1
|
Abitbol V, Martinón-Torres F, Taha MK, Nolan T, Muzzi A, Bambini S, Borrow R, Toneatto D, Serino L, Rappuoli R, Pizza M. 4CMenB journey to the 10-year anniversary and beyond. Hum Vaccin Immunother 2024; 20:2357924. [PMID: 38976659 PMCID: PMC11232649 DOI: 10.1080/21645515.2024.2357924] [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: 10/19/2023] [Accepted: 05/17/2024] [Indexed: 07/10/2024] Open
Abstract
The 4-component meningococcal serogroup B (MenB) vaccine, 4CMenB, the first broadly protective, protein-based MenB vaccine to be licensed, is now registered in more than 50 countries worldwide. Real-world evidence (RWE) from the last decade confirms its effectiveness and impact, with infant immunization programs showing vaccine effectiveness of 71-95% against invasive MenB disease and cross-protection against non-B serogroups, including a 69% decrease in serogroup W cases in 4CMenB-eligible cohorts in England. RWE from different countries also demonstrates the potential for additional moderate protection against gonorrhea in adolescents. The real-world safety profile of 4CMenB is consistent with prelicensure reports. Use of the endogenous complement human serum bactericidal antibody (enc-hSBA) assay against 110 MenB strains may enable assessment of the immunological effectiveness of multicomponent MenB vaccines in clinical trial settings. Equitable access to 4CMenB vaccination is required to better protect all age groups, including older adults, and vulnerable groups through comprehensive immunization policies.
Collapse
Affiliation(s)
| | - Federico Martinón-Torres
- Genetics, Vaccines and Infections Research Group (GENVIP), Instituto de Investigación Sanitaria de Santiago and Universidad de, Santiago de Compostela, Spain
- Translational Pediatrics and Infectious Diseases, Pediatrics Department, Hospital Clínico Universitario de Santiago, Santiago de Compostela, Spain
- Consorcio Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - Muhamed-Kheir Taha
- Institut Pasteur, Université Paris Cité, Invasive Bacterial Infections Unit, National Reference Center for Meningococci and Haemophilus influenzae, Paris, France
| | - Terry Nolan
- Peter Doherty Institute for Infection & Immunity at University of Melbourne and Murdoch Children's Research Institute, Melbourne, Australia
| | | | | | - Ray Borrow
- Meningococcal Reference Unit, UK Health Security Agency, Manchester, UK
| | | | | | | | | |
Collapse
|
2
|
Efron A, Brozzi A, Biolchi A, Bodini M, Giuliani M, Guidotti S, Lorenzo F, Moscoloni MA, Muzzi A, Nocita F, Pizza M, Rappuoli R, Tomei S, Vidal G, Vizzotti C, Campos J, Sorhouet Pereira C. Genetic characterization and estimated 4CMenB vaccine strain coverage of 284 Neisseria meningitidis isolates causing invasive meningococcal disease in Argentina in 2010-2014. Hum Vaccin Immunother 2024; 20:2378537. [PMID: 39037011 DOI: 10.1080/21645515.2024.2378537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 07/06/2024] [Indexed: 07/23/2024] Open
Abstract
Meningococcal (Neisseria meningitidis) serogroup B (MenB) strain antigens are diverse and a limited number of strains can be evaluated using the human serum bactericidal antibody (hSBA) assay. The genetic Meningococcal Antigen Typing System (gMATS) was developed to predict the likelihood of coverage for large numbers of isolates by the 4CMenB vaccine, which includes antigens Neisseria adhesin A (NadA), Neisserial Heparin-Binding Antigen (NHBA), factor H-binding protein (fHbp), and Porin A (PorA). In this study, we characterized by whole-genome analyses 284 invasive MenB isolates collected from 2010 to 2014 by the Argentinian National Laboratories Network (52-61 isolates per year). Strain coverage was estimated by gMATS on all isolates and by hSBA assay on 74 randomly selected isolates, representative of the whole panel. The four most common clonal complexes (CCs), accounting for 81.3% of isolates, were CC-865 (75 isolates, 26.4%), CC-32 (59, 20.8%), CC-35 (59, 20.8%), and CC-41/44 (38, 13.4%). Vaccine antigen genotyping showed diversity. The most prevalent variants/peptides were fHbp variant 2, NHBA peptides 24, 21, and 2, and PorA variable region 2 profiles 16-36 and 14. The nadA gene was present in 66 (23.2%) isolates. Estimated strain coverage by hSBA assay showed 78.4% of isolates were killed by pooled adolescent sera, and 51.4% and 64.9% (based on two different thresholds) were killed by pooled infant sera. Estimated coverage by gMATS (61.3%; prediction interval: 55.5%, 66.7%) was consistent with the infant hSBA assay results. Continued genomic surveillance is needed to evaluate the persistence of major MenB CCs in Argentina.
Collapse
Affiliation(s)
- Adriana Efron
- Instituto Nacional de Enfermedades Infecciosas-ANLIS "Dr. Carlos G. Malbrán", Buenos Aires, Argentina
| | | | | | | | | | | | - Federico Lorenzo
- Instituto Nacional de Enfermedades Infecciosas-ANLIS "Dr. Carlos G. Malbrán", Buenos Aires, Argentina
| | - María Alicia Moscoloni
- Instituto Nacional de Enfermedades Infecciosas-ANLIS "Dr. Carlos G. Malbrán", Buenos Aires, Argentina
| | | | | | | | | | | | | | - Carla Vizzotti
- National Ministry of Health (2010-2015 and 2019-2023), Buenos Aires, Argentina
| | - Josefina Campos
- Instituto Nacional de Enfermedades Infecciosas-ANLIS "Dr. Carlos G. Malbrán", Buenos Aires, Argentina
| | - Cecilia Sorhouet Pereira
- Instituto Nacional de Enfermedades Infecciosas-ANLIS "Dr. Carlos G. Malbrán", Buenos Aires, Argentina
| |
Collapse
|
3
|
Manoharan S, Farman TA, Piliou S, Mastroeni P. Characterisation and Immunogenicity of Neisseria cinerea outer membrane vesicles displaying NadA, NHBA and fHbp from Neisseria meningitidis serogroup B. Front Immunol 2024; 15:1473064. [PMID: 39380985 PMCID: PMC11458423 DOI: 10.3389/fimmu.2024.1473064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 09/04/2024] [Indexed: 10/10/2024] Open
Abstract
More affordable and effective vaccines against bacterial meningitis caused by Neisseria meningitidis serogroup B are still required for global prevention. We have previously shown that modified outer membrane vesicles (mOMVs) from commensal Neisseria cinerea can be used as a platform to induce immune responses against meningococcal antigens. The aim of the present study was to use a combination of two genetically engineered mOMVs to express multiple antigens from N. meningitidis known to be involved in protective immunity to meningococcal meningitis (different variants of factor H binding protein (fHbp), Neisseria Heparin Binding Antigen (NHBA) and Neisseria Adhesin A (NadA)). Antigen expression in the mOMVs was confirmed by Western blotting; detoxification of the lipooligosaccharide (LOS) was confirmed by measuring human Toll-like receptor 4 (hTLR4) activation using in vitro cell assays. Mice immunised with a combination of two mOMVs expressing fHbp, NHBA and NadA produced antibodies to all the antigens. Furthermore, serum bactericidal activity (SBA) was induced by the immunisation, with mOMVs expressing NadA displaying high SBA titres against a nadA+ MenB strain. The work highlights the potential of mOMVs from N. cinerea to induce functional immune responses against multiple antigens involved in the protective immune response to meningococcal disease.
Collapse
MESH Headings
- Antigens, Bacterial/immunology
- Antigens, Bacterial/genetics
- Animals
- Adhesins, Bacterial/immunology
- Adhesins, Bacterial/genetics
- Neisseria meningitidis, Serogroup B/immunology
- Bacterial Proteins/immunology
- Bacterial Proteins/genetics
- Mice
- Meningococcal Vaccines/immunology
- Humans
- Antibodies, Bacterial/immunology
- Antibodies, Bacterial/blood
- Meningitis, Meningococcal/immunology
- Meningitis, Meningococcal/prevention & control
- Meningitis, Meningococcal/microbiology
- Neisseria cinerea/immunology
- Bacterial Outer Membrane/immunology
- Female
- Extracellular Vesicles/immunology
- Bacterial Outer Membrane Proteins/immunology
- Bacterial Outer Membrane Proteins/genetics
- Mice, Inbred BALB C
- Carrier Proteins
Collapse
Affiliation(s)
- Shathviga Manoharan
- Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | | | | | | |
Collapse
|
4
|
Da Costa RM, Rooke JL, Wells TJ, Cunningham AF, Henderson IR. Type 5 secretion system antigens as vaccines against Gram-negative bacterial infections. NPJ Vaccines 2024; 9:159. [PMID: 39218947 PMCID: PMC11366766 DOI: 10.1038/s41541-024-00953-6] [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: 02/13/2024] [Accepted: 08/14/2024] [Indexed: 09/04/2024] Open
Abstract
Infections caused by Gram-negative bacteria are leading causes of mortality worldwide. Due to the rise in antibiotic resistant strains, there is a desperate need for alternative strategies to control infections caused by these organisms. One such approach is the prevention of infection through vaccination. While live attenuated and heat-killed bacterial vaccines are effective, they can lead to adverse reactions. Newer vaccine technologies focus on utilizing polysaccharide or protein subunits for safer and more targeted vaccination approaches. One promising avenue in this regard is the use of proteins released by the Type 5 secretion system (T5SS). This system is the most prevalent secretion system in Gram-negative bacteria. These proteins are compelling vaccine candidates due to their demonstrated protective role in current licensed vaccines. Notably, Pertactin, FHA, and NadA are integral components of licensed vaccines designed to prevent infections caused by Bordetella pertussis or Neisseria meningitidis. In this review, we delve into the significance of incorporating T5SS proteins into licensed vaccines, their contributions to virulence, conserved structural motifs, and the protective immune responses elicited by these proteins.
Collapse
Affiliation(s)
- Rochelle M Da Costa
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Jessica L Rooke
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Timothy J Wells
- Frazer Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Adam F Cunningham
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Ian R Henderson
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
5
|
Fantoni G, Boccadifuoco G, Verdirosa F, Molesti E, Manenti A, Montomoli E. Current challenges and improvements in assessing the immunogenicity of bacterial vaccines. Front Microbiol 2024; 15:1404637. [PMID: 39044946 PMCID: PMC11263209 DOI: 10.3389/fmicb.2024.1404637] [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: 03/23/2024] [Accepted: 06/26/2024] [Indexed: 07/25/2024] Open
Abstract
The increase in antimicrobial-resistant bacterial strains has highlighted the need for a new vaccine strategy. The primary goal of a candidate vaccine is to prevent disease, by inducing a persistent immunologic memory, through the activation of pathogen-specific immune response. Antibody titer is the main parameter used to assess the immunogenicity of bacterial vaccine candidates and it is the most widely used as a correlate of protection. On the other hand, the antibody titer alone cannot provide complete information on all the activity mediated by antibodies which can only be assessed by functional assays, like the serum bactericidal assay and the opsonophagocytosis assay. However, due to the involvement of many biological factors, these assays are difficult to standardize. Some improvements have been achieved in recent years, but further optimizations are needed to minimize inter- and intra-laboratories variability and to allow the applicability of these functional assays for the vaccine immunogenicity assessment on a larger scale.
Collapse
Affiliation(s)
- Giulia Fantoni
- VisMederi S.r.l., Siena, Italy
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | | | | | | | | | - Emanuele Montomoli
- VisMederi S.r.l., Siena, Italy
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| |
Collapse
|
6
|
Muzzi A, Lu MC, Mori E, Biolchi A, Fu T, Serino L. Prediction by genetic MATS of 4CMenB vaccine strain coverage of invasive meningococcal serogroup B isolates circulating in Taiwan between 2003 and 2020. mSphere 2024; 9:e0022024. [PMID: 38752729 PMCID: PMC11338074 DOI: 10.1128/msphere.00220-24] [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] [Received: 03/19/2024] [Accepted: 04/01/2024] [Indexed: 06/26/2024] Open
Abstract
Neisseria meningitidis serogroup B (NmB) strains have diverse antigens, necessitating methods for predicting meningococcal serogroup B (MenB) vaccine strain coverage. The genetic Meningococcal Antigen Typing System (gMATS), a correlate of MATS estimates, predicts strain coverage by the 4-component MenB (4CMenB) vaccine in cultivable and non-cultivable NmB isolates. In Taiwan, 134 invasive, disease-causing NmB isolates were collected in 2003-2020 (23.1%, 4.5%, 5.2%, 29.8%, and 37.3% from individuals aged ≤11 months, 12-23 months, 2-4 years, 5-29 years, and ≥30 years, respectively). NmB isolates were characterized by whole-genome sequencing and vaccine antigen genotyping, and 4CMenB strain coverage was predicted using gMATS. Analysis of phylogenetic relationships with 502 global NmB genomes showed that most isolates belonged to three global hyperinvasive clonal complexes: ST-4821 (27.6%), ST-32 (23.9%), and ST-41/44 (14.9%). Predicted strain coverage by gMATS was 62.7%, with 27.6% isolates covered, 2.2% not covered, and 66.4% unpredictable by gMATS. Age group coverage point estimates ranged from 42.9% (2-4 years) to 66.1% (≤11 months). Antigen coverage estimates and percentages predicted as covered/not covered were highly variable, with higher estimates for isolates with one or more gMATS-positive antigens than for isolates positive for one 4CMenB antigen. In conclusion, this first study on NmB strain coverage by 4CMenB in Taiwan shows 62.7% coverage by gMATS, with predictable coverage for 29.8% of isolates. These could be underestimated since the gMATS calculation does not consider synergistic mechanisms associated with simultaneous antibody binding to multiple targets elicited by multicomponent vaccines or the contributions of minor outer membrane vesicle vaccine components.IMPORTANCEMeningococcal diseases, caused by the bacterium Neisseria meningitidis (meningococcus), include meningitis and septicemia. Although rare, invasive meningococcal disease is often severe and can be fatal. Nearly all cases are caused by six meningococcal serogroups (types), including meningococcal serogroup B. Vaccines are available against meningococcal serogroup B, but the antigens targeted by these vaccines have highly variable genetic features and expression levels, so the effectiveness of vaccination may vary depending on the strains circulating in particular countries. It is therefore important to test meningococcal serogroup B strains isolated from specific populations to estimate the percentage of bacterial strains that a vaccine can protect against (vaccine strain coverage). Meningococcal isolates were collected in Taiwan between 2003 and 2020, of which 134 were identified as serogroup B. We did further investigations on these isolates, including using a method (called gMATS) to predict vaccine strain coverage by the 4-component meningococcal serogroup B vaccine (4CMenB).
Collapse
Affiliation(s)
| | - Min-Chi Lu
- School of Medicine,
China Medical University,
Taichung, Taiwan
| | | | | | | | | |
Collapse
|
7
|
Cotugno N, Neri A, Sanna M, Santilli V, Manno EC, Pascucci GR, Morrocchi E, Amodio D, Ruggiero A, Ciofi degl Atti ML, Barneschi I, Grappi S, Cocchi I, Giacomet V, Trabattoni D, Olivieri G, Bernardi S, O’Connor D, Montomoli E, Pollard AJ, Palma P. Children with perinatally acquired HIV exhibit distinct immune responses to 4CMenB vaccine. JCI Insight 2024; 9:e177182. [PMID: 38775152 PMCID: PMC11141905 DOI: 10.1172/jci.insight.177182] [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: 10/31/2023] [Accepted: 04/05/2024] [Indexed: 06/02/2024] Open
Abstract
Children with perinatally acquired HIV (PHIV) have special vaccination needs, as they make suboptimal immune responses. Here, we evaluated safety and immunogenicity of 2 doses of 4-component group B meningococcal vaccine in antiretroviral therapy-treated children with PHIV and healthy controls (HCs). Assessments included the standard human serum bactericidal antibody (hSBA) assay and measurement of IgG titers against capsular group B Neisseria meningitidis antigens (fHbp, NHBA, NadA). The B cell compartment and vaccine-induced antigen-specific (fHbp+) B cells were investigated by flow cytometry, and gene expression was investigated by multiplexed real-time PCR. A good safety and immunogenicity profile was shown in both groups; however, PHIV demonstrated a reduced immunogenicity compared with HCs. Additionally, PHIV showed a reduced frequency of fHbp+ and an altered B cell subset distribution, with higher fHbp+ frequency in activated memory and tissue-like memory B cells. Gene expression analyses on these cells revealed distinct mechanisms between PHIV and HC seroconverters. Overall, these data suggest that PHIV presents a diverse immune signature following vaccination. The impact of such perturbation on long-term maintenance of vaccine-induced immunity should be further evaluated in vulnerable populations, such as people with PHIV.
Collapse
Affiliation(s)
- Nicola Cotugno
- Clinical and Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
- Department of Systems Medicine and
| | - Alessia Neri
- Clinical and Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
- PhD Program in Immunology, Molecular Medicine and Applied Biotechnology, University of Rome “Tor Vergata,” Rome, Italy
| | - Marco Sanna
- Clinical and Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Veronica Santilli
- Clinical and Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Emma Concetta Manno
- Clinical and Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Giuseppe Rubens Pascucci
- Clinical and Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
- PhD Program in Immunology, Molecular Medicine and Applied Biotechnology, University of Rome “Tor Vergata,” Rome, Italy
| | - Elena Morrocchi
- Clinical and Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Donato Amodio
- Clinical and Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
- Department of Systems Medicine and
| | - Alessandra Ruggiero
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Marta Luisa Ciofi degl Atti
- Unit of Epidemiology, Clinical Pathways and Clinical Risk, Medical Direction, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | | | | | - Ilaria Cocchi
- Paediatric Infectious Disease Unit, “Luigi Sacco” Hospital, and
| | - Vania Giacomet
- Paediatric Infectious Disease Unit, “Luigi Sacco” Hospital, and
| | - Daria Trabattoni
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Giulio Olivieri
- Clinical and Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
- PhD Program in Immunology, Molecular Medicine and Applied Biotechnology, University of Rome “Tor Vergata,” Rome, Italy
| | - Stefania Bernardi
- Clinical and Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Daniel O’Connor
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Emanuele Montomoli
- VisMederi Life Sciences Srl, Siena, Italy
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Andrew J. Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Paolo Palma
- Clinical and Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
- Department of Systems Medicine and
| |
Collapse
|
8
|
Roe SK, Felter B, Zheng B, Ram S, Wetzler LM, Garges E, Zhu T, Genco CA, Massari P. In Vitro Pre-Clinical Evaluation of a Gonococcal Trivalent Candidate Vaccine Identified by Transcriptomics. Vaccines (Basel) 2023; 11:1846. [PMID: 38140249 PMCID: PMC10747275 DOI: 10.3390/vaccines11121846] [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: 11/06/2023] [Revised: 12/03/2023] [Accepted: 12/09/2023] [Indexed: 12/24/2023] Open
Abstract
Gonorrhea, a sexually transmitted disease caused by Neisseria gonorrhoeae, poses a significant global public health threat. Infection in women can be asymptomatic and may result in severe reproductive complications. Escalating antibiotic resistance underscores the need for an effective vaccine. Approaches being explored include subunit vaccines and outer membrane vesicles (OMVs), but an ideal candidate remains elusive. Meningococcal OMV-based vaccines have been associated with reduced rates of gonorrhea in retrospective epidemiologic studies, and with accelerated gonococcal clearance in mouse vaginal colonization models. Cross-protection is attributed to shared antigens and possibly cross-reactive, bactericidal antibodies. Using a Candidate Antigen Selection Strategy (CASS) based on the gonococcal transcriptome during human mucosal infection, we identified new potential vaccine targets that, when used to immunize mice, induced the production of antibodies with bactericidal activity against N. gonorrhoeae strains. The current study determined antigen recognition by human sera from N. gonorrhoeae-infected subjects, evaluated their potential as a multi-antigen (combination) vaccine in mice and examined the impact of different adjuvants (Alum or Alum+MPLA) on functional antibody responses to N. gonorrhoeae. Our results indicated that a stronger Th1 immune response component induced by Alum+MPLA led to antibodies with improved bactericidal activity. In conclusion, a combination of CASS-derived antigens may be promising for developing effective gonococcal vaccines.
Collapse
Affiliation(s)
- Shea K. Roe
- Department of Immunology, Tufts University School of Medicine, Boston, MA 02111, USA; (S.K.R.); (C.A.G.)
| | - Brian Felter
- Department of Immunology, Tufts University School of Medicine, Boston, MA 02111, USA; (S.K.R.); (C.A.G.)
| | - Bo Zheng
- Division of Infectious Diseases and Immunology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA (S.R.)
| | - Sanjay Ram
- Division of Infectious Diseases and Immunology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA (S.R.)
| | - Lee M. Wetzler
- Section of Infectious Diseases, Boston University School of Medicine, Boston, MA 02118, USA;
| | - Eric Garges
- Department of Preventive Medicine and Biostatistics, F. Edward Hebert School of Medicine, Uniformed Services University, Bethesda, MD 20814, USA;
| | - Tianmou Zhu
- Department of Immunology, Tufts University School of Medicine, Boston, MA 02111, USA; (S.K.R.); (C.A.G.)
| | - Caroline A. Genco
- Department of Immunology, Tufts University School of Medicine, Boston, MA 02111, USA; (S.K.R.); (C.A.G.)
| | - Paola Massari
- Department of Immunology, Tufts University School of Medicine, Boston, MA 02111, USA; (S.K.R.); (C.A.G.)
| |
Collapse
|
9
|
Gray MC, Thomas KS, Lamb ER, Werner LM, Connolly KL, Jerse AE, Criss AK. Evaluating vaccine-elicited antibody activities against Neisseria gonorrhoeae: cross-protective responses elicited by the 4CMenB meningococcal vaccine. Infect Immun 2023; 91:e0030923. [PMID: 37991382 PMCID: PMC10715150 DOI: 10.1128/iai.00309-23] [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: 08/07/2023] [Accepted: 09/27/2023] [Indexed: 11/23/2023] Open
Abstract
The bacterial pathogen Neisseria gonorrhoeae is an urgent global health problem due to increasing numbers of infections, coupled with rampant antibiotic resistance. Vaccines against gonorrhea are being prioritized to combat drug-resistant N. gonorrhoeae. Meningococcal serogroup B vaccines such as four-component meningococcal B vaccine (4CMenB) are predicted by epidemiology studies to cross-protect individuals from natural infection with N. gonorrhoeae and elicit antibodies that cross-react with N. gonorrhoeae. Evaluation of vaccine candidates for gonorrhea requires a suite of assays for predicting efficacy in vitro and in animal models of infection, including the role of antibodies elicited by immunization. Here, we present the development and optimization of assays to evaluate antibody functionality after immunization of mice: antibody binding to intact N. gonorrhoeae, serum bactericidal activity, and opsonophagocytic killing activity using primary human neutrophils [polymorphonuclear leukocytes (PMNs)]. These assays were developed with purified antibodies against N. gonorrhoeae and used to evaluate serum from mice that were vaccinated with 4CMenB or given alum as a negative control. Results from these assays will help prioritize gonorrhea vaccine candidates for advanced preclinical to early clinical studies and will contribute to identifying correlates and mechanisms of immune protection against N. gonorrhoeae.
Collapse
Affiliation(s)
- Mary C. Gray
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Keena S. Thomas
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Evan R. Lamb
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Lacie M. Werner
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Kristie L. Connolly
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, Maryland, USA
| | - Ann E. Jerse
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, Maryland, USA
| | - Alison K. Criss
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| |
Collapse
|
10
|
Peterson J, Drazan D, Czajka H, Maguire J, Pregaldien JL, Seppa II, Maansson R, O'Neill R, Balmer P, Jodar L, Jansen KU, Anderson AS, Perez JL, Beeslaar J. Immunogenicity and safety of a pentavalent meningococcal ABCWY vaccine in adolescents and young adults: an observer-blind, active-controlled, randomised trial. THE LANCET. INFECTIOUS DISEASES 2023; 23:1370-1382. [PMID: 37579773 DOI: 10.1016/s1473-3099(23)00191-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 03/10/2023] [Accepted: 03/15/2023] [Indexed: 08/16/2023]
Abstract
BACKGROUND Meningococcal serogroups A, B, C, W, and Y cause nearly all meningococcal disease, and comprehensive protection requires vaccination against all five serogroups. We aimed to assess the immunogenicity and safety of a pentavalent MenABCWY vaccine comprising two licensed vaccines-meningococcal serogroup B-factor H binding protein vaccine (MenB-FHbp) and a quadrivalent meningococcal serogroup ACWY tetanus toxoid conjugate vaccine (MenACWY-TT)-compared with two doses of MenB-FHbp and a single dose of quadrivalent meningococcal serogroup ACWY CRM197-conjugate vaccine (MenACWY-CRM) as the active control. We previously reported the primary safety and immunogenicity data relating to the two-dose MenB-FHbp schedule. Here we report secondary outcomes and ad-hoc analyses relating to MenABCWY immunogenicity and safety. METHODS We did an observer-blind, active-controlled trial at 68 sites in the USA, Czech Republic, Finland, and Poland. Healthy individuals (aged 10-25 years) who had or had not previously received a MenACWY vaccine were randomly assigned (1:2) using an interactive voice or web-based response system, stratified by previous receipt of a MenACWY vaccine, to receive 0·5 mL of MenABCWY (months 0 and 6) and placebo (month 0) or MenB-FHbp (months 0 and 6) and MenACWY-CRM (month 0) via intramuscular injection into the upper deltoid. All individuals were masked to group allocation, except staff involved in vaccine dispensation, preparation, and administration; and protocol adherence. Endpoints for serogroups A, C, W, and Y included the proportion of participants who achieved at least a four-fold increase in serum bactericidal antibody using human complement (hSBA) titres between baseline and 1 month after each vaccination. For serogroup B, secondary endpoints included the proportion of participants who achieved at least a four-fold increase in hSBA titres from baseline for each of four primary test strains and the proportion of participants who achieved titres of at least the lower limit of quantitation against all four test strains combined at 1 month after the second dose. Endpoints for serogroups A, C, W, and Y were assessed in the modified intent-to-treat (mITT) population, which included all randomly assigned participants who received at least one vaccine dose and had at least one valid and determinate MenB or serogroup A, C, W, or Y assay result before vaccination up to 1 month after the second dose, assessed in ACWY-experienced and ACWY-naive participants separately. Secondary endpoints for serogroup B were analysed in the evaluable immunogenicity population, which included all participants in the mITT population who were randomly assigned to the group of interest, received all investigational products as randomly assigned, had blood drawn for assay testing within the required time frames, had at least one valid and determinate MenB assay result after the second vaccination, and had no important protocol deviations; outcomes were assessed in both ACWY-experienced and ACWY-naive populations combined. Non-inferiority of MenABCWY to MenACWY-CRM and MenB-FHbp was determined using a -10% non-inferiority margin for these endpoints. Reactogenicity and adverse events were assessed among all participants who received at least one vaccine dose and who had available safety data. This trial is registered with Clinicaltrials.gov, NCT03135834, and is complete. FINDINGS Between April 24 and November 10, 2017, 1610 participants (809 MenACWY-naive; 801 MenACWY-experienced) were randomly assigned: 544 to receive MenABCWY and placebo (n=272 MenACWY-naive; n=272 MenACWY-experienced) and 1066 to receive MenB-FHbp and MenACWY-CRM (n=537 MenACWY-naive; n=529 MenACWY-experienced). Among MenACWY-naive or MenACWY-experienced MenABCWY recipients, 75·5% (95% CI 69·8-80·6; 194 of 257; serogroup C) to 96·9% (94·1-98·7; 254 of 262; serogroup A) and 93·0% (88·4-96·2; 174 of 187; serogroup Y) to 97·4% (94·4-99·0; 224 of 230; serogroup W) achieved at least four-fold increases in hSBA titres against serogroups ACWY after dose 1 or 2, respectively, in ad-hoc analyses. Additionally, 75·8% (71·5-79·8; 320 of 422) to 94·7% (92·1-96·7; 396 of 418) of MenABCWY and 67·4% (64·1-70·6; 563 of 835) to 95·0% (93·3-96·4; 782 of 823) of MenB-FHbp recipients achieved at least four-fold increases in hSBA titres against MenB strains after dose 2 in secondary analyses; 79·9% (334 of 418; 75·7-83·6) and 74·3% (71·2-77·3; 605 of 814), respectively, achieved composite responses. MenABCWY was non-inferior to MenACWY-CRM (single dose) and to MenB-FHbp in ad-hoc analyses based on the proportion of participants with at least a four-fold increase in hSBA titres from baseline and (for MenB-FHbp only) composite responses. Reactogenicity events after vaccination were similarly frequent across groups, were mostly mild or moderate, and were unaffected by MenACWY experience. No adverse events causing withdrawals were related to the investigational product. Serious adverse events were reported in four (1·5%; 0·4-3·7) MenACWY-naive individuals in the MenABCWY group versus six (2·2%; 0·8-4·8) among MenACWY-experienced individuals in the MenABCWY group and 14 (1·3%; 0·7-2·2) in the active control group (MenACWY-experienced and MenACWY-naive individuals combined); none of these were considered related to the investigational product. INTERPRETATION MenABCWY immune responses were robust and non-inferior to MenACWY-CRM and MenB-FHbp administered separately, and MenABCWY was well tolerated. The favourable benefit-risk profile supports further MenABCWY evaluation as a simplified schedule compared with current adolescent meningococcal vaccination programmes. FUNDING Pfizer.
Collapse
Affiliation(s)
| | - Daniel Drazan
- General Practice for Children and Adolescents, Jindrichuv Hradec, Czech Republic
| | - Hanna Czajka
- College of Medical Sciences, University of Rzeszow, Rzeszow, Poland; Individual Specialist Medical Practice, Krakow, Poland
| | - Jason Maguire
- Pfizer Vaccine Research and Development, Pearl River, NY, USA.
| | | | - IIkka Seppa
- Tampere University Vaccine Research Center, Tampere, Finland
| | - Roger Maansson
- Pfizer Vaccine Research and Development, Collegeville, PA, USA
| | - Robert O'Neill
- Pfizer Vaccine Research and Development, Pearl River, NY, USA
| | - Paul Balmer
- Pfizer Vaccine Medical Development and Scientific/Clinical Affairs, Collegeville, PA, USA
| | - Luis Jodar
- Pfizer Vaccine Medical Development and Scientific/Clinical Affairs, Collegeville, PA, USA
| | | | | | - John L Perez
- Pfizer Vaccine Research and Development, Collegeville, PA, USA
| | | |
Collapse
|
11
|
Gray MC, Thomas KS, Lamb ER, Werner LM, Connolly KL, Jerse AE, Criss AK. Evaluating vaccine-elicited antibody activities against Neisseria gonorrhoeae: cross-protective responses elicited by the 4CMenB meningococcal vaccine. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.03.551882. [PMID: 37577557 PMCID: PMC10418180 DOI: 10.1101/2023.08.03.551882] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
The bacterial pathogen Neisseria gonorrhoeae is an urgent global health problem due to increasing numbers of infections, coupled with rampant antibiotic resistance. Vaccines against gonorrhea are being prioritized to combat drug-resistant N. gonorrhoeae. Meningococcal serogroup B vaccines such as 4CMenB are predicted by epidemiology studies to cross-protect individuals from natural infection with N. gonorrhoeae and elicit antibodies that cross-react with N. gonorrhoeae. Evaluation of vaccine candidates for gonorrhea requires a suite of assays for predicting efficacy in vitro and in animal models of infection, including the role of antibodies elicited by immunization. Here we present assays to evaluate antibody functionality after immunization: antibody binding to intact N. gonorrhoeae, serum bactericidal activity, and opsonophagocytic killing activity using primary human neutrophils (polymorphonuclear leukocytes). These assays were developed with purified antibodies against N. gonorrhoeae and used to evaluate serum from mice that were vaccinated with 4CMenB or given alum as a negative control. Results from these assays will help prioritize gonorrhea vaccine candidates for advanced preclinical to early clinical study and will contribute to identifying correlates and mechanisms of immune protection against N. gonorrhoeae .
Collapse
|
12
|
Lodi L, Barbati F, Amicizia D, Baldo V, Barbui AM, Bondi A, Costantino C, Da Dalt L, Ferrara L, Fortunato F, Guarnieri V, Icardi G, Indolfi G, Martinelli D, Martini M, Moriondo M, Nieddu F, Peroni DG, Prato R, Ricci S, Russo F, Tirelli F, Vitale F, Ladhani SN, Azzari C. Four-Component Recombinant Protein-Based Vaccine Effectiveness Against Serogroup B Meningococcal Disease in Italy. JAMA Netw Open 2023; 6:e2329678. [PMID: 37594762 PMCID: PMC10439479 DOI: 10.1001/jamanetworkopen.2023.29678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 06/21/2023] [Indexed: 08/19/2023] Open
Abstract
Importance Population-based data on the 4-component recombinant protein-based (4CMenB) vaccine effectiveness and reduction in incidence rate ratios (IRRs) are continuously needed to assess vaccine performance in the prevention of serogroup B invasive meningococcal disease (IMD). Objective To assess the effectiveness and reduction in IRRs associated with the 4CMenB vaccine in the pediatric population in 6 regions in Italy. Design, Setting, and Participants This retrospective cohort screening study and case-control study included data from children aged younger than 6 years in 6 highly populated Italian regions from January 1, 2006, to January 1, 2020. Participants included children younger than 6 years diagnosed with serogroup B IMD without predisposing factors. Data were collected from regional surveillance and vaccination registries and were analyzed from September 2021 to January 2022. Exposures Routine 4CMenB vaccination, per regional vaccination programs. Main Outcomes and Measures The main outcome was the effectiveness of the 4CMenB vaccine in the prevention of serogroup B IMD in the population of children aged younger than 6 years in 6 Italian regions. The percentages of vaccine effectiveness (VE) were obtained through the concomitant use of a screening method and a case-control study. Secondary outcomes were the comparison of effectiveness results obtained using the 2 different computational methods, the description of serogroup B IMD incidence rates, and reduction in IRRs before and after 4CMenB introduction, as a proxy for vaccine impact. Results The cohort screening study included a resident population of 587 561 children younger than 6 years in 3 regions with similar surveillance protocols, and the matched-case controls study assessed a resident population of 1 080 620 children younger than 6 years in 6 regions. Analyses found that 4CMenB VE in fully immunized children was 94.9% (95% CI, 83.1%-98.4%) using the screening method and 91.7% (95% CI, 24.4%-98.6%) using the case-control method. Overall reduction in IRR was 50%, reaching 70% in regions with early-start vaccination schedules. The case-control method involving 6 highly-populated Italian regions included 26 cases and 52 controls and found an estimated VE of 92.4% (95% CI, 67.6%-97.9%) in children old enough for the first vaccine dose and 95.6% (95% CI, 71.7%-99.1%) in fully immunized children. VE was more than 90% for partially immunized children. Even in regions where the first dose was administered at age 2 months, almost 20% of unvaccinated cases were among infants too young to receive the first 4CMenB dose. Conclusions and Relevance This screening cohort study and matched case-controls study found high effectiveness of 4CMenB vaccination and greater reduction in IRR for early-start vaccination schedules in preventing invasive serogroup B meningococcal disease. The high proportion of children too young to be vaccinated among unvaccinated cases suggests that starting the vaccination even earlier may prevent more cases. Screening and case-control methods provided similar estimates of VE: either method may be used in different study settings, but concomitant use can provide more robust estimates.
Collapse
Affiliation(s)
- Lorenzo Lodi
- Immunology Unit, Meyer Children’s Hospital IRCCS, Florence, Italy
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Federica Barbati
- Immunology Unit, Meyer Children’s Hospital IRCCS, Florence, Italy
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Daniela Amicizia
- Department of Health Sciences, University of Genoa, Genoa, Italy
| | - Vincenzo Baldo
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health University of Padua, Padua, Italy
| | - Anna Maria Barbui
- S.C. Microbiology and Virology Laboratory, City of Health and Science, Turin, Italy
| | - Alessandro Bondi
- S.C. Microbiology and Virology Laboratory, City of Health and Science, Turin, Italy
| | - Claudio Costantino
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Excellence Specialties “G. D’Alessandro,” University of Palermo, Palermo, Italy
| | - Liviana Da Dalt
- Department of Woman’s and Child’s Health, Padua University Hospital, Padua, Italy
| | - Lorenza Ferrara
- Regional Epidemiology Reference Service for the Surveillance, Prevention and Control of Infectious Diseases, Local Health Unit of Alessandria, Alessandria, Italy
| | - Francesca Fortunato
- Hygiene Unit, Department of Medical and Surgical Sciences, Policlinico Foggia Hospital, University of Foggia, Foggia, Italy
| | - Valentina Guarnieri
- Immunology Unit, Meyer Children’s Hospital IRCCS, Florence, Italy
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Giancarlo Icardi
- Department of Health Sciences, University of Genoa, Genoa, Italy
| | - Giuseppe Indolfi
- Paediatric and Liver Unit, Meyer Children’s Hospital IRCCS, Florence, Italy
- Department of Neurofarba, University of Florence, Florence, Italy
| | - Domenico Martinelli
- Hygiene Unit, Department of Medical and Surgical Sciences, Policlinico Foggia Hospital, University of Foggia, Foggia, Italy
| | | | - Maria Moriondo
- Laboratory of Immunology and Molecular Microbiology, Meyer Children’s Hospital IRCCS, Florence, Italy
| | - Francesco Nieddu
- Laboratory of Immunology and Molecular Microbiology, Meyer Children’s Hospital IRCCS, Florence, Italy
| | - Diego G. Peroni
- Pediatric Clinic, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Rosa Prato
- Hygiene Unit, Department of Medical and Surgical Sciences, Policlinico Foggia Hospital, University of Foggia, Foggia, Italy
| | - Silvia Ricci
- Immunology Unit, Meyer Children’s Hospital IRCCS, Florence, Italy
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Francesca Russo
- Veneto Regional Directorate of Prevention, Food Safety, Veterinary, Public Health, Venice, Italy
| | - Francesca Tirelli
- Department of Woman’s and Child’s Health, Padua University Hospital, Padua, Italy
| | - Francesco Vitale
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Excellence Specialties “G. D’Alessandro,” University of Palermo, Palermo, Italy
| | - Shamez N. Ladhani
- National Infection Service, Public Health England, London, United Kingdom
- Paediatric Infectious Diseases Research Group, St George’s University of London, London, United Kingdom
| | - Chiara Azzari
- Immunology Unit, Meyer Children’s Hospital IRCCS, Florence, Italy
- Department of Health Sciences, University of Florence, Florence, Italy
| |
Collapse
|
13
|
Borrow R, Martinón-Torres F, Abitbol V, Andani A, Preiss S, Muzzi A, Serino L, Sohn WY. Use of expanded Neisseria meningitidis serogroup B panels with the serum bactericidal antibody assay for the evaluation of meningococcal B vaccine effectiveness. Expert Rev Vaccines 2023; 22:738-748. [PMID: 37622470 DOI: 10.1080/14760584.2023.2244596] [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: 03/21/2023] [Accepted: 08/01/2023] [Indexed: 08/26/2023]
Abstract
INTRODUCTION Neisseria meningitidis serogroup B (NmB) antigens are inherently diverse with variable expression among strains. Prediction of meningococcal B (MenB) vaccine effectiveness therefore requires an assay suitable for use against large panels of epidemiologically representative disease-causing NmB strains. Traditional serum bactericidal antibody assay using exogenous human complement (hSBA) is limited to the quantification of MenB vaccine immunogenicity on a small number of indicator strains. AREAS COVERED Additional and complementary methods for assessing strain coverage developed previously include the Meningococcal Antigen Typing System (MATS), Meningococcal Antigen Surface Expression (MEASURE) assay, and genotyping approaches, but these do not estimate vaccine effectiveness. We provide a narrative review of these methods, highlighting a more recent approach involving the hSBA assay in conjunction with expanded NmB strain panels: hSBA assay using endogenous complement in each vaccinated person's serum (enc-hSBA) against a 110-strain NmB panel and the traditional hSBA assay against 14 (4 + 10) NmB strains. EXPERT OPINION The enc-hSBA is a highly standardized, robust method that can be used in clinical trials to measure the immunological effectiveness of MenB vaccines under conditions that mimic real-world settings as closely as possible, through the use of endogenous complement and a diverse, epidemiologically representative panel of NmB strains.
Collapse
Affiliation(s)
- Ray Borrow
- Meningococcal Reference Unit, UK Health Security Agency, Manchester Royal Infirmary, Manchester, UK
| | - Federico Martinón-Torres
- Pediatrics Research Group (GENVIP), Instituto de Investigación Sanitaria de Santiago and Universidad de Santiago de Compostela, Galicia, Spain
- Translational Pediatrics and Infectious Diseases, Pediatrics Department, Hospital Clínico Universitario de Santiago, Santiago de Compostela, Spain
- Consorcio Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | | | | | | | | | | | | |
Collapse
|
14
|
Aruta MG, De Simone D, Dale H, Chirwa E, Kadwala I, Mbewe M, Banda H, Gordon M, Pizza M, Berlanda Scorza F, Nyirenda T, Canals R, Rossi O. Development and Characterization of a Luminescence-Based High-Throughput Serum Bactericidal Assay (L-SBA) to Assess Bactericidal Activity of Human Sera against Nontyphoidal Salmonella. Methods Protoc 2022; 5:100. [PMID: 36548142 PMCID: PMC9783057 DOI: 10.3390/mps5060100] [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: 09/01/2022] [Revised: 11/30/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022] Open
Abstract
Salmonella Typhimurium and Salmonella Enteritidis are leading causative agents of invasive nontyphoidal Salmonella (iNTS) disease, which represents one of the major causes of death and morbidity in sub-Saharan Africa, still partially underestimated. Large sero-epidemiological studies are necessary to unravel the burden of disease and guide the introduction of vaccines that are not yet available. Even if no correlate of protection has been determined so far for iNTS, the evaluation of complement-mediated functionality of antibodies generated towards natural infection or elicited upon vaccination may represent a big step towards this achievement. Here we present the setup and the intra-laboratory characterization in terms of repeatability, intermediate precision, linearity, and specificity of a high-throughput luminescence-based serum bactericidal assay (L-SBA). This method could be useful to perform sero-epidemiological studies across iNTS endemic countries and for evaluation of antibodies raised against iNTS vaccine candidates in upcoming clinical trials.
Collapse
Affiliation(s)
- Maria Grazia Aruta
- GSK Vaccines Institute for Global Health Srl, Via Fiorentina 1, 53100 Siena, Italy
| | - Daniele De Simone
- GSK Vaccines Institute for Global Health Srl, Via Fiorentina 1, 53100 Siena, Italy
| | - Helen Dale
- Malawi Liverpool Wellcome Trust Programme, Queen Elizabeth Central Hospital, College of Medicine, Blantyre 30096, Malawi
- Pathology Department, Kamuzu University of Health Sciences, Blantyre 312225, Malawi
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 3BX, UK
| | - Esmelda Chirwa
- Malawi Liverpool Wellcome Trust Programme, Queen Elizabeth Central Hospital, College of Medicine, Blantyre 30096, Malawi
- Pathology Department, Kamuzu University of Health Sciences, Blantyre 312225, Malawi
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 3BX, UK
| | - Innocent Kadwala
- Malawi Liverpool Wellcome Trust Programme, Queen Elizabeth Central Hospital, College of Medicine, Blantyre 30096, Malawi
- Pathology Department, Kamuzu University of Health Sciences, Blantyre 312225, Malawi
| | - Maurice Mbewe
- Malawi Liverpool Wellcome Trust Programme, Queen Elizabeth Central Hospital, College of Medicine, Blantyre 30096, Malawi
- Pathology Department, Kamuzu University of Health Sciences, Blantyre 312225, Malawi
| | - Happy Banda
- Malawi Liverpool Wellcome Trust Programme, Queen Elizabeth Central Hospital, College of Medicine, Blantyre 30096, Malawi
- Pathology Department, Kamuzu University of Health Sciences, Blantyre 312225, Malawi
| | - Melita Gordon
- Malawi Liverpool Wellcome Trust Programme, Queen Elizabeth Central Hospital, College of Medicine, Blantyre 30096, Malawi
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 3BX, UK
| | - Mariagrazia Pizza
- GSK Vaccines Institute for Global Health Srl, Via Fiorentina 1, 53100 Siena, Italy
| | | | - Tonney Nyirenda
- Pathology Department, Kamuzu University of Health Sciences, Blantyre 312225, Malawi
| | - Rocío Canals
- GSK Vaccines Institute for Global Health Srl, Via Fiorentina 1, 53100 Siena, Italy
| | - Omar Rossi
- GSK Vaccines Institute for Global Health Srl, Via Fiorentina 1, 53100 Siena, Italy
| | | |
Collapse
|
15
|
Genetic Features of a Representative Panel of 110 Meningococcal B Isolates to Assess the Efficacy of Meningococcal B Vaccines. mSphere 2022; 7:e0038522. [PMID: 36129279 PMCID: PMC9599336 DOI: 10.1128/msphere.00385-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Predictions of vaccine efficacy against Neisseria meningitidis serogroup B (NmB) disease are hindered by antigenic variability, limiting the representativeness of individual NmB isolates. A qualitative human serum bactericidal assay using endogenous complements of individual subjects (enc-hSBA) enables large panels of NmB isolates to be tested. A 110-isolate panel was randomly selected from 442 invasive NmB isolates from United States cases reported to the Centers for Disease Control (CDC) from 2000 to 2008. Typing analyses confirmed the 110-isolate panel is representative of the 442 isolates. The genetic features of the 110-isolate panel were compared against over 4,200 invasive NmB isolates collected from 2000 to 2018 in the United States, Australia, Canada, and nine European countries. Clonal complexes in the 110-isolate panel are also present in each geographical region; cumulative percentages show that these account for around 81% of the clonal complexes found in NmB isolates in other panels. For the antigens (fHbp, NHBA, PorA1.4, NadA) included in the currently licensed meningococcal serogroup B (MenB) vaccines, specifically considering the presence of at least one antigen with a matched genotype, the 110-isolate panel represents approximately 89% of the NmB isolates circulating worldwide, ranging from 87% for the European isolates to 95% and 97% for NmB isolates in the United States and Australia, respectively. The 110-isolate panel includes the most prevalent clonal complexes and genetic variants of MenB vaccine antigens found in a multinational collection of invasive NmB isolates. This panel is useful for assessing the efficacy of MenB vaccines in clinical trials worldwide. IMPORTANCENeisseria meningitidis serogroup B (NmB) is a major cause of invasive meningococcal disease (IMD). Predicting the effectiveness of vaccines against NmB is difficult because NmB is an uncommon disease and because antigens targeted by meningococcal serogroup B (MenB) vaccines have highly variable genetic features and expression levels. Therefore, a large number of NmB isolates from different regions would need to be tested to comprehensively assess vaccine effectiveness. We examined a panel of 110 isolates obtained from NmB IMD cases in the United States and compared the genetic features of this panel with those of panels from different countries around the world. We found the 110-isolate panel included the most common clonal complexes and genetic variants of MenB vaccine antigens that exist in the global collections of invasive NmB isolates. This confirms the value of the NmB 110-isolate panel in understanding the effectiveness of MenB vaccines in clinical trials worldwide.
Collapse
|
16
|
Mancini F, Micoli F, Rossi O. Setup and Characterization of a High-Throughput Luminescence-Based Serum Bactericidal Assay (L-SBA) to Determine Functionality of Human Sera against Shigella flexneri. BIOTECH 2022; 11:biotech11030029. [PMID: 35997337 PMCID: PMC9396978 DOI: 10.3390/biotech11030029] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/19/2022] [Accepted: 07/25/2022] [Indexed: 12/02/2022] Open
Abstract
Shigellosis represents a major public health problem worldwide. The morbidity of the disease, especially in children in developing countries, together with the increase of antimicrobial resistance make a vaccine against Shigella an urgent medical need. Several vaccines under development are targeting Shigella lipopolysaccharide (LPS), whose extreme diversity renders necessary the development of multivalent vaccines. Immunity against Shigella LPS can elicit antibodies capable of killing bacteria in a serotype-specific manner. Therefore, although a correlation of protection against shigellosis has not been established, demonstration of vaccine-elicited antibody bactericidal activity may provide one means of vaccine protection against Shigella. To facilitate Shigella vaccine development, we have set up a high-throughput serum bactericidal assay based on luminescence readout (L-SBA), which has been already used to determine the functionality of antibodies against S. sonnei in multiple clinical trials. Here we present the setup and intra-laboratory characterization of L-SBA against three epidemiologically relevant Shigella flexneri serotypes using human sera. We assessed the linearity, repeatability and reproducibility of the method, demonstrating high assay specificity to detect the activity of antibodies against each homologous strain without any heterologous aspecificity against species-related and non-species-related strains; this assay is ready to be used to determine bactericidal activity of clinical sera raised by multivalent vaccines and in sero-epidemiological studies.
Collapse
|
17
|
Bettencourt C, Nogueira P, Paulo Gomes J, João Simões M. Vaccines against Neisseria meningitidis serogroup B strains - What does genomics reveal on the Portuguese strain's coverage. Vaccine 2022; 40:4772-4779. [PMID: 35778280 DOI: 10.1016/j.vaccine.2022.06.038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 06/02/2022] [Accepted: 06/12/2022] [Indexed: 10/17/2022]
Abstract
In Portugal, Neisseria meningitidis serogroup B (MenB) is the most common serogroup causing invasive meningococcal disease. To protect against MenB disease two protein based MenB vaccines are available in Portugal, the 4CMenB that was licenced in 2014 and included in the routine immunization program in October 2020, and the bivalent rLP2086 vaccine licensed in 2017. The aim of this study was to predict the coverage of the 4CMenB and rLP2086 vaccines against Portuguese isolates of Neisseria meningitidis sampled between 2012 and 2019 and to evaluate the diversity of vaccine antigens based on genomic analysis. Whole-genome sequence data from 324 Portuguese Neisseria meningitidis isolates were analysed. To predict strain coverage by 4CMenB and rLP2086, vaccine antigen reactivity was assessed using the MenDeVar index available on the PubMLST Neisseria website. This study included 235 (75.6%) MenB isolates of all invasive MenB strains reported between 2012 and 2019. Moreover, 89 non MenB isolates sampled in the same period, enrolling 68 from invasive disease and 21 from healthy carriers, were also studied. The predicted strain coverage of MenB isolates was 73.5% (95% CI: 64.8%-81.2%) for 4CMenB and 100% for rLP2086. Predicted strain coverage by 4CMenB in the age group from 0 to 4 years old, was 73.9%. Most of MenB isolates were covered by a single antigen (85.4%), namely fHbp (30.3%), P1.4 (29.2%), and NHBA (24.7%). In Portugal, the most prevalent peptides in MenB isolates were: P1.4 (16.2%), NHBA peptide 2 (14.0%), and fHbp peptide 14 (7.2%), from 4CMenB and fHbp peptide 19 (10.6%) from rLP2086. No significant temporal trends were observed concerning the distribution and diversity of vaccine antigen variants. 4CMenB and rLP2086 vaccines showed potential coverage for isolates regardless serogroup. The use of both vaccines should be considered to control possible outbreaks caused by serogroups with no vaccine available.
Collapse
Affiliation(s)
- Célia Bettencourt
- National Reference Laboratory for Neisseria meningitidis, Department of Infectious Diseases, National Institute of Health (INSA), 1649-016 Lisbon, Portugal.
| | - Paulo Nogueira
- Laboratório de Biomatemática - Instituto de Medicina Preventiva e Saúde Pública, 1649-028 Lisbon, Portugal.
| | - João Paulo Gomes
- Bioinformatics Unit, Department of Infectious Diseases, National Institute of Health (INSA), 1649-016 Lisbon, Portugal.
| | - Maria João Simões
- National Reference Laboratory for Neisseria meningitidis, Department of Infectious Diseases, National Institute of Health (INSA), 1649-016 Lisbon, Portugal.
| |
Collapse
|
18
|
Rollier CS, Dold C, Blackwell L, Linder A, Silva-Reyes L, Clutterbuck E, Davis K, Ford K, Liu X, Holland A, Chan H, Harbinson H, O'Connor D, Borrow R, Snape MD, Pollard AJ. Immunogenicity of a single 4CMenB vaccine booster in adolescents 11 years after childhood immunisation. Vaccine 2022; 40:4453-4463. [PMID: 35697571 DOI: 10.1016/j.vaccine.2022.04.085] [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: 03/10/2022] [Revised: 04/17/2022] [Accepted: 04/27/2022] [Indexed: 11/26/2022]
Abstract
The clinical development of the meningococcal vaccine, 4CMenB, included 2 doses in vaccine-naïve adolescents, which was considered unlikely to be cost-effective for implementation. Theoretically, priming with 4CMenB in early childhood might drive strong immune responses after only a single booster dose in adolescents and reduce programmatic costs. To address this question, children over 11 years old who took part in previous trials involving the administration of 3-5 doses of 4CMenB at infant/preschool age from 2006 were recruited into a post licensure single-centre trial, and were divided into two groups: those who received their last dose at 12 months old (infant group) and those who received their last dose at 3 years old (infant + preschool group). Naïve age-matched controls were randomised to receive one (adolescent 1 group) or two doses at days 0 and 28 (adolescent 2 group) of 4CMenB. Serum bactericidal antibody (SBA) assays using human complement were performed against three reference strains prior to vaccination, and at 1, 6 and 12 months. Previous vaccination was associated with a higher response to a single booster dose at 11 years of age, one-month post-vaccination, when compared with a single dose in naïve age-matched controls. At day 180, the highest responses were observed in participants in the infant + preschool group against strain 5/99 (GMT 316.1 [CI 158.4 to 630.8]), as compared with naïve adolescents who received two doses (GMTs 84.5 [CI 57.7 to 123.6]). When the last dose was received at 12-months of age, responses to a single adolescent dose were not as robust (GMT 61.1 [CI 14.8 to 252.4] to strain 5/99). This descriptive study indicates that the highest SBA responses after a single dose in adolescence were observed in participants who received a preschool dose, suggesting that B cell memory responses are not sufficiently primed at less than 12 months of age. Trial registration EudraCT 2017-004732-11, ISRCTN16774163.
Collapse
Affiliation(s)
- Christine S Rollier
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford OX37LE, UK.
| | - Christina Dold
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford OX37LE, UK
| | - Luke Blackwell
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford OX37LE, UK
| | - Aline Linder
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford OX37LE, UK
| | - Laura Silva-Reyes
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford OX37LE, UK
| | - Elizabeth Clutterbuck
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford OX37LE, UK
| | - Kimberly Davis
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford OX37LE, UK
| | - Karen Ford
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford OX37LE, UK
| | - Xinxue Liu
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford OX37LE, UK
| | - Ann Holland
- UK Health Security Agency, Vaccine Evaluation Unit, Manchester Royal Infirmary, M13 9WL Manchester, UK
| | - Hannah Chan
- UK Health Security Agency, Vaccine Evaluation Unit, Manchester Royal Infirmary, M13 9WL Manchester, UK
| | - Holly Harbinson
- UK Health Security Agency, Vaccine Evaluation Unit, Manchester Royal Infirmary, M13 9WL Manchester, UK
| | - Daniel O'Connor
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford OX37LE, UK
| | - Ray Borrow
- UK Health Security Agency, Vaccine Evaluation Unit, Manchester Royal Infirmary, M13 9WL Manchester, UK
| | - Matthew D Snape
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford OX37LE, UK
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford OX37LE, UK
| |
Collapse
|
19
|
Findlow J, Borrow R, Stephens DS, Liberator P, Anderson AS, Balmer P, Jodar L. Correlates of protection for meningococcal surface protein vaccines; current approaches for the determination of breadth of coverage. Expert Rev Vaccines 2022; 21:753-769. [PMID: 35469524 DOI: 10.1080/14760584.2022.2064850] [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: 11/04/2022]
Abstract
INTRODUCTION The two currently licensed surface protein non capsular meningococcal serogroup B (MenB) vaccines both have the purpose of providing broad coverage against diverse MenB strains. However, the different antigen compositions and approaches used to assess breadth of coverage currently make direct comparisons complex. AREAS COVERED In the second of two companion papers, we comprehensively review the serology and factors influencing breadth of coverage assessments for two currently licensed MenB vaccines. EXPERT OPINION Surface protein MenB vaccines were developed using different approaches, resulting in unique formulations and thus their breadth of coverage. The surface proteins used as vaccine antigens can vary among meningococcal strains due to gene presence/absence, sequence diversity and differences in protein expression. Assessment of the breadth of coverage provided by vaccines is influenced by the ability to induce cross-reactive functional immune responses to sequence diverse protein variants; the characteristics of the circulating invasive strains from specific geographic locations; methodological differences in the immunogenicity assays; differences in human immune responses between individuals; and the maintenance of protective antibody levels over time. Understanding the proportion of meningococcal strains which are covered by the two licensed vaccines is important in understanding protection from disease and public health use.
Collapse
Affiliation(s)
- Jamie Findlow
- Vaccine Medical Development, Scientific and Clinical Affairs, Pfizer Ltd, Tadworth, UK
| | - Ray Borrow
- Meningococcal Reference Unit, Public Health England, Manchester Royal Infirmary, Manchester, UK
| | - David S Stephens
- Woodruff Health Sciences Center, Emory University, Atlanta, Georgia, USA
| | - Paul Liberator
- Vaccine Research and Development, Pfizer Inc, Pearl River, New York, USA
| | | | - Paul Balmer
- Vaccine Medical Development, Scientific and Clinical Affairs, Pfizer Inc, Collegeville, PA, USA
| | - Luis Jodar
- Vaccine Medical Development, Scientific and Clinical Affairs, Pfizer Inc, Collegeville, PA, USA
| |
Collapse
|
20
|
Drazan D, Czajka H, Maguire JD, Pregaldien JL, Maansson R, O'Neill R, Anderson AS, Balmer P, Beeslaar J, Perez JL. A phase 3 study to assess the immunogenicity, safety, and tolerability of MenB-FHbp administered as a 2-dose schedule in adolescents and young adults. Vaccine 2021; 40:351-358. [PMID: 34961633 DOI: 10.1016/j.vaccine.2021.11.053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 11/17/2021] [Accepted: 11/18/2021] [Indexed: 11/15/2022]
Abstract
BACKGROUND The MenB-FHbp vaccine is licensed to prevent meningococcal serogroup B disease on either a 2-dose (0, 6 months) or 3-dose (0, 1-2, 6 months) series. This phase 3 study further assessed the immunogenicity and safety of the 2-dose MenB-FHbp schedule. METHODS Subjects 10-25 years of age received MenB-FHbp (months 0, 6) and the quadrivalent meningococcal conjugate vaccine MenACWY-CRM (month 0). Primary immunogenicity endpoints included percentages of subjects achieving ≥ 4-fold increases from baseline in serum bactericidal antibody using human complement (hSBA) titers for 4 diverse, vaccine-heterologous primary serogroup B test strains and titers ≥ lower limit of quantitation (LLOQ; 1:8 or 1:16) for all 4 primary strains combined (composite response) after dose 2; a titer ≥ 1:4 is the accepted correlate of protection. Percentages of participants with hSBA titers ≥ LLOQ for 10 additional vaccine-heterologous strains were also assessed; positive predictive values of primary strain responses for secondary strain responses were determined. Safety was assessed. RESULTS Overall, 1057 subjects received dose 1 and 946 received dose 2 of MenB-FHbp. Percentages of participants achieving ≥ 4-fold increases in hSBA titers against each primary strain after dose 2 ranged from 67.4% to 95.0% and the composite response was 74.3%. Primary strain responses were highly predictive of secondary strain responses. Most reactogenicity events were mild-to-moderate in severity and did not lead to withdrawal from the study. Adverse events (AEs) considered by the investigator to be related to vaccination occurred in 4.2% (44/1057) of subjects, and there were no serious AEs or newly diagnosed chronic medical conditions considered related to vaccination. CONCLUSIONS MenB-FHbp administered at 0, 6 months was well tolerated and induced protective bactericidal antibody responses against diverse serogroup B strains. Findings provide further support for the continued use of MenB-FHbp on a 2-dose schedule in this population.
Collapse
Affiliation(s)
- Daniel Drazan
- General Practice for Children and Adolescents, Jindrichuv Hradec, Czech Republic
| | - Hanna Czajka
- Faculty of Medicine, University of Rzeszów, Rzeszów, Poland and Individual Specialist Medical Practice, Krakow, Poland
| | - Jason D Maguire
- Vaccine Research and Development, Pfizer Inc, Pearl River, NY, USA.
| | | | - Roger Maansson
- Vaccine Research and Development, Pfizer Inc, Collegeville, PA, USA
| | - Robert O'Neill
- Vaccine Research and Development, Pfizer Inc, Pearl River, NY, USA
| | | | - Paul Balmer
- Vaccine Medical and Scientific Affairs, Pfizer Inc, Collegeville, PA, USA
| | | | - John L Perez
- Vaccine Research and Development, Pfizer Inc, Collegeville, PA, USA
| |
Collapse
|
21
|
Hames RG, Jasiunaite Z, Wanford JJ, Carreno D, Chung WY, Dennison AR, Oggioni MR. Analyzing Macrophage Infection at the Organ Level. Methods Mol Biol 2021; 2414:405-431. [PMID: 34784049 DOI: 10.1007/978-1-0716-1900-1_22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Classical in vivo infection models are oftentimes associated with speculation due to the many physiological factors that are unseen or not accounted for when analyzing experimental outputs, especially when solely utilizing the classic approach of tissue-derived colony-forming unit (CFU) enumeration. To better understand the steps and natural progression of bacterial infection, the pathophysiology of individual organs with which the bacteria interact in their natural course of infection must be considered. In this case, it is not only important to isolate organs as much as possible from additional physiological processes, but to also consider the dynamics of the bacteria at the cellular level within these organs of interest. Here, we describe in detail two models, ex vivo porcine liver and spleen coperfusion and a murine infection model, and the numerous associated experimental outputs produced by these models that can be taken and used together to investigate the pathogen-host interactions within tissues in depth.
Collapse
Affiliation(s)
- Ryan G Hames
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | - Zydrune Jasiunaite
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | - Joseph J Wanford
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | - David Carreno
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | - Wen Y Chung
- Department of Hepatobiliary and Pancreatic Surgery, University Hospitals of Leicester, Leicester, UK
| | - Ashley R Dennison
- Department of Hepatobiliary and Pancreatic Surgery, University Hospitals of Leicester, Leicester, UK
| | - Marco R Oggioni
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK. .,Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy.
| |
Collapse
|
22
|
Martins RM, Périssé ARS, Camacho LAB, Leal ML, Maia MLS, Homma A, Jessouroun E. Phase I safety and immunogenicity study of a Brazilian serogroup B vaccine. Braz J Infect Dis 2021; 25:101652. [PMID: 34793713 PMCID: PMC9392203 DOI: 10.1016/j.bjid.2021.101652] [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: 07/12/2021] [Revised: 10/06/2021] [Accepted: 10/21/2021] [Indexed: 12/03/2022] Open
Abstract
Meningococcal disease by serogroup B has been a public health problem in Brazil in the last decades. The Brazilian Oswaldo Cruz Foundation has been working to develop a vaccine with detergent-treated outer membrane vesicles (OMV) and detoxified endotoxin (dLOS) from Neisseria meningitidis serogroup B prevalent strains. A phase I study, enrolling 26 adults (18–44 years of age) was performed using experimental vaccines combining B components and aluminum hydroxide as adjuvant. It was a dose escalation study testing vaccines made of 25, 50, and 100 µg OMV protein/mL (sum of both strains) and dLOS in half amount of total protein concentration, with three doses given two months apart. Adverse events were mild/moderate with frequency increasing with the amount of antigens. Pain in the site of injection was the most frequent reaction in all doses, reported in more than the 85% across vaccine groups. Considering all injections, cephalea was the most common systemic adverse event, detected in 11.1%, 17.2% and 32.1%, respectively with doses of 12.5 μg, 25 μg and 50 μg. High titers of total IgG (ELISA) were observed for the vaccine components before vaccination. Protective levels of bactericidal antibodies (titer ≥1:4) for both vaccine strains were also present. Considering a 4-fold increase of IgG titers compared to pre-immune values (seroconversion), 50%-70% of those who received intermediate and highest doses of antigens presented satisfactory response for OMV of N44/89 strain. The lowest dose vaccine induced no seroconversion for strain N44/89, and 11% for strain N603/95. For the three vaccines doses, 25% of seroconversion, in total IgG against LOS, was observed. Increased antibody bactericidal activity was observed for both strains in higher antigen concentrations. For IgG against LOS, all vaccine formulations showed 25% of seroconversion. In conclusion, MenB-Bio experimental vaccines were well tolerated and immunogenic, thus allowing phase II studies.
Collapse
Affiliation(s)
- R Menezes Martins
- Clinical Advisory Unit, Bio-Manguinhos/Fiocruz, Rio de Janeiro, RJ, Brazil
| | - A R S Périssé
- National School of Public Health, Fiocruz, Rio de Janeiro, RJ, Brazil
| | - L A B Camacho
- National School of Public Health, Fiocruz, Rio de Janeiro, RJ, Brazil
| | - M L Leal
- Bacterial Technology Laboratory, Bio-Manguinhos/Fiocruz, Rio de Janeiro, RJ, Brazil
| | - M L S Maia
- Clinical Advisory Unit, Bio-Manguinhos/Fiocruz, Rio de Janeiro, RJ, Brazil
| | - A Homma
- Bio-Manguinhos/Fiocruz, Rio de Janeiro, RJ, Brazil
| | - E Jessouroun
- Bacterial Technology Laboratory, Bio-Manguinhos/Fiocruz, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
23
|
Kitiyodom S, Yata T, Thompson KD, Costa J, Elumalai P, Katagiri T, Temisak S, Namdee K, Rodkhum C, Pirarat N. Immersion Vaccination by a Biomimetic-Mucoadhesive Nanovaccine Induces Humoral Immune Response of Red Tilapia ( Oreochromis sp.) against Flavobacterium columnare Challenge. Vaccines (Basel) 2021; 9:vaccines9111253. [PMID: 34835184 PMCID: PMC8624005 DOI: 10.3390/vaccines9111253] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/27/2021] [Accepted: 10/27/2021] [Indexed: 11/21/2022] Open
Abstract
Immersion vaccination with a biomimetic mucoadhesive nanovaccine has been shown to induce a strong mucosal immune response against columnaris disease, a serious bacterial disease in farmed red tilapia caused by Flavobacterium columnare. However, the induction of a systemic immune response by the vaccine is yet to be investigated. Here, we examine if a specific humoral immune response is stimulated in tilapia by a biomimetic-mucoadhesive nanovaccine against Flavobacterium columnare using an indirect-enzyme-linked immunosorbent assay (ELISA), serum bactericidal activity (SBA) and the expression of immune-related genes within the head-kidney and spleen, together with assessing the relative percent survival of vaccinated fish after experimentally infecting them with F. columnare. The anti-IgM antibody titer of fish at 14 and 21 days post-vaccination was significantly higher in chitosan complex nanoemulsion (CS-NE) vaccinated fish compared to fish vaccinated with the formalin-killed vaccine or control fish, supporting the serum bactericidal activity results at these time points. The cumulative mortality of the unvaccinated control fish was 87% after challenging fish with the pathogen, while the cumulative mortality of the CS-NE vaccinated group was 24%, which was significantly lower than the formalin-killed vaccinated and control fish. There was a significant upregulation of IgM, IgT, TNF α, and IL1-β genes in the spleen and kidney of vaccinated fish. Significant upregulation of IgM and IgT genes was observed in the spleen of CS-NE vaccinated fish. The study confirmed the charged-chitosan-based mucoadhesive nanovaccine to be an effective platform for immersion vaccination of tilapia, with fish generating a humoral systemic immune response against columnaris disease in vaccinated fish.
Collapse
Affiliation(s)
- Sirikorn Kitiyodom
- Wildlife Exotic Aquatic Animal Pathology-Research Unit, Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand;
| | - Teerapong Yata
- Biochemistry Unit, Department of Physiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand;
| | - Kim D. Thompson
- Moredun Research Institute, Pentlands Science Park, Penicuik EH26 0PZ, UK;
- Correspondence: (K.D.T.); (N.P.)
| | - Janina Costa
- Moredun Research Institute, Pentlands Science Park, Penicuik EH26 0PZ, UK;
| | - Preetham Elumalai
- School of Ocean Science and Technology, Kerala University of Fisheries and Ocean Studies, Kochi 682506, Kerala, India;
| | - Takayuki Katagiri
- Laboratory of Fish Health Management, Course of Aquatic Biosciences, Tokyo University of Marine Science and Technology, Tokyo 108-8477, Japan;
| | - Sasithon Temisak
- Bio Analysis Group, Chemical Metrology and Biometry Department, National Institute of Metrology (NIMT), Pathum Thani 12120, Thailand;
| | - Katawut Namdee
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani 12120, Thailand;
| | - Channarong Rodkhum
- Department of Microbiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand;
| | - Nopadon Pirarat
- Wildlife Exotic Aquatic Animal Pathology-Research Unit, Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand;
- Correspondence: (K.D.T.); (N.P.)
| |
Collapse
|
24
|
Increasing the High Throughput of a Luminescence-Based Serum Bactericidal Assay (L-SBA). BIOTECH 2021; 10:biotech10030019. [PMID: 35822773 PMCID: PMC9245470 DOI: 10.3390/biotech10030019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/02/2021] [Accepted: 09/16/2021] [Indexed: 11/16/2022] Open
Abstract
Serum bactericidal assay (SBA) is the method to investigate in vitro complement-mediated bactericidal activity of sera raised upon vaccination. The assay is based on incubating the target bacteria and exogenous complement with sera at different dilutions and the result of the assay is represented by the sera dilution being able to kill 50% of bacteria present in the inoculum. The traditional readout of the assay is based on measurement of colony-forming units (CFU) obtained after plating different reaction mixes on agar. This readout is at low throughput and time consuming, even when automated counting is used. We previously described a novel assay with a luminescence readout (L-SBA) based on measurement of ATP released by live bacteria, which allowed to substantially increase the throughput as well as to reduce the time necessary to perform the assay when compared to traditional methods. Here we present a further improvement of the assay by moving from a 96-well to a 384-well format, which allowed us to further increase the throughput and substantially reduce costs while maintaining the high performance of the previously described L-SBA method. The method has been successfully applied to a variety of different pathogens.
Collapse
|
25
|
Findlow J, Lucidarme J, Taha MK, Burman C, Balmer P. Correlates of protection for meningococcal surface protein vaccines: lessons from the past. Expert Rev Vaccines 2021; 21:739-751. [PMID: 34287103 DOI: 10.1080/14760584.2021.1940144] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Recombinant surface protein meningococcal serogroup B (MenB) vaccines are available but with different antigen compositions, leading to differences between vaccines in their immunogenicity and likely breadth of coverage. The serology and breadth of coverage assessment for MenB vaccines are multifaceted areas, and a comprehensive understanding of these complexities is required to appropriately compare licensed vaccines and those under development. AREAS COVERED In the first of two companion papers that comprehensively review the serology and breadth of coverage assessment for MenB vaccines, the history of early meningococcal vaccines is considered in this narrative review to identify transferable lessons applicable to the currently licensed MenB vaccines and those under development, as well as their serology. EXPERT OPINION Understanding correlates of protection and the breadth of coverage assessment for meningococcal surface protein vaccines is significantly more complex than that for capsular polysaccharide vaccines. Determination and understanding of the breadth of coverage of surface protein vaccines are clinically important and unique to each vaccine formulation. It is essential to estimate the proportion of MenB cases that are preventable by a specific vaccine to assess its overall potential impact and to compare the benefits and limitations of different vaccines in preventing invasive meningococcal disease.
Collapse
Affiliation(s)
- Jamie Findlow
- Vaccine Medical Development, Scientific and Clinical Affairs, Pfizer Ltd, Tadworth, UK
| | - Jay Lucidarme
- Meningococcal Reference Unit, Public Health England, Manchester Royal Infirmary, Manchester, UK
| | | | - Cynthia Burman
- Vaccine Medical Development, Scientific and Clinical Affairs, Pfizer Inc, Collegeville, PA, USA
| | - Paul Balmer
- Vaccine Medical Development, Scientific and Clinical Affairs, Pfizer Inc, Collegeville, PA, USA
| |
Collapse
|
26
|
Exploring the Ability of Meningococcal Vaccines to Elicit Mucosal Immunity: Insights from Humans and Mice. Pathogens 2021; 10:pathogens10070906. [PMID: 34358056 PMCID: PMC8308890 DOI: 10.3390/pathogens10070906] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 07/01/2021] [Accepted: 07/09/2021] [Indexed: 12/16/2022] Open
Abstract
Neisseria meningitidis causes a devastating invasive disease but is also a normal colonizer of the human nasopharynx. Due to the rapid progression of disease, the best tool to protect individuals against meningococcal infections is immunization. Clinical experience with polysaccharide conjugate vaccines has revealed that an ideal meningococcal vaccine must prevent both invasive disease and nasal colonization, which confers herd immunity. However, not all meningococcal vaccines are equal in their ability to prevent nasal colonization, for unknown reasons. Herein, we describe recent efforts to utilize humanized mouse models to understand the impact of different meningococcal vaccines on nasal colonization. These mice are susceptible to nasal colonization, and they become immune following live nasal infection or immunization with matched capsule-conjugate or protein-based vaccines, replicating findings from human work. We bring together insights regarding meningococcal colonization and immunity from clinical work with findings using humanized mouse models, providing new perspective into the different determinants of mucosal versus systemic immunity. Then, we use this as a framework to help focus future studies toward understanding key mechanistic aspects left unresolved, including the bacterial factors required for colonization and immune evasion, determinants of nasal mucosal protection, and characteristics of an ideal meningococcal vaccine.
Collapse
|
27
|
Laver JR, Gbesemete D, Dale AP, Pounce ZC, Webb CN, Roche EF, Guy JM, Berreen G, Belogiannis K, Hill AR, Ibrahim MM, Ahmed M, Cleary DW, Pandey AK, Humphries HE, Allen L, de Graaf H, Maiden MC, Faust SN, Gorringe AR, Read RC. A recombinant commensal bacteria elicits heterologous antigen-specific immune responses during pharyngeal carriage. Sci Transl Med 2021; 13:eabe8573. [PMID: 34233953 PMCID: PMC7615050 DOI: 10.1126/scitranslmed.abe8573] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 03/22/2021] [Accepted: 05/25/2021] [Indexed: 12/25/2022]
Abstract
The human nasopharynx contains a stable microbial ecosystem of commensal and potentially pathogenic bacteria, which can elicit protective primary and secondary immune responses. Experimental intranasal infection of human adults with the commensal Neisseria lactamica produced safe, sustained pharyngeal colonization. This has potential utility as a vehicle for sustained release of antigen to the human mucosa, but commensals in general are thought to be immunologically tolerated. Here, we show that engineered N. lactamica, chromosomally transformed to express a heterologous vaccine antigen, safely induces systemic, antigen-specific immune responses during carriage in humans. When the N. lactamica expressing the meningococcal antigen Neisseria Adhesin A (NadA) was inoculated intranasally into human volunteers, all colonized participants carried the bacteria asymptomatically for at least 28 days, with most (86%) still carrying the bacteria at 90 days. Compared to an otherwise isogenic but phenotypically wild-type strain, colonization with NadA-expressing N. lactamica generated NadA-specific immunoglobulin G (IgG)- and IgA-secreting plasma cells within 14 days of colonization and NadA-specific IgG memory B cells within 28 days of colonization. NadA-specific IgG memory B cells were detected in peripheral blood of colonized participants for at least 90 days. Over the same period, there was seroconversion against NadA and generation of serum bactericidal antibody activity against a NadA-expressing meningococcus. The controlled infection was safe, and there was no transmission to adult bedroom sharers during the 90-day period. Genetically modified N. lactamica could therefore be used to generate beneficial immune responses to heterologous antigens during sustained pharyngeal carriage.
Collapse
Affiliation(s)
- Jay R Laver
- Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK.
- NIHR Southampton Biomedical Research Centre and NIHR Southampton Clinical Research Facility, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | - Diane Gbesemete
- Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
- NIHR Southampton Biomedical Research Centre and NIHR Southampton Clinical Research Facility, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | - Adam P Dale
- Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
- NIHR Southampton Biomedical Research Centre and NIHR Southampton Clinical Research Facility, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | - Zoe C Pounce
- Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
| | - Carl N Webb
- Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
- NIHR Southampton Biomedical Research Centre and NIHR Southampton Clinical Research Facility, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | - Eleanor F Roche
- Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
| | - Jonathan M Guy
- Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
| | - Graham Berreen
- Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
| | - Konstantinos Belogiannis
- Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
| | - Alison R Hill
- Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
- NIHR Southampton Biomedical Research Centre and NIHR Southampton Clinical Research Facility, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | - Muktar M Ibrahim
- Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
| | - Muhammad Ahmed
- Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
| | - David W Cleary
- Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
- NIHR Southampton Biomedical Research Centre and NIHR Southampton Clinical Research Facility, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | - Anish K Pandey
- Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
- NIHR Southampton Biomedical Research Centre and NIHR Southampton Clinical Research Facility, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | | | - Lauren Allen
- Public Health England, Porton Down, Salisbury SP4 0JG, UK
| | - Hans de Graaf
- Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
- NIHR Southampton Biomedical Research Centre and NIHR Southampton Clinical Research Facility, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | - Martin C Maiden
- Department of Zoology, University of Oxford, Oxford OX1 3SZ, UK
| | - Saul N Faust
- Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
- NIHR Southampton Biomedical Research Centre and NIHR Southampton Clinical Research Facility, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | | | - Robert C Read
- Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
- NIHR Southampton Biomedical Research Centre and NIHR Southampton Clinical Research Facility, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| |
Collapse
|
28
|
Østergaard L, Vesikari T, Senders SD, Flodmark CE, Kosina P, Jiang HQ, Maguire JD, Absalon J, Jansen KU, Harris SL, Maansson R, Balmer P, Beeslaar J, Perez JL. Persistence of hSBA titers elicited by the meningococcal serogroup B vaccine menB-FHbp for up to 4 years after a 2- or 3-dose primary series and immunogenicity, safety, and tolerability of a booster dose through 26 months. Vaccine 2021; 39:4545-4554. [PMID: 34215452 DOI: 10.1016/j.vaccine.2021.06.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 06/01/2021] [Accepted: 06/02/2021] [Indexed: 11/24/2022]
Abstract
BACKGROUND To demonstrate extended protection against meningococcal serogroup B (MenB) disease after MenB-FHbp (bivalent rLP2086) vaccination, this study evaluated immunopersistence through 26 months following MenB-FHbp boosting after 2 or 3 primary doses in adolescents. STUDY DESIGN This phase 3, open-label study was an extension of 3 phase 2 studies with participants aged 11-18 years randomized to receive primary MenB-FHbp vaccination following 1 of 5 dosing schedules or control. A booster dose was administered 48 months after the primary series. Immunopersistence through 48 months after the last primary dose (persistence stage) and 26 months postbooster (booster stage) was determined by serum bactericidal assays using human complement (hSBAs) against 4 vaccine-heterologous test strains. Safety evaluations included adverse events (AEs) and local and systemic reactions. RESULTS Overall, 698 and 304 subjects enrolled in the persistence and booster stages, respectively. hSBA titers declined in all groups during 12 months postprimary vaccination, then remained stable through 48 months. One month postbooster, 93.4-100.0% of subjects achieved hSBA titers ≥ lower limit of quantitation against each test strain; percentages at 12 and 26 months postbooster were higher than at similar time points following primary vaccination. Primary and booster MenB-FHbp vaccinations were well tolerated, with ≤ 12.5% of subjects reporting AEs during each stage. The most common local (reported by 84.4-93.8% of subjects) and systemic (68.8-76.6%) reactions to the booster were injection site pain and fatigue and headache, respectively; ≤ 3.7% of subjects reported severe systemic events. CONCLUSION Protective hSBA titers initially declined but were retained by many subjects for 4 years irrespective of primary MenB-FHbp vaccination schedule. Boosting at 48 months after primary vaccination was safe, well tolerated, and induced immune responses indicative of immunological memory that persisted through 26 months. Booster vaccination during late adolescence may prolong protection against MenB disease.
Collapse
Affiliation(s)
- Lars Østergaard
- Department of Infectious Diseases, Aarhus University Hospital, Skejby, Palle Juul-Jensens Blvd 99, 8200 Aarhus N, Denmark.
| | - Timo Vesikari
- Nordic Research Network Ltd, Biokatu 10, 33520 Tampere, Finland
| | - Shelly D Senders
- Senders Pediatrics, 2054 South Green Road, South Euclid, OH, USA
| | - Carl-Erik Flodmark
- Department of Pediatrics, Entrance 108, Skåne University Hospital in Malmö, 205 02 Malmö, Sweden
| | - Pavel Kosina
- Department of Infectious Diseases, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic
| | - Han-Qing Jiang
- Pfizer Vaccine Research and Development, 401 North Middletown Road, Pearl River, NY, USA
| | - Jason D Maguire
- Pfizer Vaccine Research and Development, 401 North Middletown Road, Pearl River, NY, USA
| | - Judith Absalon
- Pfizer Vaccine Research and Development, 401 North Middletown Road, Pearl River, NY, USA
| | - Kathrin U Jansen
- Pfizer Vaccine Research and Development, 401 North Middletown Road, Pearl River, NY, USA
| | - Shannon L Harris
- Pfizer Vaccine Research and Development, 401 North Middletown Road, Pearl River, NY, USA
| | - Roger Maansson
- Pfizer Vaccine Research and Development, 500 Arcola Road, Collegeville, PA, USA
| | - Paul Balmer
- Pfizer Vaccine Medical and Scientific Affairs, 500 Arcola Road, Collegeville, PA, USA
| | - Johannes Beeslaar
- Pfizer UK Vaccine Research and Development, Horizon Building, Honey Lane, Hurley SL6 6RJ, UK
| | - John L Perez
- Pfizer Vaccine Research and Development, 500 Arcola Road, Collegeville, PA, USA
| |
Collapse
|
29
|
Tzanakaki G, Xirogianni A, Tsitsika A, Clark SA, Kesanopoulos K, Bratcher HB, Papandreou A, Rodrigues CMC, Maiden MCJ, Borrow R, Tsolia M. Estimated strain coverage of serogroup B meningococcal vaccines: A retrospective study for disease and carrier strains in Greece (2010-2017). Vaccine 2021; 39:1621-1630. [PMID: 33597116 DOI: 10.1016/j.vaccine.2021.01.073] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 01/07/2021] [Accepted: 01/30/2021] [Indexed: 11/17/2022]
Abstract
Invasive meningococcal disease (IMD) is associated with high case fatality rates and long-term sequelae among survivors. Meningococci belonging to six serogroups (A, B, C, W, X, and Y) cause nearly all IMD worldwide, with serogroup B meningococci (MenB) the predominant cause in many European countries, including Greece (~80% of all IMD). In the absence of protein-conjugate polysaccharide MenB vaccines, two protein-based vaccines are available to prevent MenB IMD in Greece: 4CMenB (Bexsero™, GlaxoSmithKline), available since 2014; and MenB-FHbp, (Trumenba™, Pfizer), since 2018. This study investigated the potential coverage of MenB vaccines in Greece using 107 MenB specimens, collected from 2010 to 2017 (66 IMD isolates and 41 clinical samples identified solely by non-culture PCR), alongside 6 MenB isolates from a carriage study conducted during 2017-2018. All isolates were characterized by multilocus sequence typing (MLST), PorA, and FetA antigen typing. Whole Genome Sequencing (WGS) was performed on 66 isolates to define the sequences of vaccine components factor H-binding protein (fHbp), Neisserial Heparin Binding Antigen (NHBA), and Neisseria adhesin A (NadA). The expression of fHbp was investigated with flow cytometric meningococcal antigen surface expression (MEASURE) assay. The fHbp gene was present in-frame in all isolates tested by WGS and in 41 MenB clinical samples. All three variant families of fHbp peptides were present, with subfamily B peptides (variant 1) occurring in 69.2% and subfamily A in 30.8% of the samples respectively. Sixty three of 66 (95.5%) MenB isolates expressed sufficient fHbp to be susceptible to bactericidal killing by MenB-fHbp induced antibodies, highlighting its potential to protect against most IMD in Greece.
Collapse
Affiliation(s)
- G Tzanakaki
- National Meningitis Reference Laboratory (NMRL), Dept of Public Health Policy, School of Public Health, University of West Attica, Athens, Greece.
| | - A Xirogianni
- National Meningitis Reference Laboratory (NMRL), Dept of Public Health Policy, School of Public Health, University of West Attica, Athens, Greece
| | - A Tsitsika
- Second Dept of Paediatrics, Medical School, National Kapodistrian University, Athens, Greece
| | - S A Clark
- Meningococcal Reference Unit, Public Health England, Manchester Royal Infirmary, Manchester, UK
| | - K Kesanopoulos
- National Meningitis Reference Laboratory (NMRL), Dept of Public Health Policy, School of Public Health, University of West Attica, Athens, Greece
| | - H B Bratcher
- Department of Zoology, Peter Medawar Building for Pathogen Research, University of Oxford, South Parks Road, Oxford, UK
| | - A Papandreou
- National Meningitis Reference Laboratory (NMRL), Dept of Public Health Policy, School of Public Health, University of West Attica, Athens, Greece
| | - C M C Rodrigues
- Department of Zoology, Peter Medawar Building for Pathogen Research, University of Oxford, South Parks Road, Oxford, UK
| | - M C J Maiden
- Department of Zoology, Peter Medawar Building for Pathogen Research, University of Oxford, South Parks Road, Oxford, UK
| | - R Borrow
- Meningococcal Reference Unit, Public Health England, Manchester Royal Infirmary, Manchester, UK
| | - M Tsolia
- Second Dept of Paediatrics, Medical School, National Kapodistrian University, Athens, Greece
| |
Collapse
|
30
|
Meningococcal Deduced Vaccine Antigen Reactivity (MenDeVAR) Index: a Rapid and Accessible Tool That Exploits Genomic Data in Public Health and Clinical Microbiology Applications. J Clin Microbiol 2020; 59:JCM.02161-20. [PMID: 33055180 PMCID: PMC7771438 DOI: 10.1128/jcm.02161-20] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 10/09/2020] [Indexed: 12/24/2022] Open
Abstract
As microbial genomics makes increasingly important contributions to clinical and public health microbiology, the interpretation of whole-genome sequence data by nonspecialists becomes essential. In the absence of capsule-based vaccines, two protein-based vaccines have been used for the prevention of invasive serogroup B meningococcal disease (IMD) since their licensure in 2013 and 2014. These vaccines have different components and different levels of coverage of meningococcal variants. Hence, decisions regarding which vaccine to use in managing serogroup B IMD outbreaks require information about the index case isolate, including (i) the presence of particular vaccine antigen variants, (ii) the expression of vaccine antigens, and (iii) the likely susceptibility of its antigen variants to antibody-dependent bactericidal killing. As microbial genomics makes increasingly important contributions to clinical and public health microbiology, the interpretation of whole-genome sequence data by nonspecialists becomes essential. In the absence of capsule-based vaccines, two protein-based vaccines have been used for the prevention of invasive serogroup B meningococcal disease (IMD) since their licensure in 2013 and 2014. These vaccines have different components and different levels of coverage of meningococcal variants. Hence, decisions regarding which vaccine to use in managing serogroup B IMD outbreaks require information about the index case isolate, including (i) the presence of particular vaccine antigen variants, (ii) the expression of vaccine antigens, and (iii) the likely susceptibility of its antigen variants to antibody-dependent bactericidal killing. To obtain this information requires a multitude of laboratory assays, impractical in real-time clinical settings, where the information is most urgently needed. To facilitate assessment for public health and clinical purposes, we synthesized genomic and experimental data from published sources to develop and implement the Meningococcal Deduced Vaccine Antigen Reactivity (MenDeVAR) Index, which is publicly available on PubMLST (https://pubmlst.org). Using whole-genome sequences or individual gene sequences obtained from IMD isolates or clinical specimens, the MenDeVAR Index provides rapid evidence-based information on the presence and possible immunological cross-reactivity of different meningococcal vaccine antigen variants. The MenDeVAR Index enables practitioners who are not genomics specialists to assess the likely reactivity of vaccines for individual cases, outbreak management, or the assessment of public health vaccine programs. The MenDeVAR Index has been developed in consultation with, but independently of, both the 4CMenB (Bexsero; GSK) and rLP2086 (Trumenba; Pfizer, Inc.) vaccine manufacturers.
Collapse
|
31
|
Findlow J, Bayliss CD, Beernink PT, Borrow R, Liberator P, Balmer P. Broad vaccine protection against Neisseria meningitidis using factor H binding protein. Vaccine 2020; 38:7716-7727. [PMID: 32878710 PMCID: PMC8082720 DOI: 10.1016/j.vaccine.2020.08.031] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 07/27/2020] [Accepted: 08/12/2020] [Indexed: 11/29/2022]
Abstract
Neisseria meningitidis, the causative agent of invasive meningococcal disease (IMD), is classified into different serogroups defined by their polysaccharide capsules. Meningococcal serogroups A, B, C, W, and Y are responsible for most IMD cases, with serogroup B (MenB) causing a substantial percentage of IMD cases in many regions. Vaccines using capsular polysaccharides conjugated to carrier proteins have been successfully developed for serogroups A, C, W, and Y. However, because the MenB capsular polysaccharide is poorly immunogenic, MenB vaccine development has focused on alternative antigens. The 2 currently available MenB vaccines (MenB-4C and MenB-FHbp) both include factor H binding protein (FHbp), a surface-exposed protein harboured by nearly all meningococcal isolates that is important for survival of the bacteria in human blood. MenB-4C contains a nonlipidated FHbp from subfamily B in addition to other antigens, including Neisserial Heparin Binding Antigen, Neisserial adhesin A, and outer membrane vesicles, whereas MenB-FHbp contains a lipidated FHbp from each subfamily (A and B). FHbp is highly immunogenic and a main target of bactericidal activity of antibodies elicited by both licensed MenB vaccines. FHbp is also an important vaccine component, in contrast to some other meningococcal antigens that may have limited cross-protection across strains, as FHbp-specific antibodies can provide broad cross-protection within each subfamily. Limited cross-protection between subfamilies necessitates the inclusion of FHbp variants from both subfamilies to achieve broad FHbp-based vaccine coverage. Additionally, immune responses to the lipidated form of FHbp have a superior cross-reactive profile to those elicited by the nonlipidated form. Taken together, the inclusion of lipidated FHbp variants from both FHbp subfamilies is expected to provide broad protection against the diverse disease-causing meningococcal strains expressing a wide range of FHbp sequence variants. This review describes the development of vaccines for MenB disease prevention, with a focus on the FHbp antigen.
Collapse
Affiliation(s)
- Jamie Findlow
- Vaccine Medical Development, Scientific and Clinical Affairs, Pfizer Ltd, Tadworth, UK.
| | | | - Peter T Beernink
- Department of Pediatrics, School of Medicine, University of California, San Francisco, San Francisco, CA, USA.
| | - Ray Borrow
- Public Health England, Manchester Royal Infirmary, Manchester, UK.
| | - Paul Liberator
- Vaccine Research and Development, Pfizer Inc, Pearl River, NY, USA.
| | - Paul Balmer
- Vaccine Medical Development, Scientific and Clinical Affairs, Pfizer Inc, Collegeville, PA, USA.
| |
Collapse
|
32
|
Genomic Characterization of Invasive Meningococcal Serogroup B Isolates and Estimation of 4CMenB Vaccine Coverage in Finland. mSphere 2020; 5:5/5/e00376-20. [PMID: 32938694 PMCID: PMC7494829 DOI: 10.1128/msphere.00376-20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Invasive meningococcal disease (IMD) caused by Neisseria meningitidis is a significant cause of morbidity and mortality worldwide. In Finland, the incidence rate of IMD is low, with meningococcal serogroup B (MenB) accounting for around one-third of IMD cases annually. The aim of this study was to investigate the genetic variability of invasive MenB isolates collected in Finland between 2010 and 2017 (n = 81), including the genes encoding the 4-component MenB vaccine (4CMenB; Bexsero; GSK) antigens and their promoters, and to evaluate the 4CMenB potential coverage. Whole-genome sequencing was performed. The meningococcal antigen typing system (MATS) was used to characterize MenB isolates and predict the potential coverage of 4CMenB. MATS was complemented by genetic MATS (gMATS) through association of antigen genotyping and phenotypic MATS results. Multilocus sequence typing revealed predominance of the ST-41/44 clonal complex among which sequence type (ST)-303 was the most common and was predicted to be covered by 4CMenB. Of the 4 major vaccine antigens, the factor H-binding protein variant 1, neisserial heparin binding antigen peptide 2, and the PorA P1.4 antigen were predominant, whereas Neisseria adhesin A was present in only 4% of the 81 isolates. MATS and gMATS 4CMenB strain coverage predictions were 78% and 86%, respectively, in a subpanel of 60 isolates collected during 2010 to 2014, with a gMATS prediction of 84% for all 81 isolates. The results suggest that 4CMenB could reduce the burden of IMD in Finland and that gMATS could be applied to monitor vaccine strain coverage and predict vaccine effectiveness.IMPORTANCE 4CMenB is a 4-component vaccine used against invasive meningococcal disease (IMD) caused by Neisseria meningitidis serogroup B (MenB). We investigated the genetic variability of MenB in Finland and evaluated 4CMenB strain coverage by 2 different methods: MATS (meningococcal antigen typing system) and gMATS (genetic MATS). In a set of MenB isolates, 78% (MATS) and 86% (gMATS) were predicted as covered by 4CMenB, suggesting that use of 4CMenB would help reduce IMD incidence in Finland. MATS has been used in 13 countries worldwide, generating information on phenotypic characteristics that could infer protection by 4CMenB. Based on these data and genetic information, gMATS coverage predictions can be made. gMATS predicts coverage consistent with MATS for about 94% of tested strains. Unlike MATS, gMATS does not require live isolates, thus allowing the analysis also of noncultivable strains, making the coverage predictions more accurate. Therefore, gMATS can replace MATS to assess 4CMenB coverage, including in regions with no prior MATS data.
Collapse
|
33
|
Beeslaar J, Absalon J, Anderson AS, Eiden JJ, Balmer P, Harris SL, Jones TR, O'Neill RE, Pregaldien JL, Radley D, Maansson R, Ginis J, Srivastava A, Perez JL. MenB-FHbp Vaccine Protects Against Diverse Meningococcal Strains in Adolescents and Young Adults: Post Hoc Analysis of Two Phase 3 Studies. Infect Dis Ther 2020; 9:641-656. [PMID: 32700260 PMCID: PMC7452968 DOI: 10.1007/s40121-020-00319-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Indexed: 11/17/2022] Open
Abstract
Introduction Two phase 3 studies in adolescents and young adults demonstrated that MenB-FHbp, a meningococcal serogroup B (MenB) vaccine, elicits protective immune responses after 2 or 3 doses based on serum bactericidal antibody assays using human complement (hSBA) against 4 primary and 10 additional diverse, vaccine-heterologous MenB test strains. Lower limits of quantitation (LLOQs; titers 1:8 or 1:16; titers ≥ 1:4 correlate with protection) were used to evaluate responses to individual strains and all 4 primary strains combined (composite response). A post hoc analysis evaluated percentages of subjects with protective responses to as many as 8 strains combined (4 primary plus additional strains). Methods Immune responses were measured using hSBAs against 4 primary strains in adolescents (n = 1509, MenB-FHbp; n = 898, hepatitis A virus vaccine/saline) and young adults (n = 2480, MenB-FHbp; n = 824, saline) receiving MenB-FHbp or control at 0, 2, and 6 months. Ten additional strains were evaluated in subsets of subjects from approximately 1800 MenB-FHbp recipients across both studies. Percentages of subjects with hSBA titers ≥ LLOQ for different numbers of primary strains or primary plus additional strains combined (7 or 8 strains total per subset) were determined before vaccination, 1 month post-dose 2, and 1 month post-dose 3. Results Across the panel of primary plus additional strains, at 1 month post-dose 3, titers ≥ LLOQ were elicited in 93.7–95.7% of adolescents and 91.7–95.0% of young adults for ≥ 5 test strains combined and in 70.5–85.8% of adolescents and 67.5–81.4% of young adults for ≥ 7 strains combined. Among adolescents, 99.8%, 99.0%, 92.8%, and 82.7% had titers ≥ LLOQ against at least 1, 2, 3, and all 4 primary strains, respectively; corresponding percentages for young adults were 99.7%, 97.7%, 94.0%, and 84.5%. Conclusions Results support the ability of MenB-FHbp to provide broad coverage against MenB strains expressing diverse FHbp variants. Trial Registration ClinicalTrials.gov identifiers NCT01830855, NCT01352845. Electronic supplementary material The online version of this article (10.1007/s40121-020-00319-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Johannes Beeslaar
- Vaccine Clinical Research and Development, Pfizer Ltd UK, Hurley, UK.
| | - Judith Absalon
- Vaccine Clinical Research and Development, Pfizer Inc, Pearl River, NY, USA
| | | | - Joseph J Eiden
- Vaccine Clinical Research and Development, Pfizer Inc, Pearl River, NY, USA
| | - Paul Balmer
- Vaccine Medical Development, Scientific and Clinical Affairs, Pfizer Inc, Collegeville, PA, USA
| | - Shannon L Harris
- Vaccine Research and Development, Pfizer Inc, Pearl River, NY, USA
| | - Thomas R Jones
- Vaccine Research and Development, Pfizer Inc, Pearl River, NY, USA
| | - Robert E O'Neill
- Vaccine Research and Development, Pfizer Inc, Pearl River, NY, USA
| | | | - David Radley
- Vaccine Clinical Research and Development, Pfizer Inc, Pearl River, NY, USA
| | - Roger Maansson
- Vaccine Clinical Research and Development, Pfizer Inc, Collegeville, PA, USA
| | - John Ginis
- Vaccine Research and Development, Pfizer Inc, Collegeville, PA, USA
| | - Amit Srivastava
- Vaccine Medical Development, Scientific and Clinical Affairs, Pfizer Inc, Collegeville, PA, USA
| | - John L Perez
- Vaccine Clinical Research and Development, Pfizer Inc, Collegeville, PA, USA
| |
Collapse
|
34
|
Intra-Laboratory Evaluation of Luminescence Based High-Throughput Serum Bactericidal Assay (L-SBA) to Determine Bactericidal Activity of Human Sera against Shigella. High Throughput 2020; 9:ht9020014. [PMID: 32521658 PMCID: PMC7361673 DOI: 10.3390/ht9020014] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 06/01/2020] [Accepted: 06/04/2020] [Indexed: 12/31/2022] Open
Abstract
Despite the huge decrease in deaths caused by Shigella worldwide in recent decades, shigellosis still causes over 200,000 deaths every year. No vaccine is currently available, and the morbidity of the disease coupled with the rise of antimicrobial resistance renders the introduction of an effective vaccine extremely urgent. Although a clear immune correlate of protection against shigellosis has not yet been established, the demonstration of the bactericidal activity of antibodies induced upon vaccination may provide one means of the functionality of antibodies induced in protecting against Shigella. The method of choice to evaluate the complement-mediated functional activity of vaccine-induced antibodies is the Serum Bactericidal Assay (SBA). Here we present the development and intra-laboratory characterization of a high-throughput luminescence-based SBA (L-SBA) method, based on the detection of ATP as a proxy of surviving bacteria, to evaluate the complement-mediated killing of human sera. We demonstrated the high specificity of the assay against a homologous strain without any heterologous aspecificity detected against species-related and non-species-related strains. We assessed the linearity, repeatability and reproducibility of L-SBA on human sera. This work will guide the bactericidal activity assessment of clinical sera raised against S. sonnei. The method has the potential of being applicable with similar performances to determine the bactericidal activity of any non-clinical and clinical sera that rely on complement-mediated killing.
Collapse
|
35
|
Balmer P, Beeslaar J, Findlow J, Srivastava A. Understanding immunogenicity assessments for meningococcal serogroup B vaccines. Postgrad Med 2020; 132:184-191. [PMID: 32124678 DOI: 10.1080/00325481.2019.1696582] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Invasive meningococcal disease (IMD) is a potentially devastating infection associated with high mortality and long-term sequelae; however, vaccines are available to protect against the five common disease-causing serogroups (A, B, C, W, and Y). Because traditional field efficacy clinical trials were not feasible due to low IMD incidence that necessitates a very large number of participants, serum bactericidal antibody (SBA) assays using rabbit (rSBA) or human (hSBA) complement were established as in vitro surrogates of meningococcal vaccine efficacy and are now routinely used to support vaccine licensure. Specifically, rSBA assays have been used to evaluate responses to meningococcal capsular polysaccharide-protein conjugate vaccines against serogroups A, C, W, and Y; the accepted correlate of protection for rSBA assays is a titer ≥1:8. Importantly, because the bacterial capsular polysaccharide antigen is conserved across strains, only one test strain that expresses an invariant polysaccharide capsule for each serogroup is required to assess coverage. rSBA assays are unsuitable for subcapsular protein-based serogroup B (MenB) vaccines, and therefore, hSBA assays have been used for licensure; titers ≥1:4 are considered the correlate of protection against IMD for hSBA. In contrast to MenACWY vaccines, because bacterial surface proteins are antigenically variable, MenB vaccines must be tested with hSBA assays using multiple test strains that represent the antigenic diversity of disease-causing isolates. As this complexity regarding SBA assessment methods can make data interpretation difficult, herein we describe the use of hSBA assays to evaluate MenB vaccine efficacy and to support licensure. In addition, we highlight how the two recently approved MenB vaccines differ in their use of hSBA assays in clinical studies to demonstrate broad protection against MenB IMD.
Collapse
Affiliation(s)
- Paul Balmer
- Vaccine Medical Development, Scientific & Clinical Affairs, Pfizer Inc, Collegeville, PA, USA
| | | | - Jamie Findlow
- Vaccine Medical & Scientific Affairs, Pfizer Ltd, Tadworth, UK
| | - Amit Srivastava
- Vaccine Medical Development, Scientific & Clinical Affairs, Pfizer Inc, Collegeville, PA, USA
| |
Collapse
|
36
|
Adsorption onto aluminum hydroxide adjuvant protects antigens from degradation. Vaccine 2020; 38:3600-3609. [PMID: 32063436 DOI: 10.1016/j.vaccine.2020.02.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 01/31/2020] [Accepted: 02/02/2020] [Indexed: 11/24/2022]
Abstract
Aluminum based adjuvants are widely used in commercial vaccines, since they are known to be safe and effective with a variety of antigens. The effect of antigen adsorption onto Aluminum Hydroxide is a complex area, since several mechanisms are involved simultaneously, whose impact is both antigen and formulation conditions dependent. Moreover, the mode of action of Aluminum Hydroxide is itself complex, with many mechanisms operating simultaneously. Within the literature there are contrasting theories regarding the effect of adsorption on antigen integrity and stability, with reports of antigen being stabilized by adsorption onto Aluminum Hydroxide, but also with contrary reports of antigen being destabilized. With the aim to understand the impact of adsorption on three recombinant proteins which, following in vivo immunization, are able to induce functional bactericidal antibodies against Neisseria meningitidis type B, we used a range of physico-chemical tools, such as DSC and UPLC, along with in vitro binding of antibodies that recognize structural elements of the proteins, and supported the in vitro data with in vivo evaluation in mice studies. We showed that, following exposure to accelerated degradation conditions involving heat, the recombinant proteins, although robust, were stabilized by adsorption onto Aluminum Hydroxide and retain their structural integrity unlike the not adsorbed proteins. The measure of the Melting Temperature was a useful tool to compare the behavior of proteins adsorbed and not adsorbed on Aluminum Hydroxide and to predict protein stability.
Collapse
|
37
|
Harris SL, Tan C, Perez J, Radley D, Jansen KU, Anderson AS, Jones TR. Selection of diverse strains to assess broad coverage of the bivalent FHbp meningococcal B vaccine. NPJ Vaccines 2020; 5:8. [PMID: 32025339 PMCID: PMC6989502 DOI: 10.1038/s41541-019-0154-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 11/20/2019] [Indexed: 01/14/2023] Open
Abstract
MenB-FHbp is a recombinant meningococcal serogroup B (MenB) vaccine composed of 2 factor H binding proteins (FHbps). Meningococcal vaccines targeting polysaccharide serogroup A, C, Y, and W capsules were licensed upon confirmation of bactericidal antibody induction after initial efficacy studies with serogroup A and C vaccines. Unlike meningococcal polysaccharide vaccines, wherein single strains demonstrated bactericidal antibodies per serogroup for each vaccine, MenB-FHbp required a more robust approach to demonstrate that bactericidal antibody induction could kill strains with diverse FHbp sequences. Serum bactericidal assays using human complement were developed for 14 MenB strains, representing breadth of meningococcal FHbp diversity of ~80% of circulating MenB strains. This work represents an innovative approach to license a non-toxin protein vaccine with 2 antigens representing a single virulence factor by an immune correlate, and uniquely demonstrates that such a vaccine provides coverage across bacterial strains by inducing broadly protective antibodies. Neisseria meningitidis is an important cause of invasive meningococcal disease, effective vaccines exist for some serogroups but immunogenicity to the MenB group is poor. Thomas R. Jones and colleagues examine serum bactericidal responses from volunteers challenged with MenB-FHbp – a recombinant MenB vaccine containing two Factor H (FH)-binding proteins. Serum bactericidal responses are tested against 14 MenB clinical isolates selected in an unbiased manner to cover the vast breadth of FHbp antigen and epidemiological diversity. This work demonstrates the broad efficacy of the MenB-FHbp vaccine using a serum bactericidal activity as a surrogate of protection.
Collapse
Affiliation(s)
| | - Cuiwen Tan
- Pfizer Vaccine Research and Development, Pearl River, NY USA
| | - John Perez
- 2Pfizer Vaccine Research and Development, Collegeville, PA USA
| | - David Radley
- 2Pfizer Vaccine Research and Development, Collegeville, PA USA
| | | | | | - Thomas R Jones
- Pfizer Vaccine Research and Development, Pearl River, NY USA
| |
Collapse
|
38
|
Kurtovic L, Boyle MJ, Opi DH, Kennedy AT, Tham WH, Reiling L, Chan JA, Beeson JG. Complement in malaria immunity and vaccines. Immunol Rev 2019; 293:38-56. [PMID: 31556468 PMCID: PMC6972673 DOI: 10.1111/imr.12802] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 09/03/2019] [Indexed: 12/20/2022]
Abstract
Developing efficacious vaccines for human malaria caused by Plasmodium falciparum is a major global health priority, although this has proven to be immensely challenging over the decades. One major hindrance is the incomplete understanding of specific immune responses that confer protection against disease and/or infection. While antibodies to play a crucial role in malaria immunity, the functional mechanisms of these antibodies remain unclear as most research has primarily focused on the direct inhibitory or neutralizing activity of antibodies. Recently, there is a growing body of evidence that antibodies can also mediate effector functions through activating the complement system against multiple developmental stages of the parasite life cycle. These antibody‐complement interactions can have detrimental consequences to parasite function and viability, and have been significantly associated with protection against clinical malaria in naturally acquired immunity, and emerging findings suggest these mechanisms could contribute to vaccine‐induced immunity. In order to develop highly efficacious vaccines, strategies are needed that prioritize the induction of antibodies with enhanced functional activity, including the ability to activate complement. Here we review the role of complement in acquired immunity to malaria, and provide insights into how this knowledge could be used to harness complement in malaria vaccine development.
Collapse
Affiliation(s)
- Liriye Kurtovic
- Burnet Institute, Melbourne, Vic., Australia.,Central Clinical School, Monash University, Melbourne, Vic., Australia
| | | | | | - Alexander T Kennedy
- Walter and Eliza Hall Institute, Melbourne, Vic., Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Vic., Australia
| | - Wai-Hong Tham
- Walter and Eliza Hall Institute, Melbourne, Vic., Australia
| | | | - Jo-Anne Chan
- Burnet Institute, Melbourne, Vic., Australia.,Central Clinical School, Monash University, Melbourne, Vic., Australia
| | - James G Beeson
- Burnet Institute, Melbourne, Vic., Australia.,Central Clinical School, Monash University, Melbourne, Vic., Australia.,Department of Microbiology, Monash University, Clayton, Vic., Australia.,Department of Medicine, The University of Melbourne, Parkville, Vic., Australia
| |
Collapse
|
39
|
Ndungo E, Pasetti MF. Functional antibodies as immunological endpoints to evaluate protective immunity against Shigella. Hum Vaccin Immunother 2019; 16:197-205. [PMID: 31287754 PMCID: PMC7670857 DOI: 10.1080/21645515.2019.1640427] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The development, clinical advancement and licensure of vaccines, and monitoring of vaccine effectiveness could be expedited and simplified by the ability to measure immunological endpoints that can predict a favorable clinical outcome. Antigen-specific and functional antibodies have been described in the context of naturally acquired immunity and vaccination against Shigella, and their presence in serum has been associated with reduced risk of disease in human subjects. The relevance of these antibodies as correlates of protective immunity, their mechanistic contribution to protection (e.g. target antigens, interference with pathogenesis, and participation in microbial clearance), and factors that influence their magnitude and makeup (e.g. host age, health condition, and environment) are important considerations that need to be explored. In addition to facilitating vaccine evaluation, immunological correlates of protection could be useful for identifying groups at risk and advancing immune therapies. Herein we discuss the precedent and value of functional antibodies as immunological endpoints to predict vaccine efficacy and the relevance of functional antibody activity to evaluate protective immunity against shigellosis.
Collapse
Affiliation(s)
- Esther Ndungo
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Marcela F Pasetti
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
40
|
Martinón-Torres F, Nolan T, Toneatto D, Banzhoff A. Persistence of the immune response after 4CMenB vaccination, and the response to an additional booster dose in infants, children, adolescents, and young adults. Hum Vaccin Immunother 2019; 15:2940-2951. [PMID: 31246520 PMCID: PMC6930112 DOI: 10.1080/21645515.2019.1627159] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
The multicomponent meningococcal serogroup B vaccine, 4CMenB, has demonstrated effectiveness in preventing invasive MenB disease in infants and in controlling MenB outbreaks. The need for/timing of additional booster doses is not yet established. We reviewed eight studies that evaluated antibody persistence and booster following primary 4CMenB vaccination of infants, children, adolescents, and young adults. Putative seroprotective hSBA titers for ≥1 vaccine antigen were maintained by 76-100% of children 24-36 months after priming during infancy and in 84-100% after priming in the second year of life. hSBA levels were higher in vaccinees at 4 and 7.5 years following priming during adolescence than in vaccine-naïve individuals of a similar age. Antibodies persisted at higher levels to NHBA and NadA than to PorA or fHbp. Booster vaccination induced robust anamnestic responses, demonstrating effective priming by 4CMenB across age-groups. These data can inform decision-making to optimize vaccination strategies.
Collapse
Affiliation(s)
- Federico Martinón-Torres
- Translational Pediatrics and Infectious Diseases Section, Pediatrics Department, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain
| | - Terry Nolan
- School of Population and Global Health, The University of Melbourne, and Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | | | | |
Collapse
|
41
|
Lujan E, Winter K, Rovaris J, Liu Q, Granoff DM. Serum Bactericidal Antibody Responses of Students Immunized With a Meningococcal Serogroup B Vaccine in Response to an Outbreak on a University Campus. Clin Infect Dis 2019; 65:1112-1119. [PMID: 28582542 DOI: 10.1093/cid/cix519] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 06/01/2017] [Indexed: 12/12/2022] Open
Abstract
Background MenB-4C is a recently licensed meningococcal serogroup B vaccine. For vaccine licensure, short-term efficacy was inferred from serum bactericidal antibody (SBA) titers against 3 antigen-specific indicator strains, which are not necessarily representative of US disease-causing strains. Methods A total of 4923 students were immunized with MenB-4C in response to an outbreak at a university. Serum samples were obtained at 1.5-2 months from 106 students who received the recommended 2 doses and 52 unvaccinated students. Follow-up serum samples were obtained at 7 months from 42 vaccinated and 24 unvaccinated participants. SBA was measured against strains from 4 university outbreaks. Results At 1.5-2 months, the proportion of immunized students with protective titers ≥1:4 against an isolate from the campus outbreak was 93% (95% confidence interval [CI], 87%-97%) vs 37% (95% CI, 24%-51%) in unvaccinated students. The proportion with protective titers against strains from 3 other university outbreaks was 73% (95% CI, 62%-82%) vs 26% (95% CI, 14%-41%) in unvaccinated; 71% (95% CI, 61%-79%) vs 19% (95% CI, 10%-33%) in unvaccinated; and 53% (95% CI, 42%-64%) vs 9% (95% CI, 3%-22%) in unvaccinated (P < .0001 for each strain). At 7 months, the proportion of immunized students with titers ≥1:4 was 86% (95% CI, 71%-95%) against the isolate from the campus outbreak and 57% (95% CI, 41%-72%), 38% (95% CI, 24%-54%), and 31% (95% CI, 18%-47%), respectively, for the other 3 outbreak strains. Conclusions MenB-4C elicited short-term protective titers against 4 strains responsible for recent university campus outbreaks. By 7 months the prevalence of protective titers was <40% for 2 of the 4 outbreak strains. A booster dose of MenB-4C may be needed to maintain protective titers.
Collapse
Affiliation(s)
- Eduardo Lujan
- Center for Immunobiology and Vaccine Development, UCSF Benioff Children's Hospital Oakland
| | - Kathleen Winter
- Immunization Branch, California Department of Public Health, Richmond, and
| | | | - Qin Liu
- Wistar Institute, Philadelphia, Pennsylvania
| | - Dan M Granoff
- Center for Immunobiology and Vaccine Development, UCSF Benioff Children's Hospital Oakland
| |
Collapse
|
42
|
Peschiera I, Giuliani M, Giusti F, Melero R, Paccagnini E, Donnarumma D, Pansegrau W, Carazo JM, Sorzano COS, Scarselli M, Masignani V, Liljeroos LJ, Ferlenghi I. Structural basis for cooperativity of human monoclonal antibodies to meningococcal factor H-binding protein. Commun Biol 2019; 2:241. [PMID: 31263785 PMCID: PMC6595007 DOI: 10.1038/s42003-019-0493-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 05/29/2019] [Indexed: 11/23/2022] Open
Abstract
Monoclonal antibody (mAb) cooperativity is a phenomenon triggered when mAbs couples promote increased bactericidal killing compared to individual partners. Cooperativity has been deeply investigated among mAbs elicited by factor H-binding protein (fHbp), a Neisseria meningitidis surface-exposed lipoprotein and one of the key antigens included in both serogroup B meningococcus vaccine Bexsero and Trumenba. Here we report the structural and functional characterization of two cooperative mAbs pairs isolated from Bexsero vaccines. The 3D electron microscopy structures of the human mAb-fHbp-mAb cooperative complexes indicate that the angle formed between the antigen binding fragments (fAbs) assume regular angle and that fHbp is able to bind simultaneously and stably the cooperative mAbs pairs and human factor H (fH) in vitro. These findings shed light on molecular basis of the antibody-based mechanism of protection driven by simultaneous recognition of the different epitopes of the fHbp and underline that cooperativity is crucial in vaccine efficacy.
Collapse
|
43
|
McDaniel A, Dempsey A, Srivastava A. A physician's guide to the 2-dose schedule of MenB-FHbp vaccine. Hum Vaccin Immunother 2019; 15:2729-2737. [PMID: 30932730 PMCID: PMC6930067 DOI: 10.1080/21645515.2019.1596711] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 02/27/2019] [Accepted: 03/07/2019] [Indexed: 11/24/2022] Open
Abstract
Meningococcal serogroup B (MenB) is the predominant cause of invasive meningococcal disease in the United States, with older adolescents and young adults attending college at increased risk. Notably, MenB caused all meningococcal disease outbreaks at US colleges between 2011 and 2018. MenB disease is vaccine-preventable. The MenB-FHbp vaccine can be administered on a 2-dose (0 and 6 months) schedule to healthy adolescents and young adults or as a tailored 3-dose (0, 1-2, and 6 months) schedule for individuals at increased risk. This review focuses on the 2-dose schedule (0 and 6 months) of MenB-FHbp. Clinical evidence demonstrating strong and broadly protective immunogenicity in adolescents after primary vaccination, immune persistence up to 48 months post-primary vaccination (18-61% of subjects across schedules), and immune memory evidenced by robust response to a single booster dose are described. Implementation approaches to ensure adolescents and young adults are fully vaccinated against meningococcal disease are discussed.
Collapse
Affiliation(s)
- Angee McDaniel
- Medical Development, Scientific & Clinical Affairs, Pfizer Vaccines, Pfizer Inc, Collegeville, PA, USA
| | - Amanda Dempsey
- University of Colorado Denver, Anschutz Medical Campus, Denver, CO, USA
| | - Amit Srivastava
- Medical Development, Scientific & Clinical Affairs, Pfizer Vaccines, Pfizer Inc, Cambridge, MA, USA
| |
Collapse
|
44
|
Findlow J, Balmer P, Borrow R. A review of complement sources used in serum bactericidal assays for evaluating immune responses to meningococcal ACWY conjugate vaccines. Hum Vaccin Immunother 2019; 15:2491-2500. [PMID: 30883271 PMCID: PMC6816443 DOI: 10.1080/21645515.2019.1593082] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Invasive meningococcal disease is rare and potentially devastating but often vaccine-preventable. Evaluation of meningococcal vaccine effectiveness is impractical owing to relatively low disease incidence; protection is therefore estimated using serum bactericidal antibody (SBA) assays. Original experiments on natural immunity established a titer of ≥4 as the correlate of protection for SBA assays using human complement (hSBA), but human complement is relatively difficult to obtain and standardize. Use of baby rabbit complement (rSBA assays), per standard guidelines for serogroups A and C, generally results in comparatively higher titers. Postlicensure effectiveness data for serogroup C conjugate vaccines support acceptance of rSBA titers ≥8 as the correlate of protection for this serogroup, but no thresholds have been formally established for serogroups A, W, and Y. Studies evaluating MenACWY-TT (Nimenrix®; Pfizer Inc, Sandwich, UK) immunogenicity have used both hSBA and rSBA assays, and ultimately suggest that rSBA may be more appropriate for these measurements.
Collapse
Affiliation(s)
- Jamie Findlow
- Vaccines, Medical and Scientific Affairs, International Developed Markets, Pfizer Ltd , Surrey , UK
| | - Paul Balmer
- Vaccine Medical and Scientific Affairs, Pfizer Inc , Collegeville , PA , USA
| | - Ray Borrow
- Public Health England, Manchester Royal Infirmary , Manchester , UK
| |
Collapse
|
45
|
Alderfer J, Srivastava A, Isturiz R, Burman C, Absalon J, Beeslaar J, Perez J. Concomitant administration of meningococcal vaccines with other vaccines in adolescents and adults: a review of available evidence. Hum Vaccin Immunother 2019; 15:2205-2216. [PMID: 30779683 PMCID: PMC6773407 DOI: 10.1080/21645515.2019.1581542] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Invasive meningococcal disease (IMD), a rapidly progressing and potentially fatal illness, disproportionately affects adolescents and young adults. While IMD is best prevented by vaccination, vaccine uptake in these groups is low. An evidence-based understanding of the safety and effectiveness of concomitant vaccination of meningococcal vaccines, including the newer MenB protein vaccines and the more established MenACWY conjugate vaccines, with other vaccines recommended for adolescents and young adults may help maximize vaccination opportunities. We identified 21 studies assessing concomitant administration of meningococcal vaccines with other vaccines in adolescents and adults. Although studies varied in methodology, concomitant administration generally did not affect immunogenicity of the meningococcal or coadministered vaccines. In some cases, reactogenicity increased following concomitant administration, but no definitive safety concerns were raised. In general, data suggest that meningococcal vaccines can be safely and effectively coadministered with other vaccines.
Collapse
Affiliation(s)
- Justine Alderfer
- Pfizer Vaccines Medical Development & Scientific and Clinical Affairs , Collegeville , PA , USA
| | - Amit Srivastava
- Pfizer Vaccines Medical Development & Scientific and Clinical Affairs , Collegeville , PA , USA
| | - Raul Isturiz
- Pfizer Vaccines Medical Development & Scientific and Clinical Affairs , Collegeville , PA , USA
| | - Cynthia Burman
- Pfizer Vaccines Medical Development & Scientific and Clinical Affairs , Collegeville , PA , USA
| | - Judith Absalon
- Pfizer Vaccine Clinical Research and Development , Pearl River , NY , USA
| | | | - John Perez
- Pfizer Vaccine Clinical Research and Development , Collegeville , PA , USA
| |
Collapse
|
46
|
MenB-FHbp Meningococcal Group B Vaccine (Trumenba ®): A Review in Active Immunization in Individuals Aged ≥ 10 Years. Drugs 2019; 78:257-268. [PMID: 29380290 DOI: 10.1007/s40265-018-0869-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
MenB-FHbp (bivalent rLP2086; Trumenba®) is a recombinant protein-based vaccine targeting Neisseria meningitidis serogroup B (MenB), which has recently been licensed in the EU for active immunization to prevent invasive disease caused by MenB in individuals ≥ 10 years of age. The vaccine, which contains a variant from each of the two identified subfamilies of the meningococcal surface protein factor H-binding protein (fHBP), has been licensed in the USA for active immunization in individuals 10-25 years of age since 2014. This article reviews the immunogenicity, reactogenicity and tolerability of MenB-FHbp, with a focus on the EU label and the European setting. As demonstrated in an extensive program of clinical trials in adolescents and young adults, a two-dose or three-dose series of MenB-FHbp elicits a strong immune response against a range of MenB test strains selected to be representative of strains prevalent in Europe and the USA. Follow-up studies investigating the persistence of the MenB-FHbp immune response and the effect of a booster dose of the vaccine indicate that a booster dose should be considered (following a primary vaccine series) in individuals at continued risk of invasive meningococcal disease. MenB-FHbp vaccine appears to be moderately reactogenic but, overall, is generally well tolerated, with most adverse reactions being mild to moderate in severity. Although post-marketing, population-based data will be required to establish the true effectiveness of the vaccine, currently available data indicate that MenB-FHbp, in a two-dose or three-dose series, is likely to provide broad protection against MenB strains circulating in Europe.
Collapse
|
47
|
Granoff DM, Kim H, Topaz N, MacNeil J, Wang X, McNamara LA. Differential effects of therapeutic complement inhibitors on serum bactericidal activity against non-groupable meningococcal isolates recovered from patients treated with eculizumab. Haematologica 2019; 104:e340-e344. [PMID: 30705094 DOI: 10.3324/haematol.2018.209692] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Affiliation(s)
- Dan M Granoff
- Center for Immunobiology and Vaccine Development, UCSF Benioff Children's Hospital Oakland, Oakland, CA
| | - Howard Kim
- Center for Immunobiology and Vaccine Development, UCSF Benioff Children's Hospital Oakland, Oakland, CA
| | - Nadav Topaz
- Meningitis and Vaccine Preventable Diseases Branch, Division of Bacterial Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Jessica MacNeil
- Meningitis and Vaccine Preventable Diseases Branch, Division of Bacterial Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Xin Wang
- Meningitis and Vaccine Preventable Diseases Branch, Division of Bacterial Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Lucy A McNamara
- Meningitis and Vaccine Preventable Diseases Branch, Division of Bacterial Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| |
Collapse
|
48
|
Muzzi A, Brozzi A, Serino L, Bodini M, Abad R, Caugant D, Comanducci M, Lemos AP, Gorla MC, Křížová P, Mikula C, Mulhall R, Nissen M, Nohynek H, Simões MJ, Skoczyńska A, Stefanelli P, Taha MK, Toropainen M, Tzanakaki G, Vadivelu-Pechai K, Watson P, Vazquez JA, Rajam G, Rappuoli R, Borrow R, Medini D. Genetic Meningococcal Antigen Typing System (gMATS): A genotyping tool that predicts 4CMenB strain coverage worldwide. Vaccine 2019; 37:991-1000. [PMID: 30661831 DOI: 10.1016/j.vaccine.2018.12.061] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 12/17/2018] [Accepted: 12/20/2018] [Indexed: 02/02/2023]
Abstract
BACKGROUND The Meningococcal Antigen Typing System (MATS) was developed to identify meningococcus group B strains with a high likelihood of being covered by the 4CMenB vaccine, but is limited by the requirement for viable isolates from culture-confirmed cases. We examined if antigen genotyping could complement MATS in predicting strain coverage by the 4CMenB vaccine. METHODS From a panel of 3912 MATS-typed invasive meningococcal disease isolates collected in England and Wales in 2007-2008, 2014-2015 and 2015-2016, and in 16 other countries in 2000-2015, 3481 isolates were also characterized by antigen genotyping. Individual associations between antigen genotypes and MATS coverage for each 4CMenB component were used to define a genetic MATS (gMATS). gMATS estimates were compared with England and Wales human complement serum bactericidal assay (hSBA) data and vaccine effectiveness (VE) data from England. RESULTS Overall, 81% of the strain panel had genetically predictable MATS coverage, with 92% accuracy and highly concordant results across national panels (Lin's accuracy coefficient, 0.98; root-mean-square deviation, 6%). England and Wales strain coverage estimates were 72-73% by genotyping (66-73% by MATS), underestimating hSBA values after four vaccine doses (88%) and VE after two doses (83%). The gMATS predicted strain coverage in other countries was 58-88%. CONCLUSIONS gMATS can replace MATS in predicting 4CMenB strain coverage in four out of five cases, without requiring a cultivable isolate, and is open to further improvement. Both methods underestimated VE in England. Strain coverage predictions in other countries matched or exceeded England and Wales estimates.
Collapse
Affiliation(s)
| | | | | | | | - Raquel Abad
- National Centre for Microbiology, Institute of Health Carlos III, Madrid, Spain.
| | | | | | | | | | - Pavla Křížová
- National Institute of Public Health, Prague, Czech Republic.
| | - Claudia Mikula
- Austrian Agency for Health and Food Safety, Institute for Medical Microbiology and Hygiene, Graz, Austria.
| | - Robert Mulhall
- Irish Meningitis and Sepsis Reference Laboratory (IMSRL), Dublin, Ireland.
| | - Michael Nissen
- Queensland Paediatric Infectious Diseases Laboratory, Children's Health Research Centre, University of Queensland, Lady Cilento Children's Hospital South Brisbane, Queensland, Australia.
| | - Hanna Nohynek
- National Institute for Health and Welfare (THL), Helsinki, Finland.
| | | | | | - Paola Stefanelli
- Department of Infectious Parasitic and Immune-mediated Diseases, Istituto Superiore di Sanità, Rome, Italy.
| | | | - Maija Toropainen
- National Institute for Health and Welfare (THL), Helsinki, Finland.
| | - Georgina Tzanakaki
- National Meningitis Reference Laboratory, National School of Public Health, Athens, Greece.
| | | | | | - Julio A Vazquez
- National Centre for Microbiology, Institute of Health Carlos III, Madrid, Spain.
| | | | | | - Ray Borrow
- Meningococcal Reference Unit, Public Health England, Manchester Royal Infirmary, Manchester, UK.
| | | |
Collapse
|
49
|
Rappuoli R, Pizza M, Masignani V, Vadivelu K. Meningococcal B vaccine (4CMenB): the journey from research to real world experience. Expert Rev Vaccines 2018; 17:1111-1121. [DOI: 10.1080/14760584.2018.1547637] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Rino Rappuoli
- Chief Scientist & Head of External Research and Development, GSK, Siena, Italy
| | - Mariagrazia Pizza
- Senior Scientific Director, Bacterial Vaccines, Chief Scientist & Head of External Research and Development, Siena, Italy
| | - Vega Masignani
- Discovery Project Leader, Research and Development Centre, Siena, Italy
| | - Kumaran Vadivelu
- Vaccine Development Leader, Research and Development Centre, Rockville, MD, USA
| |
Collapse
|
50
|
Sadarangani M. Protection Against Invasive Infections in Children Caused by Encapsulated Bacteria. Front Immunol 2018; 9:2674. [PMID: 30515161 PMCID: PMC6255856 DOI: 10.3389/fimmu.2018.02674] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 10/30/2018] [Indexed: 11/13/2022] Open
Abstract
The encapsulated bacteria Streptococcus pneumoniae, Neisseria meningitis, Haemophilus influenzae, and Streptococcus agalactiae (Group B Streptococcus) have been responsible for the majority of severe infections in children for decades, specifically bacteremia and meningitis. Isolates which cause invasive disease are usually surrounded by a polysaccharide capsule, which is a major virulence factor and the key antigen in protective protein-polysaccharide conjugate vaccines. Protection against these bacteria is largely mediated via polysaccharide-specific antibody and complement, although the contribution of these and other components, and the precise mechanisms, vary between species and include opsonophagocytosis and complement-dependent bacteriolysis. Further studies are required to more precisely elucidate mechanisms of protection against non-type b H. influenzae and Group B Streptococcus.
Collapse
Affiliation(s)
- Manish Sadarangani
- Vaccine Evaluation Center, BC Children's Hospital Research Institute, Vancouver, BC, Canada.,Division of Infectious Diseases, Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|