upadhyay
- Emory National Primate Research Center and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA; Department of Pathology and Laboratory Medicine, Emory School of Medicine, Atlanta, GA, USA
| | - Swati Saha
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, USA
| | - Arpan Pradhan
- Emory National Primate Research Center and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Oleksandr Kalyuzhiny
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA, USA; Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA; IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, USA
| | - Alessia Liguori
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA, USA; Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA; IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, USA
| | - Paul G Lopez
- La Jolla Institute for Immunology, La Jolla, CA, USA; Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA, USA
| | - Ivy Phung
- La Jolla Institute for Immunology, La Jolla, CA, USA; Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA, USA
| | - Claudia Flynn
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, USA
| | - Amelia Zhou
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, USA
| | - Mariane B Melo
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Ashley Lemnios
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Nicole Phelps
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA, USA; Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Erik Georgeson
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA, USA; Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Nushin Alavi
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA, USA; Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Michael Kubitz
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA, USA; Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Danny Lu
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA, USA; Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Saman Eskandarzadeh
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA, USA; Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Amanda Metz
- Emory National Primate Research Center and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA; Department of Pathology and Laboratory Medicine, Emory School of Medicine, Atlanta, GA, USA
| | - Oscar L Rodriguez
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, USA
| | - Kaitlyn Shields
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, USA
| | - Steven Schultze
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, USA
| | - Melissa L Smith
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, USA
| | - Brandon S Healy
- Emory National Primate Research Center and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Deuk Lim
- Emory National Primate Research Center and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Vanessa R Lewis
- Emory National Primate Research Center and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Elana Ben-Akiva
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - William Pinney
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Justin Gregory
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Shuhao Xiao
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Diane G Carnathan
- Emory National Primate Research Center and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Sudhir Pai Kasturi
- Emory National Primate Research Center and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Corey T Watson
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, USA
| | - Steven E Bosinger
- Emory National Primate Research Center and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA; Department of Pathology and Laboratory Medicine, Emory School of Medicine, Atlanta, GA, USA
| | - Guido Silvestri
- Emory National Primate Research Center and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
| | - William R Schief
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA, USA; Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA; IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, USA; Moderna, Inc., Cambridge, MA, USA
| | - Darrell J Irvine
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA, USA; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA.
| | - Shane Crotty
- La Jolla Institute for Immunology, La Jolla, CA, USA; Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA, USA; Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
8
|
Guillaume SM, Foster WS, San Martín Molina I, Watson EM, Innocentin S, Kennedy GM, Denton AE, Linterman MA. Lung B cells in ectopic germinal centers undergo affinity maturation. Proc Natl Acad Sci U S A 2025; 122:e2416855122. [PMID: 40168127 PMCID: PMC12002176 DOI: 10.1073/pnas.2416855122] [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: 08/20/2024] [Accepted: 02/18/2025] [Indexed: 04/03/2025] Open
Abstract
The lungs are constantly exposed to the external environment and a myriad of antigenic challenges within the air. Chronic exposure to allergens and other airborne antigens can result in the formation of lymphocyte aggregates in the lung, which can harbor ectopic germinal centers (GCs). After allergen exposure, GCs that form in the lung are much smaller and less densely packed with B cells than lymph node GCs. Despite this, ectopic lung GCs support somatic hypermutation and affinity-based maturation as in lymph node GCs, and export memory B cells (MBCs) directly into the lung tissue. This demonstrates that the lung can locally diversify B cell responses and supports the generation of tissue MBC populations in situ.
Collapse
Affiliation(s)
| | - William S. Foster
- Immunology Program, Babraham Institute, CambridgeCB22 3AT, United Kingdom
| | - Isabel San Martín Molina
- Immunology Program, Babraham Institute, CambridgeCB22 3AT, United Kingdom
- Babraham Imaging Core, Babraham Institute, CambridgeCB22 3AT, United Kingdom
| | - Emily M. Watson
- Immunology Program, Babraham Institute, CambridgeCB22 3AT, United Kingdom
| | - Silvia Innocentin
- Immunology Program, Babraham Institute, CambridgeCB22 3AT, United Kingdom
| | - Grant M. Kennedy
- Immunology Program, Babraham Institute, CambridgeCB22 3AT, United Kingdom
- Department of Physics, University of Warwick, CoventryCV4 7AL, United Kingdom
| | - Alice E. Denton
- Department of Immunology and Inflammation, Imperial College London, LondonW12 0NN, United Kingdom
| | | |
Collapse
|
9
|
Riolo G, Biagini V, Guerrini N, Roscia G, Antonelli R, Giglioli G, Stincarelli MA, Piu P, Bonifazi C, De Grazia S, Pizzo M, Lovreglio P, Stufano A, Trombetta CM, Manenti A, Montomoli E, Dapporto F. Design and validation of a semi-quantitative microneutralization assay for human Metapneumovirus A1 and B1 subtypes. Sci Rep 2025; 15:11614. [PMID: 40185841 PMCID: PMC11971440 DOI: 10.1038/s41598-025-96567-6] [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: 01/14/2025] [Accepted: 03/28/2025] [Indexed: 04/07/2025] Open
Abstract
Since 2001, human Metapneumovirus has been a significant cause of human respiratory disease worldwide, and no vaccine or preventive treatment is currently available. The ELISA-based live virus microneutralization assay is a method to detect neutralizing antibodies against a target pathogen. The aim of this study was to demonstrate the suitability of this approach to quantifying neutralizing antibodies against A1 and B1 virus subtypes in human serum samples. To standardize and validate this microneutralization assay, we carried out analytical procedures according to the International Council of Harmonization guidelines; these procedures are described in detail. In addition, we compared the validated method with the indirect ELISA, and confirmed that the ELISA-based microneutralization assay provides reliable, accurate and reproducible results. The use of this high-throughput method for large-scale serological studies could effectively support the evaluation of the immunogenicity of new vaccines, thereby improving therapeutical strategies against human Metapneumovirus.
Collapse
Affiliation(s)
| | | | - Noemi Guerrini
- VisMederi S.R.L., Siena, Italy
- Department of Life Sciences, University of Siena, Siena, Italy
| | | | | | | | | | | | | | - Simona De Grazia
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties "G. D'Alessandro", University of Palermo, Palermo, Italy
| | - Mariangela Pizzo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties "G. D'Alessandro", University of Palermo, Palermo, Italy
| | - Piero Lovreglio
- Interdisciplinary Department of Medicine, Section of Occupational Medicine, University of Bari, Bari, Italy
| | - Angela Stufano
- Interdisciplinary Department of Medicine, Section of Occupational Medicine, University of Bari, Bari, Italy
| | - Claudia Maria Trombetta
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
- VaepiX, Joint Research Laboratory, University of Siena, Siena, Italy
| | | | - Emanuele Montomoli
- VisMederi S.R.L., Siena, Italy
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
- VaepiX, Joint Research Laboratory, University of Siena, Siena, Italy
| | | |
Collapse
|
10
|
Wagoner ZW, Yates TB, Hernandez-Davies JE, Sureshchandra S, Joloya EM, Jain A, de Assis R, Kastenschmidt JM, Sorn AM, Mitul MT, Tamburini I, Ahuja G, Zhong Q, Trask D, Seldin M, Davies DH, Wagar LE. Systems immunology analysis of human immune organoids identifies host-specific correlates of protection to different influenza vaccines. Cell Stem Cell 2025; 32:529-546.e6. [PMID: 39986275 PMCID: PMC11974613 DOI: 10.1016/j.stem.2025.01.014] [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: 04/24/2024] [Revised: 11/27/2024] [Accepted: 01/27/2025] [Indexed: 02/24/2025]
Abstract
Vaccines are an essential tool to significantly reduce pathogen-related morbidity and mortality. However, our ability to rationally design vaccines and identify correlates of protection remains limited. Here, we employed an immune organoid approach to capture human adaptive immune response diversity to influenza vaccines and systematically identify host and antigen features linked to vaccine response variability. Our investigation identified established and unique immune signatures correlated with neutralizing antibody responses across seven different influenza vaccines and antigens. Unexpectedly, heightened ex vivo tissue frequencies of T helper (Th)1 cells emerged as both a predictor and a correlate of neutralizing antibody responses to inactivated influenza vaccines (IIVs). Secondary analysis of human public data confirmed that elevated Th1 signatures are associated with antibody responses following in vivo vaccination. These findings demonstrate the utility of human in vitro models for identifying in vivo correlates of protection and establish a role for Th1 functions in influenza vaccination.
Collapse
Affiliation(s)
- Zachary W Wagoner
- Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, USA; Institute for Immunology, University of California, Irvine, Irvine, CA, USA; Center for Virus Research, University of California, Irvine, Irvine, CA, USA; Vaccine R&D Center, University of California, Irvine, Irvine, CA, USA
| | - Timothy B Yates
- Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, USA; Institute for Immunology, University of California, Irvine, Irvine, CA, USA; Center for Virus Research, University of California, Irvine, Irvine, CA, USA; Vaccine R&D Center, University of California, Irvine, Irvine, CA, USA
| | - Jenny E Hernandez-Davies
- Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, USA; Institute for Immunology, University of California, Irvine, Irvine, CA, USA; Center for Virus Research, University of California, Irvine, Irvine, CA, USA; Vaccine R&D Center, University of California, Irvine, Irvine, CA, USA
| | - Suhas Sureshchandra
- Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, USA; Institute for Immunology, University of California, Irvine, Irvine, CA, USA; Center for Virus Research, University of California, Irvine, Irvine, CA, USA; Vaccine R&D Center, University of California, Irvine, Irvine, CA, USA
| | - Erika M Joloya
- Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, USA; Institute for Immunology, University of California, Irvine, Irvine, CA, USA; Center for Virus Research, University of California, Irvine, Irvine, CA, USA; Vaccine R&D Center, University of California, Irvine, Irvine, CA, USA
| | - Aarti Jain
- Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, USA; Institute for Immunology, University of California, Irvine, Irvine, CA, USA; Vaccine R&D Center, University of California, Irvine, Irvine, CA, USA
| | - Rafael de Assis
- Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, USA; Institute for Immunology, University of California, Irvine, Irvine, CA, USA; Vaccine R&D Center, University of California, Irvine, Irvine, CA, USA
| | - Jenna M Kastenschmidt
- Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, USA; Institute for Immunology, University of California, Irvine, Irvine, CA, USA; Center for Virus Research, University of California, Irvine, Irvine, CA, USA; Vaccine R&D Center, University of California, Irvine, Irvine, CA, USA
| | - Andrew M Sorn
- Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, USA; Institute for Immunology, University of California, Irvine, Irvine, CA, USA; Center for Virus Research, University of California, Irvine, Irvine, CA, USA; Vaccine R&D Center, University of California, Irvine, Irvine, CA, USA
| | - Mahina Tabassum Mitul
- Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, USA; Institute for Immunology, University of California, Irvine, Irvine, CA, USA; Center for Virus Research, University of California, Irvine, Irvine, CA, USA; Vaccine R&D Center, University of California, Irvine, Irvine, CA, USA
| | - Ian Tamburini
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, USA; Center for Epigenetics and Metabolism, University of California, Irvine, Irvine, CA, USA
| | - Gurpreet Ahuja
- Department of Pediatric Otolaryngology, Children's Hospital of Orange County, Orange, CA, USA; Department of Otolaryngology-Head and Neck Surgery, University of California, Irvine, Orange, CA, USA
| | - Qiu Zhong
- Department of Pediatric Otolaryngology, Children's Hospital of Orange County, Orange, CA, USA; Department of Otolaryngology-Head and Neck Surgery, University of California, Irvine, Orange, CA, USA
| | - Douglas Trask
- Department of Otolaryngology-Head and Neck Surgery, University of California, Irvine, Orange, CA, USA
| | - Marcus Seldin
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, USA; Center for Epigenetics and Metabolism, University of California, Irvine, Irvine, CA, USA
| | - D Huw Davies
- Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, USA; Institute for Immunology, University of California, Irvine, Irvine, CA, USA; Center for Virus Research, University of California, Irvine, Irvine, CA, USA; Vaccine R&D Center, University of California, Irvine, Irvine, CA, USA
| | - Lisa E Wagar
- Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, USA; Institute for Immunology, University of California, Irvine, Irvine, CA, USA; Center for Virus Research, University of California, Irvine, Irvine, CA, USA; Vaccine R&D Center, University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
11
|
Gilbert SC. Priorities for rapid and cost-effective vaccines to improve outbreak responses. PLoS Biol 2025; 23:e3003114. [PMID: 40193351 PMCID: PMC11975085 DOI: 10.1371/journal.pbio.3003114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2025] Open
Abstract
The use of vaccine platform technologies greatly reduces the time and money required to develop a novel vaccine against an infectious disease, but much work is still needed if vaccines are to protect the world from the next pandemic.
Collapse
Affiliation(s)
- 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
|
12
|
Nassuuna J, Sterk J, Walusimbi B, Natukunda A, Nkangi R, Amongin R, Zirimenya L, Webb EL, Elliott AM, Nkurunungi G. Helminth driven gut inflammation and microbial translocation associate with altered vaccine responses in rural Uganda. NPJ Vaccines 2025; 10:56. [PMID: 40140378 PMCID: PMC11947158 DOI: 10.1038/s41541-025-01116-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 03/17/2025] [Indexed: 03/28/2025] Open
Abstract
Vaccine responses are sometimes impaired in rural, low-income settings. Helminth-associated gut barrier dysfunction and microbial translocation (MT) may be implicated. We used samples from a trial of praziquantel treatment-effects on vaccine responses in Schistosoma mansoni (Sm)-endemic Ugandan islands, measuring intestinal fatty acid-binding protein 2 (I-FABP2), lipopolysaccharide-binding protein, anti-endotoxin core antibodies (EndoCab), soluble CD14 (sCD14) in plasma, and faecal lipocalin-2, occult blood (FOB), and calprotectin (fCAL), and evaluating their associations with baseline helminth infection, praziquantel treatment, and responses to BCG, yellow fever, typhoid, HPV, and tetanus-diphtheria vaccines. Sm associated positively with fCAL and FOB, hookworm with I-FABP2, and any helminth with EndoCab IgM, fCAL and FOB. Sm associated inversely with sCD14. Praziquantel treatment reduced all marker concentrations, significantly fCAL and FOB, implying that Sm-associated gut inflammation and MT is reversible. Associations of assessed markers with vaccine-specific responses were predominantly inverse. Interventions to improve gut barrier function may enhance vaccine responsiveness.
Collapse
Affiliation(s)
- Jacent Nassuuna
- Immunomodulation and Vaccines Focus Area, Vaccine Research Theme, MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda
| | | | - Bridgious Walusimbi
- Immunomodulation and Vaccines Focus Area, Vaccine Research Theme, MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK
| | - Agnes Natukunda
- Immunomodulation and Vaccines Focus Area, Vaccine Research Theme, MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda
- International Statistics and Epidemiology Group, Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK
| | - Ronald Nkangi
- Immunomodulation and Vaccines Focus Area, Vaccine Research Theme, MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda
- Leiden University Center for Infectious Diseases (LU-CID), Leiden University Medical Center, Leiden, The Netherlands
| | - Rebecca Amongin
- Immunomodulation and Vaccines Focus Area, Vaccine Research Theme, MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda
| | - Ludoviko Zirimenya
- Immunomodulation and Vaccines Focus Area, Vaccine Research Theme, MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda
- Department of Clinical Research, London School of Hygiene and Tropical Medicine, London, UK
| | - Emily L Webb
- Immunomodulation and Vaccines Focus Area, Vaccine Research Theme, MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda
- International Statistics and Epidemiology Group, Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK
| | - Alison M Elliott
- Immunomodulation and Vaccines Focus Area, Vaccine Research Theme, MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda
- Department of Clinical Research, London School of Hygiene and Tropical Medicine, London, UK
| | - Gyaviira Nkurunungi
- Immunomodulation and Vaccines Focus Area, Vaccine Research Theme, MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda.
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK.
| |
Collapse
|
13
|
Olivo PD, Ghatak S, Rasch R. A Multiplex Bead Serology Panel For Vaccine-Preventable Diseases Using Dried Blood Spots. Risk Manag Healthc Policy 2025; 18:923-932. [PMID: 40129703 PMCID: PMC11932034 DOI: 10.2147/rmhp.s506025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 03/14/2025] [Indexed: 03/26/2025] Open
Abstract
Background Vaccines are effective tools to improve public health. The effectiveness of vaccines is, however, dependent on the overall level of protection in a population. Antibodies to vaccine-related antigens are good biomarkers of protection and serosurveillance can help target vaccination programs. An integrated approach to perform serosurveillance on multiple vaccine-preventable diseases (VPDs) has been advocated and would be facilitated by a standardized multiplex immunoassay. In this report, we describe the evaluation of the performance of a multi-lyte bead-based serology panel for 12 VPDs which uses a dried blood spot sample from a finger prick (ImmunoProfile Antibody Test System). Methods Verification/validation studies were performed at a CLIA-certified clinical laboratory (BioAgilytix Labs, Boston, MA) on blood collected from dried blood spot (DBS) card samples from adults and children. In addition, proof-of-principle pilot serosurveillance studies were performed to demonstrate the potential of this test to identify protection gaps in adult and pediatric populations. Results This study demonstrates that the ImmunoProfile Antibody Test System has the requisite analytical performance to be a reliable tool for determining levels of protection to VPDs. The pilot serosurveillance studies demonstrate that this test reveals gaps in protection comparable to what has been shown using immunoassays for individual antibodies using serum samples. Conclusion Serological survey data obtained with the validated ImmunoProfile Antibody Test System could provide a wealth of information on levels of protection and could unearth vaccination gaps that may not have been anticipated.
Collapse
Affiliation(s)
- Paul D Olivo
- Department of Microbiology and Microbial Pathogenesis, Washington University Medical School, St. Louis, MO, USA
| | | | - Randolph Rasch
- College of Nursing, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
14
|
Fant P, Laurent S, Desert P, Combadière B, Palazzi X, Choudhary S, Gervais F, Broudic K, Rossi R, Gauthier BE. Proceedings of the 2023 Annual Scientific Meeting of the French Society of Toxicologic Pathology (SFPT) on Preclinical Development and Therapeutic Applications of mRNA-Based Technologies. Toxicol Pathol 2025:1926233251326089. [PMID: 40110665 DOI: 10.1177/01926233251326089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
The 2023 annual scientific meeting of the French Society of Toxicologic Pathology (Société Française de Pathologie Toxicologique, SFPT), entitled "mRNA-based technologies: preclinical development and therapeutic applications," was held in Lyon (France) on May 25 to 26, 2023. The aim of the meeting was to discuss the biology, immunology, and preclinical development of messenger RNA (mRNA)-based vaccines and therapeutics, including immuno-oncology and rare diseases, as well as the regulatory aspect of the COVID-19 vaccines and an overview of the principles and applications of in situ hybridization techniques. This article presents the summary of five lectures along with selected figures, tables, and key literature references on this topic.
Collapse
Affiliation(s)
- Pierluigi Fant
- Charles River Laboratories Safety Assessment, Saint Germain-Nuelles, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Streif S, Baeumner AJ. Advances in Surrogate Neutralization Tests for High-Throughput Screening and the Point-of-Care. Anal Chem 2025; 97:5407-5423. [PMID: 40035475 PMCID: PMC11923957 DOI: 10.1021/acs.analchem.5c00666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Affiliation(s)
- Simon Streif
- Institute of Analytical Chemistry, Chemo- and Biosensors, University of Regensburg, Universitaetsstrasse 31, 93053 Regensburg, Germany
| | - Antje J Baeumner
- Institute of Analytical Chemistry, Chemo- and Biosensors, University of Regensburg, Universitaetsstrasse 31, 93053 Regensburg, Germany
| |
Collapse
|
16
|
Sun N, Su Z, Zheng X. Research progress of mosquito-borne virus mRNA vaccines. Mol Ther Methods Clin Dev 2025; 33:101398. [PMID: 39834558 PMCID: PMC11743085 DOI: 10.1016/j.omtm.2024.101398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
In recent years, mRNA vaccines have emerged as a leading technology for preventing infectious diseases due to their rapid development and high immunogenicity. These vaccines encode viral antigens, which are translated into antigenic proteins within host cells, inducing both humoral and cellular immune responses. This review systematically examines the progress in mRNA vaccine research for major mosquito-borne viruses, including dengue virus, Zika virus, Japanese encephalitis virus, Chikungunya virus, yellow fever virus, Rift Valley fever virus, and Venezuelan equine encephalitis virus. Enhancements in mRNA vaccine design, such as improvements to the 5' cap structure, 5'UTR, open reading frame, 3'UTR, and polyadenylation tail, have significantly increased mRNA stability and translation efficiency. Additionally, the use of lipid nanoparticles and polymer nanoparticles has greatly improved the delivery efficiency of mRNA vaccines. Currently, mRNA vaccines against mosquito-borne viruses are under development and clinical trials, showing promising protective effects. Future research should continue to optimize vaccine design and delivery systems to achieve broad-spectrum and long-lasting protection against various mosquito-borne virus infections.
Collapse
Affiliation(s)
- Ningze Sun
- Beijing Institute of Tropical Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory for Research on Prevention and Treatment of Tropical Diseases, Beijing, China
| | - Zhiwei Su
- Beijing Institute of Tropical Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory for Research on Prevention and Treatment of Tropical Diseases, Beijing, China
| | - Xiaoyan Zheng
- Beijing Institute of Tropical Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory for Research on Prevention and Treatment of Tropical Diseases, Beijing, China
| |
Collapse
|
17
|
Nakayama T, Hamada S, Kawamura A, Sogawa Y, Sakakibara S, Nakatsu T, Kimata M, Oe K. Non-inferiority and vaccine titer-confirmation studies of MMR vaccine (JVC-001; measles AIK-C, mumps RIT4385, and rubella Takahashi strains) in healthy 1-year-old Japanese children. Vaccine 2025; 49:126698. [PMID: 39920023 DOI: 10.1016/j.vaccine.2024.126698] [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] [Accepted: 12/18/2024] [Indexed: 02/09/2025]
Abstract
OBJECTIVES JVC-001 is a new live attenuated measles-mumps-rubella vaccine (measles AIK-C, mumps RIT4385, and rubella Takahashi strains). Two phase 3 studies were conducted, one to verify the non-inferior immunogenicity of JVC-001 versus the approved mumps and measles-rubella vaccines (J301 study) and another to compare the immunogenicity and safety of different titers (J302 study). METHODS Both studies were multicenter, randomized, observer-blinded, phase 3 studies. J301 compared the immunogenicity elicited with a single dose of JVC-001 or control vaccines (measles-rubella vaccine + mumps vaccine [Hoshino strain]). J302 was a titer-confirmation study of a single dose of a low- or high-titer formulation of JVC-001. Both studies enrolled healthy Japanese children (aged 1 year) and had a primary efficacy endpoint of seropositive rate on Day 43. RESULTS Overall, 861 participants completed J301 (JVC-001, n = 429; control, n = 432) and 100 participants completed J302 (low-titer, n = 48; high-titer, n = 52). For measles and rubella virus antibody titer, non-inferiority of JVC-001 was demonstrated: seropositive rates were ≥ 99.5 %. For mumps virus genotype D antibody titer, seropositive rates were 80.6 % (95 % confidence interval 76.5 % to 84.4 %) with JVC-001 and 88.1 % (84.6 % to 91.0 %) with control vaccination. Thus, non-inferiority for mumps virus genotype D antibody titer was not confirmed. Seropositive rates were similar in the low- and high-titer groups. There were no events leading to discontinuation, or cases of aseptic meningitis in either study. CONCLUSIONS Although the non-inferiority of JVC-001 to currently approved vaccines was not demonstrated for the mumps component, clinical significance and consistent efficacy were indicated. Vaccine titer did not affect immunogenicity. JVC-001 is expected to have a lower risk of aseptic meningitis to currently approved vaccines and raised no new safety signals emerged.
Collapse
Affiliation(s)
- Tetsuo Nakayama
- Laboratory of Viral Infection, Ömura Satoshi Memorial Institute, Kitasato University, 5-9-1 Shirokane, Minato-Ku, Tokyo 108-8641, Japan.
| | - Shunsuke Hamada
- Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-Ku, Tokyo 140-8710, Japan.
| | - Asuka Kawamura
- Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-Ku, Tokyo 140-8710, Japan.
| | - Yoshitaka Sogawa
- Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-Ku, Tokyo 140-8710, Japan.
| | - Sachiko Sakakibara
- Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-Ku, Tokyo 140-8710, Japan.
| | - Takafumi Nakatsu
- Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-Ku, Tokyo 140-8710, Japan.
| | - Motoshi Kimata
- Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-Ku, Tokyo 140-8710, Japan.
| | - Keiji Oe
- Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-Ku, Tokyo 140-8710, Japan.
| |
Collapse
|
18
|
Kobayashi T, Fujiwara S, Ide A, Toya T, Shingai N, Shimizu H, Najima Y, Kobayashi T, Doki N, Jo A. Impact of DTaP-IPV and DTaP Vaccination Among Adult Allogeneic Hematopoietic Stem Cell Transplant Recipients: A Prospective Observational Study. Vaccines (Basel) 2025; 13:275. [PMID: 40266146 PMCID: PMC11946051 DOI: 10.3390/vaccines13030275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 02/27/2025] [Accepted: 03/02/2025] [Indexed: 04/24/2025] Open
Abstract
Background/Objectives: Hematopoietic stem cell transplantation (HSCT) can potentially cure hematological malignancies; however, post-transplant patients have a high risk of infection owing to their immunocompromised status. Vaccination against pathogens, such as diphtheria, tetanus, pertussis, and polio, is essential post-transplantation, but neither the long-term efficacy of vaccines nor the optimal vaccination schedule has been fully established. Methods: In this prospective observational study, we assessed the short- and long-term immunogenicity of three doses of the diphtheria, tetanus, acellular pertussis, and inactivated poliovirus (DTaP-IPV) vaccines or DTaP vaccines in 29 adult allogeneic HSCT (allo-HSCT) recipients, with antibody levels measured at baseline, 1-3 months post-vaccination, and 1-year after vaccine completion. Results: At baseline, a substantial proportion of patients lacked protective antibody levels for the targeted pathogens. However, within 1-3 months post-vaccination, seropositivity rates significantly increased, reaching 78-100% for diphtheria, tetanus, pertussis, and poliovirus. Despite this, antibody levels significantly declined 1-year post-vaccination, especially for pertussis, with only 58-65% of patients maintaining protective levels. In contrast, 85-96% of patients retained protective levels for diphtheria, tetanus, and poliovirus, although antibody values also decreased. Compared to human leukocyte antigen (HLA)-mismatched cases, HLA-matched cases showed significantly higher antibody levels for diphtheria, pertussis, and poliovirus types 1 and 3. Conclusions: This study demonstrates the short-term effectiveness of DTaP-IPV and DTaP vaccines in adult allo-HSCT patients but emphasizes the challenge of maintaining long-term immunity. Given the difficulties in sustaining long-term vaccine efficacy in allo-HSCT recipients, particularly in HLA-mismatched cases, re-evaluating the current vaccination schedule may be necessary to maintain protection.
Collapse
Affiliation(s)
- Taiichiro Kobayashi
- Vaccine Clinic, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-ku, Tokyo 113-8677, Japan; (S.F.); (A.I.); (A.J.)
- Department of Infectious Diseases, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-ku, Tokyo 113-8677, Japan
| | - Sho Fujiwara
- Vaccine Clinic, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-ku, Tokyo 113-8677, Japan; (S.F.); (A.I.); (A.J.)
- Department of Infectious Diseases, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-ku, Tokyo 113-8677, Japan
| | - Ayako Ide
- Vaccine Clinic, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-ku, Tokyo 113-8677, Japan; (S.F.); (A.I.); (A.J.)
- Department of Pediatrics, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-ku, Tokyo 113-8677, Japan
| | - Takashi Toya
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-ku, Tokyo 113-8677, Japan; (T.T.); (N.S.); (H.S.); (Y.N.); (T.K.); (N.D.)
| | - Naoki Shingai
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-ku, Tokyo 113-8677, Japan; (T.T.); (N.S.); (H.S.); (Y.N.); (T.K.); (N.D.)
| | - Hiroaki Shimizu
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-ku, Tokyo 113-8677, Japan; (T.T.); (N.S.); (H.S.); (Y.N.); (T.K.); (N.D.)
| | - Yuho Najima
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-ku, Tokyo 113-8677, Japan; (T.T.); (N.S.); (H.S.); (Y.N.); (T.K.); (N.D.)
| | - Takeshi Kobayashi
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-ku, Tokyo 113-8677, Japan; (T.T.); (N.S.); (H.S.); (Y.N.); (T.K.); (N.D.)
| | - Noriko Doki
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-ku, Tokyo 113-8677, Japan; (T.T.); (N.S.); (H.S.); (Y.N.); (T.K.); (N.D.)
| | - Aoi Jo
- Vaccine Clinic, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-ku, Tokyo 113-8677, Japan; (S.F.); (A.I.); (A.J.)
- Department of Pediatrics, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-ku, Tokyo 113-8677, Japan
| |
Collapse
|
19
|
Deming ME, Toapanta FR, Pasetti M, Golding H, Khurana S, Hamouda T, Fattom A, Liang Y, Tennant SM, McGilvray MF, Bernal PJ, Oshinsky JJ, Datta S, Booth JP, Coughlan L, Neuzil KM, Costley CD, Kotloff KL, Sztein MB, Ortiz JR. An intranasal adjuvanted, recombinant influenza A/H5 vaccine candidate induces broad priming against diverse influenza A/H5N1 virus clades in a phase I randomized trial in healthy adults. RESEARCH SQUARE 2025:rs.3.rs-6059149. [PMID: 40092447 PMCID: PMC11908355 DOI: 10.21203/rs.3.rs-6059149/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
We conducted a randomized, controlled phase I trial (NCT05397119) of a novel adjuvanted recombinant influenza A/H5 (A/Indonesia/05/2005, clade 2.1) hemagglutinin vaccine, administered intranasally in two doses 28 days apart at three antigen levels. Control groups received unadjuvanted recombinant H5 or formulation buffer placebo. Six months later, participants received a heterologous unadjuvanted inactivated influenza A/H5N1 (A/Vietnam/1203/2004, clade 1) vaccine intramuscularly. All vaccines were safe and well tolerated. After the primary intranasal series, serum hemagglutination inhibition and microneutralization responses were minimal. Increases in mucosal and serum IgG/IgA, serum surface plasmon resonance antibody binding, memory B cell and CD4 T cell activity, and antibody-dependent cell-mediated cytotoxicity were observed only in recipients primed intranasally with adjuvanted H5 vaccine. Following the inactivated H5N1 boost, robust responses across all immune assays, as well as microneutralization responses against diverse H5N1 clades (including currently circulating clade 2.3.4.4b), occurred in adjuvanted vaccine recipients, demonstrating successful priming and broad responses.
Collapse
Affiliation(s)
- Meagan E Deming
- Center for Vaccine Development, University of Maryland School of Medicine
| | | | - Marcela Pasetti
- Center for Vaccine Development, University of Maryland School of Medicine
| | - Hana Golding
- Center for Biologics Evaluation and Research (CBER), Food and Drug Administration
| | - Surender Khurana
- Center for Biologics Evaluation and Research (CBER), Food and Drug Administration
| | | | | | - Yuanyuan Liang
- Center for Vaccine Development, University of Maryland School of Medicine
| | - Sharon M Tennant
- Center for Vaccine Development, University of Maryland School of Medicine
| | - Megan F McGilvray
- Center for Vaccine Development, University of Maryland School of Medicine
| | - Paula J Bernal
- Center for Vaccine Development, University of Maryland School of Medicine
| | | | - Shrimati Datta
- Center for Vaccine Development, University of Maryland School of Medicine
| | | | - Lynda Coughlan
- Center for Vaccine Development, University of Maryland School of Medicine
| | - Kathleen M Neuzil
- Center for Vaccine Development, University of Maryland School of Medicine
| | | | - Karen L Kotloff
- Center for Vaccine Development, University of Maryland School of Medicine
| | - Marcelo B Sztein
- Center for Vaccine Development, University of Maryland School of Medicine
| | - Justin R Ortiz
- Center for Vaccine Development, University of Maryland School of Medicine
| |
Collapse
|
20
|
Arkell P, Tanesi MY, Martins N, Gomes N, Oakley T, Solano V, David M, Amaral S, Sheridan SL, Yip TW, Draper AD, Sarmento N, Soares da Silva E, Alves L, Nicholson S, Stambos V, Pedrina K, Freitas C, de Neri Machado F, Gusmão CA, da Costa Barreto I, Fancourt NS, Macartney K, Yan J, Francis JR. A population-representative serosurvey estimating vaccine-induced immunity against measles, rubella, hepatitis B and severe acute respiratory syndrome coronavirus 2 in Timor-Leste. THE LANCET REGIONAL HEALTH. SOUTHEAST ASIA 2025; 34:100525. [PMID: 40084157 PMCID: PMC11905827 DOI: 10.1016/j.lansea.2024.100525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 10/25/2024] [Accepted: 12/23/2024] [Indexed: 03/16/2025]
Abstract
Background Serosurveillance can be used to assess population immunity to vaccine-preventable diseases (VPDs). This study aimed to determine seroprevalence of four VPDs across Timor-Leste and identify immunity gaps. Methods A population-representative three-stage cluster random sample of census-enumerated households were visited between October 2021 and February 2023. Occupants aged above one year were tested for measles immunoglobulin G (IgG), rubella IgG, hepatitis B surface antibody (HBsAb), hepatitis B core antibody (HBcAb) and severe acute respiratory syndrome 2 (SARS-CoV-2) IgG, using serological assays with a priori determined cut-offs. Sample- and response-weighted mixed effects logistic regression models were used to estimate seroprevalence in relevant age-strata. Findings Of 2613 eligible households, 1908 (73.0%) participated. Of 8427 occupants, 4750 (56.4%) participated. Measles IgG seroprevalence was low among children, particularly those aged 10-14 (33.2%, 95% confidence interval (CI) 27.8-38.6%). Rubella IgG seroprevalence was high in all ages (93.2%, 95% CI 92.2-94.2%). SARS-CoV-2 IgG seroprevalence was high, including in young children not eligible for vaccination (74.0%, 95% CI 70.4-77.6%). HBsAb seroprevalence was lowest among adolescents aged 15-19 (12.1%, 95% CI 6.8-17.5%) but higher among younger children, who also had low HBcAb seroprevalence. Interpretation The pattern of measles immunity is consistent with low virus transmission and suboptimal childhood vaccine uptake. These data have informed supplementary immunisation activities. High rubella IgG seroprevalence suggests recent or ongoing virus transmission and a need for congenital rubella syndrome surveillance. Hepatitis B data provide evidence of recent improvements in vaccine-induced immunity and protection. This study demonstrates how serosurveillance can directly influence national vaccine strategies. Funding This study was funded by the Department for Foreign Affairs and Trade, Australian Government (Complex Grant Agreement Number 75889).
Collapse
Affiliation(s)
- Paul Arkell
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Dili, Timor-Leste
| | - Maria Y. Tanesi
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Dili, Timor-Leste
| | - Nelson Martins
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Dili, Timor-Leste
| | - Nelia Gomes
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Dili, Timor-Leste
| | - Tessa Oakley
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Dili, Timor-Leste
| | - Vanessa Solano
- Research Institute for the Environment and Livelihoods, Charles Darwin University, Darwin, Australia
| | - Michael David
- Daffodil Centre, The University of Sydney, a Joint Venture with Cancer Council New South Wales, Sydney, NSW, Australia
- School of Medicine & Dentistry, Griffith University, Gold Coast, QLD, Australia
| | - Salvador Amaral
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Dili, Timor-Leste
| | - Sarah L. Sheridan
- National Centre for Immunisation Research and Surveillance (NCIRS), Westmead, NSW, Australia
| | - Teem-Wing Yip
- National Centre for Immunisation Research and Surveillance (NCIRS), Westmead, NSW, Australia
- College of Medicine and Public Health, Flinders University, Australia
| | - Anthony D.K. Draper
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Dili, Timor-Leste
- Northern Territory Centre for Disease Control, Darwin, Australia
- National Centre for Epidemiology and Population Health, Australian National University, Canberra, Australia
| | - Nevio Sarmento
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Dili, Timor-Leste
| | | | - Lucsendar Alves
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Dili, Timor-Leste
| | - Suellen Nicholson
- Victorian Infectious Diseases Reference Laboratory, Royal Melbourne Hospital, Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Vicki Stambos
- Victorian Infectious Diseases Reference Laboratory, Royal Melbourne Hospital, Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Kate Pedrina
- Victorian Infectious Diseases Reference Laboratory, Royal Melbourne Hospital, Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | | | | | | | - Ismael da Costa Barreto
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Dili, Timor-Leste
| | - Nicholas S.S. Fancourt
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Dili, Timor-Leste
| | - Kristine Macartney
- National Centre for Immunisation Research and Surveillance (NCIRS), Westmead, NSW, Australia
- Faculty of Medicine and Health, University of Sydney, Australia
| | - Jennifer Yan
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Dili, Timor-Leste
| | - Joshua R. Francis
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Dili, Timor-Leste
| |
Collapse
|
21
|
Richardson S, Medhavi FNU, Tanner T, Lundy S, Omosun Y, Igietseme JU, Eko FO. Role of route of delivery on Chlamydia abortus vaccine-induced immune responses and genital tract immunity in mice. Microbes Infect 2025; 27:105463. [PMID: 39645188 DOI: 10.1016/j.micinf.2024.105463] [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: 05/12/2024] [Revised: 12/03/2024] [Accepted: 12/04/2024] [Indexed: 12/09/2024]
Abstract
We investigated if the efficacy of a Chlamydia abortus (Cab) subunit vaccine is influenced by route of administration. Thus, female CBA/J mice were immunized either by mucosal or systemic routes with Vibrio cholerae ghost (VCG)-based vaccine expressing T and B cell epitopes of Cab polymorphic membrane protein (Pmp) 18D, termed rVCG-Pmp18.3. Vaccine evaluation revealed that all routes of vaccine delivery induced a Th1-type antibody response after a prime boost or three-dose immunization regimen. Also, the intranasal and rectal mucosal and intramuscular systemic routes induced cross-reactive neutralizing antibodies against homologous and heterologous Cab strains. Irrespective of the route of immunization, the vaccine elicited a Th1-type cytokine response (IFN-γ/IL-4 >1) in immunized mice. Analysis of reduction in genital Cab burden as an index of protection showed that immunization induced substantial degrees of protection against infection, irrespective of route of delivery with the intranasal and rectal mucosal routes showing superior levels of protection 12 days postchallenge. Furthermore, there was correlation between the humoral and cellular immune response and protection was associated with the Cab-specific serum IgG antibody avidity and IFN-γ. Thus, while route of administration impacts vaccine efficacy, the rVCG-Pmp18.3-induced protective immunity against Cab respiratory infection can be accomplished by both mucosal and systemic immunization.
Collapse
Affiliation(s)
- Shakyra Richardson
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, USA
| | - F N U Medhavi
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Tayhlor Tanner
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Stephanie Lundy
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Yusuf Omosun
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Joseph U Igietseme
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, USA; National Center for Emerging Zoonotic and Infectious Diseases, Center for Disease Control and Prevention (CDC), Atlanta, GA, USA
| | - Francis O Eko
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
22
|
Goggin KP, Sun E, Yun E, Kamel M, Perez MA, Hsiao HM, DiMaggio LS, Liverman R, Anderson EJ, Shane AL, Garro R, George RP, Rostad CA. Serologic Responses to COVID-19 Vaccination in Pediatric Kidney Transplant Recipients. Transplant Direct 2025; 11:e1756. [PMID: 39936133 PMCID: PMC11809963 DOI: 10.1097/txd.0000000000001756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 10/31/2024] [Indexed: 02/13/2025] Open
Abstract
Background There are limited data describing the immune responses to COVID-19 vaccination in pediatric kidney transplant recipients, and expanding upon this information could help inform vaccination strategies in this unique population. Methods We performed a prospective, observational, single-center cohort study using remnant blood samples of pediatric kidney transplant recipients from routine clinic visits to examine longitudinal serological responses after COVID-19 vaccination. We enrolled 61 pediatric kidney transplant recipients who had at least 1 sample available for analysis. Sera or plasma were analyzed for ancestral SARS-CoV-2 and Omicron (B.1.1.529; BA.1) spike IgG and nucleocapsid IgG using a Meso Scale Discovery platform. Results One month after a 3-dose COVID-19 vaccination series, the IgG geometric mean titer to the SARS-CoV-2 ancestral spike was 684 binding antibody units/mL (95% confidence interval, 269-1739), but titers waned by 4-6 mo. A fourth dose of the COVID-19 vaccine boosted IgG geometric mean titer to 1606 binding antibody units/mL (95% confidence interval, 868-2972), and titers persisted through 6 mo. IgG titers against Omicron (B.1.1.529; BA.1) were overall lower than ancestral SARS-CoV-2. They were higher in participants with prior infection and were not significantly impacted by receipt of belatacept. Conclusions Additional doses of the COVID-19 vaccine bolstered durable serologic responses in pediatric kidney transplant recipients, and this study broadens our understanding of immune responses to COVID-19 vaccinations in this population.
Collapse
Affiliation(s)
- Kathryn P. Goggin
- Department of Pediatrics, Children’s Healthcare of Atlanta, Atlanta, GA
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
| | - Elizabeth Sun
- Division of Nephrology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
| | - Emily Yun
- Division of Nephrology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
| | - Margret Kamel
- Division of Nephrology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
| | - Maria A. Perez
- Department of Pediatrics, Children’s Healthcare of Atlanta, Atlanta, GA
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
| | - Hui-Mien Hsiao
- Department of Pediatrics, Children’s Healthcare of Atlanta, Atlanta, GA
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
| | - Langdon S. DiMaggio
- Department of Pediatrics, Children’s Healthcare of Atlanta, Atlanta, GA
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
| | - Rochelle Liverman
- Department of Pediatrics, Children’s Healthcare of Atlanta, Atlanta, GA
| | - Evan J. Anderson
- Department of Pediatrics, Children’s Healthcare of Atlanta, Atlanta, GA
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA
| | - Andi L. Shane
- Department of Pediatrics, Children’s Healthcare of Atlanta, Atlanta, GA
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
| | - Rouba Garro
- Department of Pediatrics, Children’s Healthcare of Atlanta, Atlanta, GA
- Division of Nephrology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
| | - Roshan P. George
- Department of Pediatrics, Children’s Healthcare of Atlanta, Atlanta, GA
- Division of Nephrology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
| | - Christina A. Rostad
- Department of Pediatrics, Children’s Healthcare of Atlanta, Atlanta, GA
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
| |
Collapse
|
23
|
Rubio-Casillas A, Redwan EM, Uversky VN. More antibodies are not always better: Fc effector functions play a critical role in SARS-CoV-2 infection and protection. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2025; 213:413-447. [PMID: 40246351 DOI: 10.1016/bs.pmbts.2025.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
Traditional vaccinology has primarily focused on neutralizing antibody titers as the main correlate of vaccine efficacy, often overlooking the multifaceted roles of antibody Fc effector functions in orchestrating protective immune responses. Fc-mediated immune responses play a pivotal role in immune modulation and pathogen clearance. Emerging evidence from natural infections and vaccine studies highlights the critical contribution of Fc effector functions in determining the quality and durability of immunity. This work explores the limitations of current vaccine evaluation paradigms that prioritize neutralization over Fc effector mechanisms. It also describes findings from a study showing an unexpected role for SARS-CoV-2 anti-spike antibodies: both convalescent plasma and patient-derived monoclonal antibodies (mAbs) lead to maximum phagocytic capacity by monocytes at low concentrations, whereas at higher concentrations the phagocytic capacity was reduced. Given that the severity of COVID-19 disease and antibody titers are strongly positively correlated, this work challenges the paradigm that high antibodies offer better protection against severe disease. It is proposed that humoral and cellular responses elicited by vaccination should never be higher than those produced by natural infection. By integrating antibody Fc effector functions into vaccine development, a paradigm shift is proposed that emphasizes synergic antibody responses. Such an approach could transform vaccine efficacy assessment, enhance protection against dangerous pathogens, and drive innovation in vaccine design.
Collapse
Affiliation(s)
- Alberto Rubio-Casillas
- Autlan Regional Hospital, Jalisco Health Services, Autlan, Jalisco, Mexico; Biology Laboratory, Autlan Regional Preparatory School, University of Guadalajara, Autlan, Jalisco, Mexico.
| | - Elrashdy M Redwan
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia; Therapeutic and Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City for Scientific Research and Technology Applications, New Borg El-Arab, Alexandria, Egypt
| | - Vladimir N Uversky
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States; USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| |
Collapse
|
24
|
Odio CD, Yek C, Hasund CM, Man S, Ly P, Nhek S, Chea S, Lon C, Voirin C, Huy R, Leang R, Huch C, Lamirande EW, Whitehead SS, Oliveira F, Manning JE, Katzelnick LC. Immunity to Non-Dengue Flaviviruses Impacts Dengue Virus Immunoglobulin G Enzyme-Linked Immunosorbent Assay Specificity in Cambodia. J Infect Dis 2025; 231:e337-e344. [PMID: 39297691 PMCID: PMC11841641 DOI: 10.1093/infdis/jiae422] [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: 05/24/2024] [Indexed: 09/25/2024] Open
Abstract
BACKGROUND Seroprevalence studies are the standard for disease surveillance, and serology determined eligibility for the first dengue vaccine. Expanding flavivirus co-circulation and vaccination complicate testing. We evaluate the accuracy of a common dengue virus serological assay, examine immunity to non-dengue flaviviruses as a contributor to decreased performance, and assess whether alternative cut points may improve assay performance. METHODS Children (n = 770) aged 2-9 years in Kampong Speu, Cambodia were enrolled in a prospective longitudinal study, and PanBio indirect dengue virus immunoglobulin G (IgG) enzyme-linked immunosorbent assay (ELISA) was performed. Plaque reduction neutralization tests (PRNTs) using dengue viruses were performed on a subset to assess the accuracy of the IgG ELISA, and PRNTs with Zika, Japanese encephalitis, and West Nile viruses evaluated immunity to non-dengue flaviviruses. Receiver operating curve analysis identified an alternative cut point to improve IgG ELISA accuracy. RESULTS The dengue IgG ELISA had a lower specificity than previously reported (58% vs 93%-100%). Of those with false-positive IgG results, 46% had detectable neutralizing antibodies against other flaviviruses including 14% against West Nile virus. A higher IgG cut point improved the test accuracy in this population. CONCLUSIONS Physicians and public health authorities should be alert for West Nile in Cambodia. Immunity to non-dengue flaviviruses can impact dengue surveillance. CLINICAL TRIALS REGISTRATION NCT03534245.
Collapse
Affiliation(s)
- Camila D Odio
- Viral Epidemiology and Immunity Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Christina Yek
- International Center of Excellence in Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Phnom Penh, Cambodia
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Chloe M Hasund
- Viral Epidemiology and Immunity Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Somnang Man
- International Center of Excellence in Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Phnom Penh, Cambodia
| | - Piseth Ly
- International Center of Excellence in Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Phnom Penh, Cambodia
| | - Sreynik Nhek
- International Center of Excellence in Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Phnom Penh, Cambodia
| | - Sophana Chea
- International Center of Excellence in Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Phnom Penh, Cambodia
| | - Chanthap Lon
- International Center of Excellence in Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Phnom Penh, Cambodia
| | - Charlie Voirin
- Viral Epidemiology and Immunity Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Rekol Huy
- National Center for Parasitology, Entomology, and Malaria Control, Ministry of Health, Phnom Penh, Cambodia
| | - Rithea Leang
- National Center for Parasitology, Entomology, and Malaria Control, Ministry of Health, Phnom Penh, Cambodia
| | - Chea Huch
- National Center for Parasitology, Entomology, and Malaria Control, Ministry of Health, Phnom Penh, Cambodia
| | - Elaine W Lamirande
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Stephen S Whitehead
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Fabiano Oliveira
- International Center of Excellence in Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Phnom Penh, Cambodia
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Jessica E Manning
- International Center of Excellence in Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Phnom Penh, Cambodia
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Leah C Katzelnick
- Viral Epidemiology and Immunity Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
25
|
Ma KSK, Shen CH, Chiang MH, Blumenthal KG, Chen ST. Severe Cutaneous Adverse Reactions Following Vaccination: A Systematic Review and Meta-Analysis. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2025:S2213-2198(25)00169-2. [PMID: 39978545 DOI: 10.1016/j.jaip.2025.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 12/10/2024] [Accepted: 02/04/2025] [Indexed: 02/22/2025]
Abstract
BACKGROUND An increasing number of vaccine-related severe cutaneous adverse reactions (SCARs) have been reported in the literature. OBJECTIVES To provide comprehensive information on the clinical spectrum of vaccine-associated SCARs. METHODS This was a systematic review and meta-analysis of case reports, case series, cohort studies, case-control studies, and post-market surveillance data on SCARs following vaccination. Data were extracted on demographic information, vaccine types, past medical history, medication use, and SCAR types, manifestations, management, and prognosis. The SCARs examined included acute generalized exanthematous pustulosis, drug reaction with eosinophilia and systemic symptoms, Stevens-Johnson syndrome/toxic epidermal necrolysis (SJS/TEN), and generalized bullous fixed drug eruptions. RESULTS A total of 255 cases of SCARs following vaccination were identified. Of these, 231 (91%) were classified as SJS/TEN. The pooled incidence of SCARs following vaccination was 1.676 per million (95% confidence interval = 0.136-20.668; I2 = 97%). The most frequently implicated vaccines were H1N1 vaccines (n = 52), coronavirus disease 2019 (COVID-19) vaccines (n = 38, of which 23 [61%] were mRNA vaccines), and influenza vaccines (n = 33). Acute generalized exanthematous pustulosis and drug reaction with eosinophilia and systemic symptoms were most frequently reported with COVID-19 vaccines, particularly mRNA vaccines (57.1% [4 of 7] and 83.3% [5 of 6], respectively) and viral vector vaccines (28.6% [2 of 7] and 16.7% [1 of 6], respectively). No SCARs were reported for protein-based COVID-19 vaccines. Six cases of fatal SJS/TEN were identified, including 2 cases associated with the COVID-19 vaccine. CONCLUSIONS SCARs following vaccination are extremely rare, with SJS/TEN comprising the majority of reported cases. The most frequently suspected vaccine culprits include H1N1, influenza, varicella, and COVID-19 vaccines, particularly mRNA vaccines. However, concurrent medication use may confound the attribution of SCARs to vaccines.
Collapse
Affiliation(s)
- Kevin Sheng-Kai Ma
- Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, Mass
| | - Chin-Hsuan Shen
- Department of Dermatology, Chang Gung Memorial Hospital Linkou Medical Center, Taoyuan, Taiwan
| | - Meng-Han Chiang
- Clinical Metabolomics Core Laboratory, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Kimberly G Blumenthal
- Division of Rheumatology, Allergy, and Immunology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Mass.
| | - Steven T Chen
- Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, Mass.
| |
Collapse
|
26
|
Keay S, Alberts F, O’Connor AM, Friendship R, O’Sullivan T, Poljak Z. The case for development of a core outcome set (COS) and supplemental reporting guidelines for influenza vaccine challenge trial research in swine. Front Vet Sci 2025; 12:1465926. [PMID: 40007748 PMCID: PMC11851948 DOI: 10.3389/fvets.2025.1465926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 01/27/2025] [Indexed: 02/27/2025] Open
Abstract
Previously, we systematically reviewed more than 20 years of influenza vaccine challenge trial research in pigs to answer the question, "does vaccinating sows protect offspring?" Overall, most studies were well designed but clinical heterogeneity made between-study comparisons challenging. Studies varied by samples, outcomes, and assays selected for measurement. Additionally, data essential for inclusion of findings in meta-analyses were often insufficiently reported and as a result, summary effect measures were either not derived or were not meaningful. Clinical heterogeneity and reporting issues complicate and limit what can be learned cumulatively from research and both represent two types of avoidable research waste. Here, we illustrate each concern using data collected tangentially during the systematic review and propose two corrective strategies, both of which have broad applicability across veterinary intervention research; (i) develop a Core Outcome Set (COS) to reduce unnecessary clinical heterogeneity in future research and (ii) encourage funders and journal editors to require submitted research protocols and manuscripts adhere to established reporting guidelines. As a reporting corollary, we developed a supplemental checklist specific to influenza vaccine challenge trial research in swine and propose that it is completed by researchers and included with all study protocol and manuscript submissions. The checklist serves two purposes: as a reminder of details essential to report for inclusion of findings in meta-analyses and sub-group meta-analyses (e.g., antigenic or genomic descriptions of influenza vaccine and challenge viruses), and as an aid to help synthesis researchers fully characterize and comprehensively include studies in reviews.
Collapse
Affiliation(s)
- Sheila Keay
- Department of Population Medicine, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Famke Alberts
- Department of Population Medicine, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Annette M. O’Connor
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, MI, United States
| | - Robert Friendship
- Department of Population Medicine, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Terri O’Sullivan
- Department of Population Medicine, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Zvonimir Poljak
- Department of Population Medicine, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
27
|
Eskenazi B, Rauch S, Elsiwi B, Bornman R, Obida M, Brewer A, Ward BJ, Chevrier J. Undernutrition and antibody response to measles, tetanus and Haemophilus Influenzae type b (Hib) vaccination in pre-school south African children: The VHEMBE birth cohort study. Vaccine 2025; 46:126564. [PMID: 39665976 PMCID: PMC11750586 DOI: 10.1016/j.vaccine.2024.126564] [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: 07/05/2024] [Revised: 11/14/2024] [Accepted: 11/23/2024] [Indexed: 12/13/2024]
Abstract
BACKGROUND Under-vaccination is undoubtedly driving recent worldwide measles outbreaks, but undernutrition may also be playing a role in low- and middle-income countries. Studies have shown reduced immune response to vaccines in undernourished children but few have followed children beyond infancy, when they are more likely to be exposed to infectious diseases. METHODS In the Venda Health Examination of Mothers, Babies and the Environment (VHEMBE) South African birth cohort study, we examined the relationship between undernutrition, as measured by stunting and other growth measures, and vaccine-specific serum antibody level to three different vaccine types: measles, tetanus and Haemophilus influenzae type b (Hib). We included 621 fully-vaccinated children with anthropometric measurements at ages 1, 2, and 3.5 years and antibody levels at 3.5 and 5 years. RESULTS At 5 years of age, 90.4% of fully-vaccinated children were protected against measles, 66.7% against tetanus, and 56.1% against Hib. Children who were stunted or had any indicator of diminished growth at 3.5 years averaged a 24.1% (95% CI = -44.2, 0.6) or a 27.2% (95% CI = -45.1, -1.3) lower antibody titer for measles, respectively, relative to those with normal growth. In addition, girls, but not boys, with any indicator of diminished growth at 3.5 years averaged a 36.8% (-59.3, -7.0) lower antibody titer for tetanus. We found no association between undernutrition and Hib antibody titers. CONCLUSIONS Early life undernutrition may be associated with lower induction or persistence of antibody responses to certain vaccines. Addressing child undernutrition may improve vaccine efficacy and reduce the burden of vaccine-preventable diseases.
Collapse
Affiliation(s)
- Brenda Eskenazi
- Center for Environmental Research and Community Health (CERCH), School of Public Health, University of California at Berkeley, Berkeley, CA 94720, USA.
| | - Stephen Rauch
- Center for Environmental Research and Community Health (CERCH), School of Public Health, University of California at Berkeley, Berkeley, CA 94720, USA
| | - Basant Elsiwi
- Department of Epidemiology, Biostatistics and Occupational Health, School of Population and Global Health, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada
| | - Riana Bornman
- School of Health Systems and Public Health, University of Pretoria, Pretoria, Gauteng, South Africa
| | - Muvhulawa Obida
- School of Health Systems and Public Health, University of Pretoria, Pretoria, Gauteng, South Africa
| | - Angela Brewer
- Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada; Division of Experimental Medicine, Department of Medicine, Montreal, Quebec, Canada
| | - Brian J Ward
- Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada; Division of Experimental Medicine, Department of Medicine, Montreal, Quebec, Canada
| | - Jonathan Chevrier
- Department of Epidemiology, Biostatistics and Occupational Health, School of Population and Global Health, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
28
|
Mathew S, Alex D, Demosthenes JP, Rose W, Chacko A, Kompithra RZ, Ramalingam VV, Prakash JAJ, Mathai J, Fletcher GJ, Abraham P, Verghese VP, Kannangai R. Heterogeneity and Hierarchy of Immune Response to Primary Immunization in HIV-Infected Children on HAART and the Impact of an Additional Dose of Vaccine. Indian J Pediatr 2025; 92:181-184. [PMID: 38801497 DOI: 10.1007/s12098-024-05148-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 04/25/2024] [Indexed: 05/29/2024]
Abstract
The nature of vaccine response inferiority is not well studied in children living with HIV (CLHIV). The authors investigated Hepatitis B Virus (HBV) and Diphtheria/Pertussis/Tetanus toxoid (DPT) vaccination responses following primary immunization in CLHIV (n = 42) and healthy controls (HC) (n = 38) and the effect of an additional vaccine dose. Antibody responses, CD4 and HBV-specific T/B cells were analysed using CMIA/ELISA and flow-cytometry. CLHIV had significantly lower baseline median antibody titres for all vaccines than HC (p <0.02). Differential seroprotection rates observed in CLHIV were, 4.8% for pertussis; 9.5% for HBV; 26.2% for diphtheria and 66.7% for tetanus. WHO staging significantly influenced anti-HBs levels (p = 0.0095). HBsAg-specific CD4+T-cells were significantly higher in CLHIV than HC (p = 0.042). An additional vaccine dose (HBV and Tdap) conferred a higher protection rate for tetanus and diphtheria (p <0.040) in CLHIV. These findings suggest that CLHIV exhibit a hierarchy of vaccine responses affecting antibody levels and protection rate, which was rescued by administering additional vaccine dose.
Collapse
Affiliation(s)
- Sonia Mathew
- Department of Clinical Virology, Christian Medical College, Vellore, Tamil Nadu, 632004, India
| | - Diviya Alex
- Department of Clinical Virology, Christian Medical College, Vellore, Tamil Nadu, 632004, India
| | - John Paul Demosthenes
- Department of Clinical Virology, Christian Medical College, Vellore, Tamil Nadu, 632004, India
| | - Winsley Rose
- Department of Pediatric Infectious Diseases, Christian Medical College, Vellore, Tamil Nadu, 632004, India
| | - Anila Chacko
- Department of Pediatric Infectious Diseases, Christian Medical College, Vellore, Tamil Nadu, 632004, India
| | | | | | - John Antony Jude Prakash
- Department of Clinical Microbiology, Christian Medical College, Vellore, Tamil Nadu, 632004, India
| | - John Mathai
- Department of Pediatric Surgery, Christian Medical College, Vellore, Tamil Nadu, 632004, India
| | | | - Priya Abraham
- Department of Clinical Virology, Christian Medical College, Vellore, Tamil Nadu, 632004, India
| | - Valsan Philip Verghese
- Department of Pediatric Infectious Diseases, Christian Medical College, Vellore, Tamil Nadu, 632004, India.
| | - Rajesh Kannangai
- Department of Clinical Virology, Christian Medical College, Vellore, Tamil Nadu, 632004, India.
| |
Collapse
|
29
|
Ridelfi M, Pierleoni G, Zucconi Galli Fonseca V, Batani G, Rappuoli R, Sala C. State of the Art and Emerging Technologies in Vaccine Design for Respiratory Pathogens. Semin Respir Crit Care Med 2025. [PMID: 39870103 DOI: 10.1055/a-2500-1878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2025]
Abstract
In this review, we present the efforts made so far in developing effective solutions to prevent infections caused by seven major respiratory pathogens: influenza virus, respiratory syncytial virus (RSV), the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), Bordetella pertussis, Streptococcus pneumoniae (pneumococcus), Mycobacterium tuberculosis, and Pseudomonas aeruginosa. Advancements driven by the recent coronavirus disease 2019 (COVID-19) crisis have largely focused on viruses, but effective prophylactic solutions for bacterial pathogens are also needed, especially in light of the antimicrobial resistance (AMR) phenomenon. Here, we discuss various innovative key technologies that can help address this critical need, such as (a) the development of Lung-on-Chip ex vivo models to gain a better understanding of the pathogenesis process and the host-microbe interactions; (b) a more thorough investigation of the mechanisms behind mucosal immunity as the first line of defense against pathogens; (c) the identification of correlates of protection (CoPs) which, in conjunction with the Reverse Vaccinology 2.0 approach, can push a more rational and targeted design of vaccines. By focusing on these critical areas, we expect substantial progress in the development of new vaccines against respiratory bacterial pathogens, thereby enhancing global health protection in the framework of the increasingly concerning AMR emergence.
Collapse
Affiliation(s)
- Matteo Ridelfi
- Monoclonal Antibody Discovery (MAD) Lab, Fondazione Toscana Life Sciences, Siena, Italy
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Giulio Pierleoni
- Monoclonal Antibody Discovery (MAD) Lab, Fondazione Toscana Life Sciences, Siena, Italy
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | | | - Giampiero Batani
- Monoclonal Antibody Discovery (MAD) Lab, Fondazione Toscana Life Sciences, Siena, Italy
| | | | - Claudia Sala
- Monoclonal Antibody Discovery (MAD) Lab, Fondazione Toscana Life Sciences, Siena, Italy
| |
Collapse
|
30
|
Farooq MA, Johnston APR, Trevaskis NL. Impact of nanoparticle properties on immune cell interactions in the lymph node. Acta Biomater 2025; 193:65-82. [PMID: 39701340 DOI: 10.1016/j.actbio.2024.12.039] [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: 08/07/2024] [Revised: 11/21/2024] [Accepted: 12/16/2024] [Indexed: 12/21/2024]
Abstract
The lymphatic system plays an important role in health and many diseases, such as cancer, autoimmune, cardiovascular, metabolic, hepatic, viral, and other infectious diseases. The lymphatic system is, therefore, an important treatment target site for a range of diseases. Lymph nodes (LNs), rich in T cells, B cells, dendritic cells, and macrophages, are also primary sites of action for vaccines and immunotherapies. Promoting the delivery of therapeutics and vaccines to LNs can, therefore, enhance treatment efficacy and facilitate avoidance of off-target side effects by enabling a reduction in therapeutic dose. Several nanoparticle (NP) based delivery systems, such as polymeric NPs, lipid NPs, liposomes, micelles, and dendrimers, have been reported to enhance the delivery of therapeutics and/or vaccines to LNs. Specific uptake into the lymph following injection into tissues is highly dependent on particle properties, particularly particle size, as small molecules are more likely to be taken up by blood capillaries due to higher blood flow rates, whereas larger molecules and NPs can be specifically transported via the lymphatic vessels to LNs as the initial lymphatic capillaries are more permeable than blood capillaries. Once NPs enter LNs, particle properties also have an important influence on their disposition within the node and association with immune cells, which has significant implications for the design of vaccines and immunotherapies. This review article focuses on the impact of NP properties, such as size, surface charge and modification, and route of administration, on lymphatic uptake, retention, and interactions with immune cells in LNs. We suggest that optimizing all these factors can enhance the efficacy of vaccines or therapeutics with targets in the lymphatics and also be helpful for the rational design of vaccines. STATEMENT OF SIGNIFICANCE: The lymphatic system plays an essential role in health and is an important treatment target site for a range of diseases. Promoting the delivery of immunotherapies and vaccines to immune cells in lymph nodes can enhance efficacy and facilitate avoidance of off-target side effects by enabling a reduction in therapeutic dose. One of the major approaches used to deliver therapeutics and vaccines to lymph nodes is via injection in nanoparticle delivery systems. This review aims to provide an overview of the impact of nanoparticle properties, such as size, surface charge, modification, and route of administration, on lymphatic uptake, lymph node retention, and interactions with immune cells in lymph nodes. This will inform the design of future improved nanoparticle systems for vaccines and immunotherapies.
Collapse
Affiliation(s)
- Muhammad Asim Farooq
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, VIC 3052, Australia
| | - Angus P R Johnston
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, VIC 3052, Australia
| | - Natalie L Trevaskis
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, VIC 3052, Australia.
| |
Collapse
|
31
|
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
|
32
|
Zhang B, Fong Y, Dang L, Fintzi J, Chen S, Wang J, Rouphael NG, Branche AR, Diemert DJ, Falsey AR, Graciaa DS, Baden LR, Frey SE, Whitaker JA, Little SJ, Kamidani S, Walter EB, Novak RM, Rupp R, Jackson LA, Yu C, Magaret CA, Molitor C, Borate B, Busch S, Benkeser D, Netzl A, Smith DJ, Babu TM, Kottkamp AC, Luetkemeyer AF, Immergluck LC, Presti RM, Bäcker M, Winokur PL, Mahgoub SM, Goepfert PA, Fusco DN, Atmar RL, Posavad CM, Mu J, Makowski M, Makhene MK, Nayak SU, Roberts PC, Gilbert PB, Follmann D. Neutralizing antibody immune correlates in COVAIL trial recipients of an mRNA second COVID-19 vaccine boost. Nat Commun 2025; 16:759. [PMID: 39824819 PMCID: PMC11748719 DOI: 10.1038/s41467-025-55931-w] [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: 07/19/2024] [Accepted: 01/05/2025] [Indexed: 01/20/2025] Open
Abstract
Neutralizing antibody titer has been a surrogate endpoint for guiding COVID-19 vaccine approval and use, although the pandemic's evolution and the introduction of variant-adapted vaccine boosters raise questions as to this surrogate's contemporary performance. For 985 recipients of an mRNA second bivalent or monovalent booster containing various Spike inserts [Prototype (Ancestral), Beta, Delta, and/or Omicron BA.1 or BA.4/5] in the COVAIL trial (NCT05289037), titers against 5 strains were assessed as correlates of risk of symptomatic COVID-19 ("COVID-19") and as correlates of relative (Pfizer-BioNTech Omicron vs. Prototype) booster protection against COVID-19 over 6 months of follow-up during the BA.2-BA.5 Omicron-dominant period. Consistently across the Moderna and Pfizer-BioNTech vaccine platforms and across all variant Spike inserts assessed, both peak and exposure-proximal ("predicted-at-exposure") titers correlated with lower Omicron COVID-19 risk in individuals previously infected with SARS-CoV-2, albeit significantly less so in naïve individuals [e.g., exposure-proximal hazard ratio per 10-fold increase in BA.1 titer 0.74 (95% CI 0.59, 0.94) for naïve vs. 0.41 (95% CI 0.23, 0.64) for non-naïve; interaction p = 0.013]. Neutralizing antibody titer was a strong inverse correlate of Omicron COVID-19 in non-naïve individuals and a weaker correlate in naïve individuals, posing questions about how prior infection alters the neutralization correlate.
Collapse
Affiliation(s)
- Bo Zhang
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Youyi Fong
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA, USA
| | - Lauren Dang
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jonathan Fintzi
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Shiyu Chen
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Jing Wang
- Clinical Monitoring Research Program Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | | | - Angela R Branche
- Vaccine and Treatment Evaluation Unit, University of Rochester, Rochester, NY, USA
| | - David J Diemert
- George Washington Vaccine Research Unit, George Washington University, Washington, DC, USA
| | - Ann R Falsey
- Vaccine and Treatment Evaluation Unit, University of Rochester, Rochester, NY, USA
| | | | - Lindsey R Baden
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Sharon E Frey
- Center for Vaccine Development, Saint Louis University, St Louis, MO, USA
| | - Jennifer A Whitaker
- Department of Molecular Virology and Microbiology and Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Susan J Little
- Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Satoshi Kamidani
- Center for Childhood Infections and Vaccines, Children's Healthcare of Atlanta, Atlanta, GA, USA
- Department of Pediatrics, Emory University, Atlanta, GA, USA
| | - Emmanuel B Walter
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Richard M Novak
- Project WISH, University of Illinois at Chicago, Chicago, IL, USA
| | - Richard Rupp
- Department of Pediatrics, University of Texas Medical Branch, Galveston, TX, USA
| | - Lisa A Jackson
- Kaiser Permanente Washington Health Research Institute, Seattle, WA, USA
| | - Chenchen Yu
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Craig A Magaret
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Cindy Molitor
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Bhavesh Borate
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Sydney Busch
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - David Benkeser
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Antonia Netzl
- Center for Pathogen Evolution, Department of Zoology, University of Cambridge, Cambridge, UK
| | - Derek J Smith
- Center for Pathogen Evolution, Department of Zoology, University of Cambridge, Cambridge, UK
| | - Tara M Babu
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Angelica C Kottkamp
- Vaccine and Treatment Evaluation Unit, Manhattan Research Clinic, New York University Grossman School of Medicine, New York, NY, USA
| | - Anne F Luetkemeyer
- Division of HIV, Infectious Diseases and Global Medicine, Zuckerberg San Francisco General Hospital, University of California, San Francisco, CA, USA
| | - Lilly C Immergluck
- Clinical Research Center, Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, GA, USA
- Biological Sciences Division, The University of Chicago, Chicago, IL, USA
| | - Rachel M Presti
- Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Martín Bäcker
- Department of Internal Medicine, University of Utah Schoole of Medicine, Salt Lake City, Utah, USA
| | - Patricia L Winokur
- Department of Medicine, University of Iowa College of Medicine, Iowa City, IA, USA
| | - Siham M Mahgoub
- Howard University College of Medicine, Howard University Hospital, Washington, DC, USA
| | - Paul A Goepfert
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Dahlene N Fusco
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Robert L Atmar
- Department of Molecular Virology and Microbiology and Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Christine M Posavad
- Infectious Diseases Clinical Research Consortium (IDCRC) Laboratory Operations Unit, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Jinjian Mu
- The Emmes Company LLC, Rockville, MD, USA
| | | | - Mamodikoe K Makhene
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Seema U Nayak
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Paul C Roberts
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Peter B Gilbert
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA, USA
| | - Dean Follmann
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
33
|
Odio CD, Daag JV, Crisostomo MV, Voirin CJ, Coello Escoto A, Adams C, Dahora Hein L, Aogo RA, Mpingabo PI, Raimundi Rodriguez G, Firdous S, Abad Fernandez M, White LJ, Agrupis KA, Deen J, de Silva AM, Ylade M, Katzelnick LC. Dengue virus IgG and neutralizing antibody titers measured with standard and mature viruses are protective. Nat Commun 2025; 16:191. [PMID: 39747846 PMCID: PMC11697199 DOI: 10.1038/s41467-024-53916-9] [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: 03/29/2024] [Accepted: 10/25/2024] [Indexed: 01/04/2025] Open
Abstract
The standard dengue virus (DENV) neutralization assay inconsistently predicts dengue protection. We compare how IgG ELISA, envelope domain III (EDIII), or non-structural protein 1 (NS1) binding antibodies, and titers from plaque reduction neutralization tests (PRNTs) using standard and mature viruses are associated with dengue. The ELISA measures IgG antibodies that bind to inactivated DENV1-4. The EDIII and NS1 assays measure binding antibodies, and the PRNTs measure neutralizing antibodies to each specific DENV serotype. Healthy children (n = 1206) in Cebu, Philippines were followed for 5 years. ELISA IgG≥3 was associated with reduced dengue probability relative to naïve children (3% vs. 10%, p = 0.007). Serotype-specific antibodies binding EDIII or NS1 had no association with dengue risk. Standard virus PRNT geometric mean titers (GMT) > 200 and mature GMT > 100 were associated with reduced dengue disease overall (p < 0.01). High DENV2 and DENV3 titers against either standard or mature viruses protected against the matched serotype (p < 0.01). While 43% of dengue cases had standard virus PRNT titers>100, only 2% of cases had mature virus PRNT titers>100 (p < 0.001), indicating a lower, more consistent threshold for protection. These assays may serve as correlates of protection.
Collapse
Affiliation(s)
- Camila D Odio
- Viral Epidemiology and Immunity Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Jedas Veronica Daag
- Institute of Child Health and Human Development, National Institutes of Health, University of the Philippines Manila, Manila, Philippines
| | - Maria Vinna Crisostomo
- Institute of Child Health and Human Development, National Institutes of Health, University of the Philippines Manila, Manila, Philippines
| | - Charlie J Voirin
- Viral Epidemiology and Immunity Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Ana Coello Escoto
- Viral Epidemiology and Immunity Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Cameron Adams
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Lindsay Dahora Hein
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Rosemary A Aogo
- Viral Epidemiology and Immunity Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Patrick I Mpingabo
- Viral Epidemiology and Immunity Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Guillermo Raimundi Rodriguez
- Viral Epidemiology and Immunity Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Saba Firdous
- Viral Epidemiology and Immunity Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Maria Abad Fernandez
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Laura J White
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Kristal An Agrupis
- Institute of Child Health and Human Development, National Institutes of Health, University of the Philippines Manila, Manila, Philippines
| | - Jacqueline Deen
- Institute of Child Health and Human Development, National Institutes of Health, University of the Philippines Manila, Manila, Philippines
| | - Aravinda M de Silva
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Michelle Ylade
- Institute of Child Health and Human Development, National Institutes of Health, University of the Philippines Manila, Manila, Philippines.
| | - Leah C Katzelnick
- Viral Epidemiology and Immunity Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA.
| |
Collapse
|
34
|
Ong F, Molenberghs G, Callegaro A, Van Der Elst W, Verbeke G, Stijven F, Keilegom IV, Abad AA. Evaluating Hemagglutination Inhibition Antibody Titers as a Correlate of Protection for Influenza: A Sensitivity Analysis Based on Information Theory and Causal Inference. J Glob Infect Dis 2025; 17:17-23. [PMID: 40290204 PMCID: PMC12021349 DOI: 10.4103/jgid.jgid_89_24] [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: 04/20/2024] [Revised: 07/25/2024] [Accepted: 08/02/2024] [Indexed: 04/30/2025] Open
Abstract
Introduction Identifying hemagglutination inhibition (HI) antibody titers as a key immune correlate of protection (CoP) is crucial for developing, licensing, and monitoring the ongoing effectiveness of new influenza vaccines. Using a new statistical methodology, we explored the link between an inactivated quadrivalent influenza vaccine's impact on HI antibody titers and its effectiveness against A/H1N1-associated influenza illness. Methods We utilized data from a phase 3, observer-blind, randomized, controlled trial in children aged 6-35 months to assess HI antibody titers as an immune CoP. The assessment used a statistical method developed within a causal inference framework and a new information-theoretic metric of surrogacy, the so-called individual causal association (ICA). Results The 75% and 85% uncertainty intervals of the ICA are 0.5511-0.8282 and 0.3632-0.8684, respectively, indicating a substantial reduction in the uncertainty about the vaccine's effect on the absence of infection when its impact on the HI antibody titers is known. Conclusions The evaluation yielded evidence supporting the validity of HI antibody titers as a CoP for influenza infection.
Collapse
Affiliation(s)
- Fenny Ong
- I-BioStat, Universiteit Hasselt, Diepenbeek, Belgium
| | - Geert Molenberghs
- I-BioStat, Universiteit Hasselt, Diepenbeek, Belgium
- I-BioStat, KU Leuven, Leuven, Belgium
| | | | - Wim Van Der Elst
- The Janssen Pharmaceutical Companies of Johnson and Johnson, Beerse, Belgium
| | | | | | | | | |
Collapse
|
35
|
Maltseva M, Keeshan A, Cooper C, Langlois MA. Immune imprinting: The persisting influence of the first antigenic encounter with rapidly evolving viruses. Hum Vaccin Immunother 2024; 20:2384192. [PMID: 39149872 PMCID: PMC11328881 DOI: 10.1080/21645515.2024.2384192] [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: 05/02/2024] [Revised: 07/08/2024] [Accepted: 07/21/2024] [Indexed: 08/17/2024] Open
Abstract
Immune imprinting is a phenomenon that stems from the fundamentals of immunological memory. Upon recurrent exposures to an evolving pathogen, the immune system must weigh the benefits of rapidly recalling established antibody repertoires with greater affinity to the initial variant or invest additional time and energy in producing de novo responses specific to the emerging variant. In this review, we delve into the mechanistic complexities of immune imprinting and its role in shaping subsequent immune responses, both de novo and recall, against rapidly evolving respiratory viruses such as influenza and coronaviruses. By exploring the duality of immune imprinting, we examine its potential to both enhance or hinder immune protection against disease, while emphasizing the role of host and viral factors. Finally, we explore how different vaccine platforms may affect immune imprinting and comment on vaccine strategies that can favor de novo variant-specific antibody responses.
Collapse
Affiliation(s)
- Mariam Maltseva
- Department of Biochemistry, Microbiology & Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Alexa Keeshan
- School of Epidemiology and Public Health, University of Ottawa Faculty of Medicine, Ottawa, ON, Canada
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Curtis Cooper
- School of Epidemiology and Public Health, University of Ottawa Faculty of Medicine, Ottawa, ON, Canada
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Medicine, University of Ottawa, Ottawa, ON, Canada
- Division of Infectious Diseases, Ottawa Hospital Research Institute Clinical Epidemiology Program, Ottawa, ON, Canada
| | - Marc-André Langlois
- Department of Biochemistry, Microbiology & Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
- Center for Infection, Immunity, and Inflammation (CI3), University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
36
|
Guerra A, Costantino C, Martinon-Torres F, Westerholt S, Lambeth C, Chen Z, Lumley J, Marcek T, Johnson D, Wilck M. A phase 4, open-label study to evaluate the safety and immunogenicity of DTaP5-HBV-IPV-Hib in children previously vaccinated with DTaP2-HBV-IPV-Hib or DTaP5-HBV-IPV-Hib (V419-016). Hum Vaccin Immunother 2024; 20:2310900. [PMID: 38327239 PMCID: PMC10857551 DOI: 10.1080/21645515.2024.2310900] [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: 11/15/2023] [Accepted: 01/24/2024] [Indexed: 02/09/2024] Open
Abstract
DTaP5-HBV-IPV-Hib (Vaxelis®) is a hexavalent combination vaccine (HV) indicated in infants and toddlers for the prevention of diphtheria, tetanus, pertussis, hepatitis B, poliomyelitis, and invasive disease due to Haemophilus influenzae type b. Switching between HVs during the childhood vaccination series is sometimes necessary due to, for example, vaccine availability, health-care provider preference, and/or tender awards. The purpose of this study was to describe the safety, tolerability, and immunogenicity of a booster dose of Vaxelis® in participants who previously received a primary infant series of either DTaP2-HBV-IPV-Hib (Hexyon®) or Vaxelis®. Healthy participants approximately 11-13 months of age who previously received a two-dose primary series of Hexyon® (HHV group) or Vaxelis® (VVV group) all received a Vaxelis® booster dose. Immunogenicity was evaluated by measuring antibody levels to individual vaccine antigens approximately 30 days following booster vaccination. Safety was evaluated as the proportion of participants with adverse events (AEs). The proportions of participants with antibody-specific responses for antigens contained in both Vaxelis® and Hexyon® at 30 days post-toddler-booster vaccination with Vaxelis® were comparable between groups, and higher in the VVV group for Vaxelis® antigens PRN and FIM2/3. The overall proportions of participants with AEs were generally comparable between groups. Following a booster dose of Vaxelis®, immune responses were comparable between groups for all shared antigens, and higher in the VVV group for antigens found only in Vaxelis®. The booster was well tolerated in both groups. These data support the use of Vaxelis® as a booster in mixed HV regimens.
Collapse
Affiliation(s)
| | - Claudio Costantino
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Excellence Specialties, University of Palermo, Palermo, Italy
| | - Federico Martinon-Torres
- Translational Pediatrics and Infectious Diseases, Hospital Clínico Universitario de Santiago, Santiago de Compostela, Spain
- GENVIP Research Group (www.genvip.eu), Instituto de Investigación Sanitaria de Santiago, Universidad de Santiago de Compostela, Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - Soeren Westerholt
- Pediatrics, Praxis für Kinder- und Jugendmedizin, Wolfsburg, Germany
| | | | | | | | | | | | | |
Collapse
|
37
|
Hollister J, Porter C, Sprissler R, Beitel SC, Romine JK, Uhrlaub JL, Grant L, Yoo YM, Fowlkes A, Britton A, Olsho LEW, Newes-Adeyi G, Fuller S, Zheng PQ, Gaglani M, Rose S, Dunnigan K, Naleway AL, Gwynn L, Caban-Martinez A, Schaefer Solle N, Tyner HL, Philips AL, Hegmann KT, Yoon S, Lutrick K, Burgess JL, Ellingson KD. Risk reduction in SARS-CoV-2 infection and reinfection conferred by humoral antibody levels among essential workers during Omicron predominance. PLoS One 2024; 19:e0306953. [PMID: 39739951 DOI: 10.1371/journal.pone.0306953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 12/13/2024] [Indexed: 01/02/2025] Open
Abstract
The extent to which semi-quantitative antibody levels confer protection against SARS-CoV-2 infection in populations with heterogenous immune histories is unclear. Two nested case-control studies were designed within the multisite HEROES/RECOVER prospective cohort of frontline workers to study the relationship between antibody levels and protection against first-time post-vaccination infection and reinfection with SARS-CoV-2 from December 2021 to January 2023. All participants submitted weekly nasal swabs for rRT-PCR testing and blood samples quarterly and following infection or vaccination. Cases of first-time post-vaccination infection following a third dose of monovalent (origin strain WA-1) mRNA vaccine (n = 613) and reinfection (n = 350) were 1:1 matched to controls based on timing of blood draw and other potential confounders. Conditional logistic regression models were fit to estimate infection risk reductions associated with 3-fold increases in end titers for receptor binding domain (RBD). In first-time post-vaccination and reinfection study samples, most were female (67%, 57%), non-Hispanic (82%, 68%), and without chronic conditions (65%, 65%). The odds of first-time post-vaccination infection were reduced by 21% (aOR = 0.79, 95% CI = [0.66-0.96]) for each 3-fold increase in RBD end titers. The odds of reinfection associated with a 3-fold increase in RBD end titers were reduced by 23% (aOR = 0.77, 95% CI = [0.65-0.92] for unvaccinated individuals and 58% (aOR = 0.42, 95% CI = [0.22-0.84]) for individuals with three mRNA vaccine doses following their first infection. Frontline workers with higher antibody levels following a third dose of mRNA COVID-19 vaccine were at reduced risk of SARS-CoV-2 during Omicron predominance. Among those with previous infections, the point estimates of risk reduction associated with antibody levels was greater for those with three vaccine doses compared to those who were unvaccinated.
Collapse
Affiliation(s)
- James Hollister
- Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, Arizona, United States of America
| | - Cynthia Porter
- Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, Arizona, United States of America
| | - Ryan Sprissler
- University of Arizona Genetics Core-Center for Applied Genetics and Genomic Medicine, University of Arizona, Tucson, Arizona, United States of America
| | - Shawn C Beitel
- Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, Arizona, United States of America
| | - James K Romine
- Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, Arizona, United States of America
| | - Jennifer L Uhrlaub
- Immunobiology, College of Medicine-Tucson, University of Arizona Health Sciences, University of Arizona, Tucson, Arizona, United States of America
| | - Lauren Grant
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Young M Yoo
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Ashley Fowlkes
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Amadea Britton
- Coronavirus and Other Respiratory Viruses Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | | | | | | | - Pearl Q Zheng
- Abt Associates, Rockville, Maryland, United States of America
| | - Manjusha Gaglani
- Baylor Scott & White Health, Temple, Texas, United States of America
- Texas A&M University College of Medicine, Temple, Texas, United States of America
| | - Spencer Rose
- Baylor Scott & White Health, Temple, Texas, United States of America
| | - Kayan Dunnigan
- Baylor Scott & White Health, Temple, Texas, United States of America
| | - Allison L Naleway
- Kaiser Permanente Center for Health Research, Portland, Oregon, United States of America
| | - Lisa Gwynn
- Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, United States of America
| | - Alberto Caban-Martinez
- Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, United States of America
| | - Natasha Schaefer Solle
- Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, United States of America
| | - Harmony L Tyner
- St. Luke's Regional Health Care System, Duluth, Minnesota, United States of America
| | - Andrew L Philips
- Rocky Mountain Center for Occupational and Environmental Health, Department of Family and Preventive Medicine, University of Utah Health, Salt Lake City, Utah, United States of America
| | - Kurt T Hegmann
- Rocky Mountain Center for Occupational and Environmental Health, Department of Family and Preventive Medicine, University of Utah Health, Salt Lake City, Utah, United States of America
| | - Sarang Yoon
- Rocky Mountain Center for Occupational and Environmental Health, Department of Family and Preventive Medicine, University of Utah Health, Salt Lake City, Utah, United States of America
| | - Karen Lutrick
- Family and Community Medicine, College of Medicine-Tucson, University of Arizona Health Sciences, Tucson, Arizona, United States of America
| | - Jefferey L Burgess
- Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, Arizona, United States of America
| | - Katherine D Ellingson
- Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, Arizona, United States of America
| |
Collapse
|
38
|
Chen L, Qiu Q, Zhu Q, Li J, Xie X, Shao X, Liang J, Zhang W, Zheng H, Li B, Xu L, Zeng H, Sun L. Serological investigation on the prevalence of poliovirus in Guangdong province: A cross-sectional study. Hum Vaccin Immunother 2024; 20:2300156. [PMID: 38189143 PMCID: PMC10793669 DOI: 10.1080/21645515.2023.2300156] [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: 10/05/2023] [Accepted: 12/23/2023] [Indexed: 01/09/2024] Open
Abstract
In 2019, we conducted a cross-sectional study for polio virus seroprevalence in Guangdong province, China. We assessed the positivity rates of poliomyelitis NA and GMT in serum across various demographic groups, and the current findings were compared with pre-switch data from 2014. Using multistage random sampling method, four counties/districts were randomly selected per city, and within each, one general hospital and two township hospitals were chosen. Healthy individuals coming for medical checkups or vaccination were invited. A total of 1318 individual samples were collected and tested. In non-newborn population, age-dependent positivity rates ranged from 77.8% to 100% for PV1 NA and 70.3% to 98.9% for PV3 NA (p < .01). The lowest GMT values for both types (17.03 and 8.46) occurred in the 20 to <30 years age group, while peak GMTs for PV1 and PV3 were observed in 1 to <2 (340.14) and 0 to <1-year (168.90) age groups, respectively. GMTs for PV1 (P = .002) and PV3 (P = .007) in Eastern Guangdong were lower than those in the other three regions. Male participants showed higher GMTs than females (P = .016 and .033, respectively). In newborn population, both males and females showed higher PV1 NA positivity rates and GMTs compared to PV3 (p < .05). Post-switch PV3 NA positivity rates were higher than pre-switch rates (p = .016). GMTs of both PV1 and PV3 were significantly higher post-switch (p < .001). The positivity rates of NAs and GMTs remain high level, which play an important role in resisting poliomyelitis infection. Effect of the converted immunization program was more pronounced than that before.
Collapse
Affiliation(s)
- Linxiang Chen
- Department of Immunization Planning, Luohu District Center for Disease Control and Prevention, Shenzhen, China
- Institute of Immunization Programme, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
| | - Quan Qiu
- Institute of Immunization Programme, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
| | - Qi Zhu
- Institute of Immunization Programme, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
| | - Jialing Li
- Institute of Immunization Programme, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
| | - Xin Xie
- Institute of Immunization Programme, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
| | - Xiaoping Shao
- Institute of Immunization Programme, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
| | - Jian Liang
- Institute of Immunization Programme, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
| | - Wei Zhang
- Institute of Pathogenic Microbiology, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Institute of Pathogenic Microbiology, Guangdong Provincial Key Laboratory of Pathogen Detection for Emerging Infectious Disease Response, China
| | - Huanying Zheng
- Institute of Pathogenic Microbiology, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Institute of Pathogenic Microbiology, Guangdong Provincial Key Laboratory of Pathogen Detection for Emerging Infectious Disease Response, China
| | - Baisheng Li
- Institute of Pathogenic Microbiology, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Institute of Pathogenic Microbiology, Guangdong Provincial Key Laboratory of Pathogen Detection for Emerging Infectious Disease Response, China
| | - Lin Xu
- School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Hanri Zeng
- Institute of Pathogenic Microbiology, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Institute of Pathogenic Microbiology, Guangdong Provincial Key Laboratory of Pathogen Detection for Emerging Infectious Disease Response, China
| | - Limei Sun
- Institute of Immunization Programme, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
| |
Collapse
|
39
|
Bates JT, Lirette ST, Farmer AP, Bierdeman MA, Seyfarth KB, Ederer DR, Montgomery DD, Burnett GC, Pham AT, Marshall GD. Serological assessment of the durability of vaccine-mediated protection against SARS-CoV-2 infection. Hum Vaccin Immunother 2024; 20:2308375. [PMID: 38361363 PMCID: PMC10877977 DOI: 10.1080/21645515.2024.2308375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 01/18/2024] [Indexed: 02/17/2024] Open
Abstract
Virus-neutralizing antibodies are often accepted as a correlate of protection against infection, though questions remain about which components of the immune response protect against SARS-CoV-2 infection. In this small observational study, we longitudinally measured spike receptor binding domain (RBD)-specific and nucleocapsid (NP)-specific serum IgG in a human cohort immunized with the Pfizer BNT162b2 vaccine. NP is not encoded in the vaccine, so an NP-specific response is serological evidence of natural infection. A greater than fourfold increase in NP-specific antibodies was used as the serological marker of infection. Using the RBD-specific IgG titers prior to seroconversion for NP, we calculated a protective threshold for RBD-specific IgG. On average, the RBD-specific IgG response wanes below the protective threshold 169 days following vaccination. Many participants without a history of a positive test result for SARS-CoV-2 infection seroconverted for NP-specific IgG. As a group, participants who seroconverted for NP-specific IgG had significantly higher levels of RBD-specific IgG following NP-seroconversion. RBD-specific IgG titers may serve as one correlate of protection against SARS-CoV-2 infection. These titers wane below the proposed protective threshold approximately six months following immunization. Based on serological evidence of infection, the frequency of breakthrough infections and consequently the level of SARS-CoV-2-specific immunity in the population may be higher than what is predicted based on the frequency of documented infections.
Collapse
Affiliation(s)
- John T. Bates
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, USA
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
- Center for Immunology and Microbial Research, University of Mississippi Medical Center, Jackson, MS, USA
| | - Seth T. Lirette
- School of Population Health, University of Mississippi Medical Center, Jackson, MS, USA
| | - Andrew P. Farmer
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Michael A. Bierdeman
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Kristina B. Seyfarth
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Dallas R. Ederer
- Medical Student Research Program, University of Mississippi Medical Center, Jackson, MS, USA
| | - Denise D. Montgomery
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Grace C. Burnett
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Amanda T. Pham
- Medical Student Research Program, University of Mississippi Medical Center, Jackson, MS, USA
| | - Gailen D. Marshall
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
- Center for Immunology and Microbial Research, University of Mississippi Medical Center, Jackson, MS, USA
- School of Population Health, University of Mississippi Medical Center, Jackson, MS, USA
| |
Collapse
|
40
|
Irvine EB, Darrah PA, Wang S, Wang C, McNamara RP, Roederer M, Seder RA, Lauffenburger DA, Flynn JL, Fortune SM, Alter G. Humoral correlates of protection against Mycobacterium tuberculosis following intravenous BCG vaccination in rhesus macaques. iScience 2024; 27:111128. [PMID: 39669431 PMCID: PMC11634979 DOI: 10.1016/j.isci.2024.111128] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 03/06/2024] [Accepted: 10/04/2024] [Indexed: 12/14/2024] Open
Abstract
Altering Bacille Calmette-Guérin (BCG) immunization from low-dose intradermal (i.d.) to high-dose intravenous (i.v.) vaccination provides a high level of protection against Mycobacterium tuberculosis (Mtb). In addition to strong T cell immunity, i.v. BCG drives robust humoral immune responses that track with bacterial control. However, given the near-complete protection afforded by high-dose i.v. BCG immunization, a precise correlate of protection was difficult to define. Here we leveraged plasma and bronchoalveolar lavage fluid (BAL) from a cohort of rhesus macaques that received decreasing doses of i.v. BCG and aimed to define correlates of immunity following Mtb challenge. We show an i.v. BCG dose-dependent induction of mycobacterial-specific humoral immune responses. Antibody responses at peak immunogenicity predicted bacterial control post-challenge. Multivariate analyses revealed antibody-mediated complement and natural killer (NK) cell-activating humoral networks as key signatures of protective immunity. This work extends our understanding of humoral biomarkers and potential mechanisms of i.v. BCG-mediated protection against Mtb.
Collapse
Affiliation(s)
- Edward B. Irvine
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Patricia A. Darrah
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD 20892, USA
| | - Shu Wang
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Chuangqi Wang
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ryan P. McNamara
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Mario Roederer
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD 20892, USA
| | - Robert A. Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD 20892, USA
| | - Douglas A. Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - JoAnne L. Flynn
- Department of Microbiology and Molecular Genetics and Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Sarah M. Fortune
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| |
Collapse
|
41
|
Brandi R, Paganelli A, D’Amelio R, Giuliani P, Lista F, Salemi S, Paganelli R. mRNA Vaccines Against COVID-19 as Trailblazers for Other Human Infectious Diseases. Vaccines (Basel) 2024; 12:1418. [PMID: 39772079 PMCID: PMC11680146 DOI: 10.3390/vaccines12121418] [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: 11/06/2024] [Revised: 12/07/2024] [Accepted: 12/13/2024] [Indexed: 01/03/2025] Open
Abstract
mRNA vaccines represent a milestone in the history of vaccinology, because they are safe, very effective, quick and cost-effective to produce, easy to adapt should the antigen vary, and able to induce humoral and cellular immunity. METHODS To date, only two COVID-19 mRNA and one RSV vaccines have been approved. However, several mRNA vaccines are currently under development for the prevention of human viral (influenza, human immunodeficiency virus [HIV], Epstein-Barr virus, cytomegalovirus, Zika, respiratory syncytial virus, metapneumovirus/parainfluenza 3, Chikungunya, Nipah, rabies, varicella zoster virus, and herpes simplex virus 1 and 2), bacterial (tuberculosis), and parasitic (malaria) diseases. RESULTS RNA viruses, such as severe acute respiratory syndrome coronavirus (SARS-CoV)-2, HIV, and influenza, are characterized by high variability, thus creating the need to rapidly adapt the vaccines to the circulating viral strain, a task that mRNA vaccines can easily accomplish; however, the speed of variability may be higher than the time needed for a vaccine to be adapted. mRNA vaccines, using lipid nanoparticles as the delivery system, may act as adjuvants, thus powerfully stimulating innate as well as adaptive immunity, both humoral, which is rapidly waning, and cell-mediated, which is highly persistent. Safety profiles were satisfactory, considering that only a slight increase in prognostically favorable anaphylactic reactions in young females and myopericarditis in young males has been observed. CONCLUSIONS The COVID-19 pandemic determined a shift in the use of RNA: after having been used in medicine as micro-RNAs and tumor vaccines, the new era of anti-infectious mRNA vaccines has begun, which is currently in great development, to either improve already available, but unsatisfactory, vaccines or develop protective vaccines against infectious agents for which no preventative tools have been realized yet.
Collapse
Affiliation(s)
- Rossella Brandi
- Istituto di Science Biomediche della Difesa, Stato Maggiore Della Difesa, 00184 Rome, Italy; (R.B.); (F.L.)
| | | | | | - Paolo Giuliani
- Poliambulatorio Montezemolo, Ente Sanitario Militare del Ministero Della Difesa Presso la Corte dei Conti, 00195 Rome, Italy;
| | - Florigio Lista
- Istituto di Science Biomediche della Difesa, Stato Maggiore Della Difesa, 00184 Rome, Italy; (R.B.); (F.L.)
| | - Simonetta Salemi
- Division of Internal Medicine, Azienda Ospedaliero-Universitaria S. Andrea, 00189 Rome, Italy
| | - Roberto Paganelli
- Internal Medicine, Faculty of Medicine and Surgery, Unicamillus, International School of Medicine, 00131 Rome, Italy
| |
Collapse
|
42
|
Clark TW, Tregoning JS, Lister H, Poletti T, Amin F, Nguyen-Van-Tam JS. Recent advances in the influenza virus vaccine landscape: a comprehensive overview of technologies and trials. Clin Microbiol Rev 2024; 37:e0002524. [PMID: 39360831 PMCID: PMC11629632 DOI: 10.1128/cmr.00025-24] [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] [Indexed: 10/05/2024] Open
Abstract
SUMMARYIn the United Kingdom (UK) in 2022/23, influenza virus infections returned to the levels recorded before the COVID-19 pandemic, exerting a substantial burden on an already stretched National Health Service (NHS) through increased primary and emergency care visits and subsequent hospitalizations. Population groups ≤4 years and ≥65 years of age, and those with underlying health conditions, are at the greatest risk of influenza-related hospitalization. Recent advances in influenza virus vaccine technologies may help to mitigate this burden. This review aims to summarize advances in the influenza virus vaccine landscape by describing the different technologies that are currently in use in the UK and more widely. The review also describes vaccine technologies that are under development, including mRNA, and universal influenza virus vaccines which aim to provide broader or increased protection. This is an exciting and important era for influenza virus vaccinations, and advances are critical to protect against a disease that still exerts a substantial burden across all populations and disproportionately impacts the most vulnerable, despite it being over 80 years since the first influenza virus vaccines were deployed.
Collapse
Affiliation(s)
- Tristan W. Clark
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - John S. Tregoning
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | | | | | | | | |
Collapse
|
43
|
Astakhova EA, Baranov KO, Shilova NV, Polyakova SM, Zuev EV, Poteryaev DA, Taranin AV, Filatov AV. Antibody Avidity Maturation Following Booster Vaccination with an Intranasal Adenovirus Salnavac Vaccine. Vaccines (Basel) 2024; 12:1362. [PMID: 39772024 PMCID: PMC11680177 DOI: 10.3390/vaccines12121362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 11/14/2024] [Accepted: 11/19/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND The COVID-19 pandemic has led to the rapid development of new vaccines and methods of testing vaccine-induced immunity. Despite the extensive research that has been conducted on the level of specific antibodies, less attention has been paid to studying the avidity of these antibodies. The avidity of serum antibodies is associated with a vaccine showing high effectiveness and reflects the process of affinity maturation. In the context of vaccines against SARS-CoV-2, only a limited number of studies have investigated the avidity of antibodies, often solely focusing on the wild-type virus following vaccination. This study provides new insights into the avidity of serum antibodies following adenovirus-based boosters. We focused on the effects of an intranasal Salnavac booster, which is compared, using a single analytical platform, to an intramuscular Sputnik V. METHODS The avidity of RBD-specific IgGs and IgAs was investigated through ELISA using urea and biolayer interferometry. RESULTS The results demonstrated the similar avidities of serum antibodies, which were induced by both vaccines for six months post-booster. However, an increase in antibody avidity was observed for the wild-type and Delta variants, but not for the BA.4/5 variant. CONCLUSIONS Collectively, our data provide the insights into antibody avidity maturation after the adenovirus-based vaccines against SARS-CoV-2.
Collapse
Affiliation(s)
- Ekaterina A. Astakhova
- Laboratory of Immunochemistry, National Research Center Institute of Immunology, Federal Medical Biological Agency of Russia, 115522 Moscow, Russia;
- Department of Immunology, Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
- Moscow Center for Advanced Studies, Kulakova Street 20, 123592 Moscow, Russia
| | - Konstantin O. Baranov
- Laboratory of Immunogenetics, Institute of Molecular and Cellular Biology, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia (A.V.T.)
| | - Nadezhda V. Shilova
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Svetlana M. Polyakova
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | | | | | - Alexander V. Taranin
- Laboratory of Immunogenetics, Institute of Molecular and Cellular Biology, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia (A.V.T.)
| | - Alexander V. Filatov
- Laboratory of Immunochemistry, National Research Center Institute of Immunology, Federal Medical Biological Agency of Russia, 115522 Moscow, Russia;
- Department of Immunology, Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
| |
Collapse
|
44
|
Sharma H, Parekh S, Pujari P, Shewale S, Desai S, Kawade A, Lalwani S, Ravi MD, Ramanan PV, Kamath V, Agarwal A, Dogar V, Gautam M, Jaganathan KS, Kumar R, Sharma I, Gairola S. A randomized, active-controlled, multi-centric, phase-II clinical study to assess safety and immunogenicity of a fully liquid DTwP-HepB-IPV-Hib hexavalent vaccine (HEXASIIL®) in Indian toddlers. Vaccine 2024; 42:126380. [PMID: 39303376 DOI: 10.1016/j.vaccine.2024.126380] [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: 11/08/2023] [Revised: 09/12/2024] [Accepted: 09/15/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND Combination vaccines are effective in simplifying complex vaccination schedules involving multiple vaccines. A fully liquid hexavalent diphtheria (D)-tetanus (T)-whole-cell pertussis (wP)- hepatitis B (HepB)-inactivated poliovirus (IPV)-Haemophilus influenzae b (Hib) vaccine (HEXASIIL®), manufactured by Serum Institute of India Pvt. Ltd. was tested for safety and immunogenicity following booster vaccination. METHODS This was a phase-II/III, open label, multicentric, controlled trial in toddlers (phase II) and infants (phase III) in India. This manuscript presents results of phase II. Healthy toddlers aged 12-24 months were randomized (1:1) to receive a 0.5 ml booster dose of HEXASIIL® or comparator Pentavac SD + Poliovac, intramuscularly and followed for 28 days for safety assessment. Blood samples were collected pre-vaccination and 28 days post-vaccination to assess immunogenicity. Descriptive summary statistics were provided for safety and immunogenecity analyses. RESULTS A total of 223 subjects were randomized. One subject droped out prior to dosing, due to consent withdrawal. Thus, 222 subjects received study vaccine (110 HEXASIIL® and 112 comparator). Frequency of solicited adverse events was comparable between HEXASIIL® and comparator (85.5 % vs 90.2 %). Most local and systemic solicited AEs were mild to moderate in severity. All events resolved completely without any sequelae and none led to subject discontinuation. No vaccine related serious AE was reported. Post vaccination, seroprotection rates against tetanus, Hib and polio type 1 and 3 were 100 % in both the groups. Seroprotection rates for diphtheria (99.1 % vs 100 %) and polio type 2 (98.2 % vs 100 %) were observed in HEXASIIL® and comparator group, respectively. For Hepatitis B, seroprotection was >99 % in both groups. Seroconversion observed for Bordetella Pertussis (94.5 % vs 95.4 %) and Pertussis Toxin (77.1 % vs 87.2 %) in HEXASIIL® and comparator group, respectively. CONCLUSION HEXASIIL® vaccine was found to be safe and immunogenic in toddlers and supported its further clinical development in infants. Clinical Trial Registration - CTRI/2019/11/022052.
Collapse
MESH Headings
- Humans
- Infant
- Haemophilus Vaccines/immunology
- Haemophilus Vaccines/adverse effects
- Haemophilus Vaccines/administration & dosage
- Diphtheria-Tetanus-Pertussis Vaccine/immunology
- Diphtheria-Tetanus-Pertussis Vaccine/adverse effects
- Diphtheria-Tetanus-Pertussis Vaccine/administration & dosage
- Vaccines, Combined/immunology
- Vaccines, Combined/adverse effects
- Vaccines, Combined/administration & dosage
- Male
- Hepatitis B Vaccines/immunology
- Hepatitis B Vaccines/adverse effects
- Hepatitis B Vaccines/administration & dosage
- Female
- India
- Antibodies, Bacterial/blood
- Poliovirus Vaccine, Inactivated/immunology
- Poliovirus Vaccine, Inactivated/adverse effects
- Poliovirus Vaccine, Inactivated/administration & dosage
- Antibodies, Viral/blood
- Immunization, Secondary
- Immunization Schedule
- Immunogenicity, Vaccine
- Child, Preschool
Collapse
Affiliation(s)
- Hitt Sharma
- Dept. of Clinical Research, Serum Institute of India Pvt. Ltd., Pune, India.
| | - Sameer Parekh
- Dept. of Clinical Research, Serum Institute of India Pvt. Ltd., Pune, India
| | - Pramod Pujari
- Dept. of Clinical Research, Serum Institute of India Pvt. Ltd., Pune, India
| | - Sunil Shewale
- Dept. of Clinical Research, Serum Institute of India Pvt. Ltd., Pune, India
| | - Shivani Desai
- Dept. of Clinical Research, Serum Institute of India Pvt. Ltd., Pune, India
| | - Anand Kawade
- Dept. of Pediatrics, KEM Hospital Research Centre, Vadu, Pune, India
| | - Sanjay Lalwani
- Dept. of Pediatrics, Bharati Vidyapeeth (Deemed to be University) Medical College & Hospital, Pune, India
| | - M D Ravi
- Dept. of Pediatrics, JSS Hospital, Mysuru, India
| | | | - Veena Kamath
- Dept. of Community Medicine, Kasturba Medical College at Dr TMA Pai Hospital, Udupi, India
| | - Anurag Agarwal
- Dept. of Pediatrics, Maulana Azad Medical College & Lok Nayak Hospital, New Delhi, India
| | - Vikas Dogar
- Dept. of Quality Control, Serum Institute of India Pvt. Ltd., Pune, India
| | - Manish Gautam
- Dept. of Quality Control, Serum Institute of India Pvt. Ltd., Pune, India
| | - K S Jaganathan
- Production Department, Serum Institute of India Pvt. Ltd., Pune, India
| | - Rakesh Kumar
- Production Department, Serum Institute of India Pvt. Ltd., Pune, India
| | - Inderjit Sharma
- Production Department, Serum Institute of India Pvt. Ltd., Pune, India
| | - Sunil Gairola
- Dept. of Quality Control, Serum Institute of India Pvt. Ltd., Pune, India
| |
Collapse
|
45
|
Kimotho J, Sein Y, Sayed S, Shah R, Mwai K, Saleh M, Wanjiku P, Mwacharo J, Nyagwange J, Karanja H, Kutima B, Gitonga JN, Mugo D, Karanu A, Moranga L, Oluoch V, Shah J, Mutiso J, Mburu A, Nneka Z, Betti P, Usyu Mutinda W, Issak Abdi A, Bejon P, Isabella Ochola-Oyier L, M.Warimwe G, Nduati EW, M. Ndungu F. Kinetics of naturally induced binding and neutralising anti-SARS-CoV-2 antibody levels and potencies among SARS-CoV-2 infected Kenyans with diverse grades of COVID-19 severity: an observational study. Wellcome Open Res 2024; 8:350. [PMID: 39640868 PMCID: PMC11617823 DOI: 10.12688/wellcomeopenres.19414.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/26/2024] [Indexed: 12/07/2024] Open
Abstract
Background Given the low levels of coronavirus disease 2019 (COVID-19) vaccine coverage in sub-Saharan Africa (sSA), despite high levels of natural severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) exposures, strategies for extending the breadth and longevity of naturally acquired immunity are warranted. Designing such strategies will require a good understanding of naturally acquired immunity. Methods We measured whole-spike immunoglobulin G (IgG) and spike-receptor binding domain (RBD) total immunoglobulins (Igs) on 585 plasma samples collected longitudinally over five successive time points within six months of COVID-19 diagnosis in 309 COVID-19 patients. We measured antibody-neutralising potency against the wild-type (Wuhan) SARS-CoV-2 pseudovirus in a subset of 51 patients over three successive time points. Binding and neutralising antibody levels and potencies were then tested for correlations with COVID-19 severities. Results Rates of seroconversion increased from day 0 (day of PCR testing) to day 180 (six months) (63.6% to 100 %) and (69.3 % to 97%) for anti-spike-IgG and anti-spike-RBD binding Igs, respectively. Levels of these binding antibodies peaked at day 28 (p<0.01) and were subsequently maintained for six months without significant decay (p>0.99). Similarly, antibody-neutralising potencies peaked at day 28 (p<0.01) but declined by three-fold, six months after COVID-19 diagnosis (p<0.01). Binding antibody levels were highly correlated with neutralising antibody potencies at all the time points analysed (r>0.60, p<0.01). Levels and potencies of binding and neutralising antibodies increased with disease severity. Conclusions Most COVID-19 patients generated SARS-CoV-2 specific binding antibodies that remained stable in the first six months of infection. However, the respective neutralising antibodies decayed three-fold by month-six of COVID-19 diagnosis suggesting that they are short-lived, consistent with what has been observed elsewhere in the world. Thus, regular vaccination boosters are required to sustain the high levels of anti-SARS-CoV-2 naturally acquired neutralising antibody potencies in our population.
Collapse
Affiliation(s)
- John Kimotho
- KEMRI-Wellcome Trust Research Programme, KILIFI, Coast, 230-80108, Kenya
- Pwani University, KILIFI, 230-80108, Kenya
| | - Yiakon Sein
- KEMRI-Wellcome Trust Research Programme, KILIFI, Coast, 230-80108, Kenya
| | - Shahin Sayed
- Aga Khan University Hospital, 3rd Parklands Avenue, Nairobi, 30270 - 00100, Kenya
| | - Reena Shah
- Aga Khan University Hospital, 3rd Parklands Avenue, Nairobi, 30270 - 00100, Kenya
| | - Kennedy Mwai
- KEMRI-Wellcome Trust Research Programme, KILIFI, Coast, 230-80108, Kenya
| | - Mansoor Saleh
- Aga Khan University Hospital, 3rd Parklands Avenue, Nairobi, 30270 - 00100, Kenya
| | - Perpetual Wanjiku
- KEMRI-Wellcome Trust Research Programme, KILIFI, Coast, 230-80108, Kenya
| | - Jedidah Mwacharo
- KEMRI-Wellcome Trust Research Programme, KILIFI, Coast, 230-80108, Kenya
| | - James Nyagwange
- KEMRI-Wellcome Trust Research Programme, KILIFI, Coast, 230-80108, Kenya
| | - Henry Karanja
- KEMRI-Wellcome Trust Research Programme, KILIFI, Coast, 230-80108, Kenya
| | - Bernadette Kutima
- KEMRI-Wellcome Trust Research Programme, KILIFI, Coast, 230-80108, Kenya
| | - John N. Gitonga
- KEMRI-Wellcome Trust Research Programme, KILIFI, Coast, 230-80108, Kenya
| | - Daisy Mugo
- KEMRI-Wellcome Trust Research Programme, KILIFI, Coast, 230-80108, Kenya
| | - Ann Karanu
- Aga Khan University Hospital, 3rd Parklands Avenue, Nairobi, 30270 - 00100, Kenya
| | - Linda Moranga
- KEMRI-Wellcome Trust Research Programme, KILIFI, Coast, 230-80108, Kenya
| | - Viviane Oluoch
- Aga Khan University Hospital, 3rd Parklands Avenue, Nairobi, 30270 - 00100, Kenya
| | - Jasmit Shah
- Aga Khan University Hospital, 3rd Parklands Avenue, Nairobi, 30270 - 00100, Kenya
| | - Julius Mutiso
- Aga Khan University Hospital, 3rd Parklands Avenue, Nairobi, 30270 - 00100, Kenya
| | - Alfred Mburu
- Aga Khan University Hospital, 3rd Parklands Avenue, Nairobi, 30270 - 00100, Kenya
| | - Zaitun Nneka
- Aga Khan University Hospital, 3rd Parklands Avenue, Nairobi, 30270 - 00100, Kenya
| | - Peter Betti
- Aga Khan University Hospital, 3rd Parklands Avenue, Nairobi, 30270 - 00100, Kenya
| | | | - Abdirahman Issak Abdi
- KEMRI-Wellcome Trust Research Programme, KILIFI, Coast, 230-80108, Kenya
- Pwani University, KILIFI, 230-80108, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Philip Bejon
- KEMRI-Wellcome Trust Research Programme, KILIFI, Coast, 230-80108, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Lynette Isabella Ochola-Oyier
- KEMRI-Wellcome Trust Research Programme, KILIFI, Coast, 230-80108, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - George M.Warimwe
- KEMRI-Wellcome Trust Research Programme, KILIFI, Coast, 230-80108, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Eunice W. Nduati
- KEMRI-Wellcome Trust Research Programme, KILIFI, Coast, 230-80108, Kenya
- Pwani University, KILIFI, 230-80108, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Francis M. Ndungu
- KEMRI-Wellcome Trust Research Programme, KILIFI, Coast, 230-80108, Kenya
- Pwani University, KILIFI, 230-80108, Kenya
- Aga Khan University Hospital, 3rd Parklands Avenue, Nairobi, 30270 - 00100, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Division of Infectious Diseases, Department of Medicine Solna and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
46
|
Taha, Eskandari S, Slesarenko VA, Haselhorst T, Semchenko EA, Seib KL. Refinement and optimisation of Neisseria gonorrhoeae NHBA and MetQ vaccine candidates. Vaccine 2024; 42:126416. [PMID: 39368128 DOI: 10.1016/j.vaccine.2024.126416] [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: 07/23/2024] [Revised: 09/06/2024] [Accepted: 09/30/2024] [Indexed: 10/07/2024]
Abstract
Neisseria gonorrhoeae has a significant impact on reproductive health with an estimated 82 million new cases of infection per year worldwide. Due to the ongoing emergence of multidrug-resistant N. gonorrhoeae strains, the high number of asymptomatic cases, and the risk of disease sequelae, the development of a gonococcal vaccine is urgently needed. We have previously described two potential gonococcal vaccine antigens, cNHBA (C-terminal fragment of the Neisseria Heparin Binding Antigen) and MetQ (methionine-binding protein). This study aimed to optimise these antigens for improved immune responses and to facilitate vaccine production, by investigating cNHBA fusions with the full-length MetQ protein or N-terminal and C-terminal MetQ fragments (Met1 and Met2, respectively) adjuvanted with aluminium hydroxide. The cNHBA and MetQ fragments and fusion antigens were all immunogenic in mice, generating a predominantly IgG1 response. Antibodies mediated bacterial killing via both serum bactericidal activity (SBA) and opsonophagocytic activity (OPA), and reduced adherence to cervical and urethral epithelial cells. Among the antigen fusions tested, MetQ-cNHBA and cNHBA-Met2 generated the highest SBA, OPA and adherence blocking titres and are proposed as promising optimised antigens for N. gonorrhoeae vaccine development.
Collapse
Affiliation(s)
- Taha
- Institute for Biomedicine and Glycomics, Griffith University, Gold Coast, QLD, Australia
| | - Sharareh Eskandari
- Institute for Biomedicine and Glycomics, Griffith University, Gold Coast, QLD, Australia
| | - Valentin A Slesarenko
- Institute for Biomedicine and Glycomics, Griffith University, Gold Coast, QLD, Australia
| | - Thomas Haselhorst
- Institute for Biomedicine and Glycomics, Griffith University, Gold Coast, QLD, Australia
| | - Evgeny A Semchenko
- Institute for Biomedicine and Glycomics, Griffith University, Gold Coast, QLD, Australia.
| | - Kate L Seib
- Institute for Biomedicine and Glycomics, Griffith University, Gold Coast, QLD, Australia.
| |
Collapse
|
47
|
Hay JA, Routledge I, Takahashi S. Serodynamics: A primer and synthetic review of methods for epidemiological inference using serological data. Epidemics 2024; 49:100806. [PMID: 39647462 DOI: 10.1016/j.epidem.2024.100806] [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: 01/18/2024] [Revised: 11/21/2024] [Accepted: 11/25/2024] [Indexed: 12/10/2024] Open
Abstract
We present a review and primer of methods to understand epidemiological dynamics and identify past exposures from serological data, referred to as serodynamics. We discuss processing and interpreting serological data prior to fitting serodynamical models, and review approaches for estimating epidemiological trends and past exposures, ranging from serocatalytic models applied to binary serostatus data, to more complex models incorporating quantitative antibody measurements and immunological understanding. Although these methods are seemingly disparate, we demonstrate how they are derived within a common mathematical framework. Finally, we discuss key areas for methodological development to improve scientific discovery and public health insights in seroepidemiology.
Collapse
Affiliation(s)
- James A Hay
- Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom.
| | - Isobel Routledge
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA.
| | - Saki Takahashi
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| |
Collapse
|
48
|
Richel E, Cordsmeier A, Bauer L, Fraedrich K, Vestweber R, Roshani B, Stolte-Leeb N, Ensser A, Stahl-Hennig C, Überla K. Mechanisms of sterilizing immunity provided by an HIV-1 neutralizing antibody against mucosal infection. PLoS Pathog 2024; 20:e1012777. [PMID: 39724193 DOI: 10.1371/journal.ppat.1012777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 11/26/2024] [Indexed: 12/28/2024] Open
Abstract
Broadly neutralizing antibodies (bnAbs) against HIV-1 have been shown to protect from systemic infection. When employing a novel challenge virus that uses HIV-1 Env for entry into target cells during the first replication cycle, but then switches to SIV Env usage, we demonstrated that bnAbs also prevented mucosal infection of the first cells. However, it remained unclear whether antibody Fc-effector functions contribute to this sterilizing immunity. Therefore, additional challenge viruses were produced that contain SIV Env and graded doses of a fusion-defective trimer of HIV-1 Env, to which the bnAb, PGT121 can bind without interfering with the SIV Env-based cell entry. After administration of either PGT121 or its mutant deficient in Fc-effector functions, rhesus macaques were intrarectally exposed to these challenge viruses and to those using either HIV-1 Env or SIV Env for entry into the first cells. Both antibodies similarly reduced infection events with the challenge virus using HIV-1 Env by a factor close to 200. Incorporating fusion-defective HIV-1 Env trimers into the particles of the challenge viruses at densities observed in primary virus isolates did not reduce SIV Env-mediated infection events. The results indicate that the sparsity of bnAb binding-sites on HIV-1 virions limits the contribution of Fc-effector functions to provide sterilizing immunity against mucosal viral infection. Hence, harnessing Fc-effector functions for sterilizing immunity against mucosal HIV-1 infection may require strategies to increase the degree of antibody opsonization.
Collapse
Affiliation(s)
- Elie Richel
- University Hospital Erlangen, Institute of Clinical and Molecular Virology, Friedrich-Alexander Universität Erlangen-Nürnberg, Germany
| | - Arne Cordsmeier
- University Hospital Erlangen, Institute of Clinical and Molecular Virology, Friedrich-Alexander Universität Erlangen-Nürnberg, Germany
| | - Larissa Bauer
- University Hospital Erlangen, Institute of Clinical and Molecular Virology, Friedrich-Alexander Universität Erlangen-Nürnberg, Germany
| | - Kirsten Fraedrich
- University Hospital Erlangen, Institute of Clinical and Molecular Virology, Friedrich-Alexander Universität Erlangen-Nürnberg, Germany
| | | | | | | | - Armin Ensser
- University Hospital Erlangen, Institute of Clinical and Molecular Virology, Friedrich-Alexander Universität Erlangen-Nürnberg, Germany
| | | | - Klaus Überla
- University Hospital Erlangen, Institute of Clinical and Molecular Virology, Friedrich-Alexander Universität Erlangen-Nürnberg, Germany
| |
Collapse
|
49
|
Cagigi A, Tinnirello R, Iannolo G, Douradinha B. Orthoflavivirus zikaense (Zika) vaccines: What are we waiting for? Int J Antimicrob Agents 2024; 64:107367. [PMID: 39490448 DOI: 10.1016/j.ijantimicag.2024.107367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/08/2024] [Accepted: 10/20/2024] [Indexed: 11/05/2024]
Affiliation(s)
- Alberto Cagigi
- International Vaccine Institute (IVI) Europe Regional Office, Solna, Sweden
| | | | | | - Bruno Douradinha
- Vaccine Technology Subgroup, Emerging Pathogens Group, Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
50
|
Kundu J, Le HT, Logan M, Hockman D, Landi A, Crawford K, Wininger M, Johnson J, Kundu JK, Tiffney EA, Urbanowicz RA, Ball JK, Bailey JR, Bukh J, Law M, Foung S, Tyrrell DL, Houghton M, Law JL. Recombinant H77C gpE1/gpE2 heterodimer elicits superior HCV cross-neutralisation than H77C gpE2 alone. J Hepatol 2024; 81:941-948. [PMID: 38986744 PMCID: PMC11830426 DOI: 10.1016/j.jhep.2024.06.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 06/11/2024] [Accepted: 06/13/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND & AIMS An optimal HCV vaccine requires the induction of antibodies that neutralise the infectivity of many heterogenous viral isolates. In this study, we have focused on determining the optimal recombinant envelope glycoprotein component to elicit cross-neutralising antibodies against global HCV genotypes. We compared the immunoreactivity and antigenicity of the HCV genotype 1a strain H77C-derived envelope glycoprotein heterodimer gpE1/gpE2 with that of recombinant gpE2 alone. METHODS Characterisation of the envelope glycoproteins was accomplished by determining their ability to bind to a panel of broadly cross-neutralising monoclonal antibodies. Immunogenicity was determined by testing the ability of vaccine antisera to neutralise the infectivity in vitro of a panel of pseudotyped HCV particles in which gpE1/gpE2 derived from representative isolates of the major global HCV genotypes were displayed. RESULTS gpE1/gpE2 binds to more diverse broadly cross-neutralising antibodies than gpE2 alone and elicits a broader profile of cross-neutralising antibodies in animals, especially against more heterologous, non-1a genotypes. While not all heterologous HCV strains can be potently inhibited in vitro by gpE1/gpE2 antisera derived from a single HCV strain, the breadth of heterologous cross-neutralisation is shown to be substantial. CONCLUSIONS Our work supports the inclusion of gpE1/gpE2 in an HCV vaccine in order to maximise the cross-neutralisation of heterogenous HCV isolates. Our data also offers future directions in formulating a cocktail of gpE1/gpE2 antigens from a small selection of HCV genotypes to further enhance cross-neutralisation of global HCV strains and hopefully advance the development of a globally effective HCV vaccine. IMPACT AND IMPLICATIONS An HCV vaccine is urgently required to prevent the high global incidence of HCV infection and disease. Since HCV is a highly heterogeneous virus, it is desirable for a vaccine to elicit antibodies that neutralise the infectivity of most global strains. To this end, we have compared the immunoreactivity and antigenicity of recombinant H77C E1E2 heterodimer with that of H77C E2 alone and show that the former exhibits more cross-neutralising epitopes and demonstrates a broader cross-neutralisation profile in vitro. In addition, our data suggests a way to further broaden cross-neutralisation using a combination of E1E2 antigens derived from a few different HCV clades. Our work is relevant for the development of an effective global HCV vaccine.
Collapse
Affiliation(s)
- Juthika Kundu
- Li Ka Shing Applied Virology Institute, Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
| | - Hoa T Le
- Li Ka Shing Applied Virology Institute, Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
| | - Michael Logan
- Li Ka Shing Applied Virology Institute, Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
| | - Darren Hockman
- Li Ka Shing Applied Virology Institute, Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
| | - Abdolamir Landi
- Li Ka Shing Applied Virology Institute, Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
| | - Kevin Crawford
- Li Ka Shing Applied Virology Institute, Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
| | - Mark Wininger
- Li Ka Shing Applied Virology Institute, Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
| | - Janelle Johnson
- Li Ka Shing Applied Virology Institute, Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
| | - Joydeb K Kundu
- Li Ka Shing Applied Virology Institute, Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
| | - E Alana Tiffney
- Dept of Infection Biology and Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Richard A Urbanowicz
- Dept of Infection Biology and Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Jonathan K Ball
- Wolfson Centre for Global Virus Infections, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom; Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Justin R Bailey
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jens Bukh
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre and Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mansun Law
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, USA
| | - Steven Foung
- Department of Pathology, Stanford University, Palo Alto, California, USA
| | - D Lorne Tyrrell
- Li Ka Shing Applied Virology Institute, Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
| | - Michael Houghton
- Li Ka Shing Applied Virology Institute, Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada.
| | - John Lokman Law
- Li Ka Shing Applied Virology Institute, Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|