1
|
Li Z, Murthy AK, Hao L, Andrew L, Anderson AS. Factor H binding protein (FHbp): An evaluation of genotypic diversity across Neisseria meningitidis serogroups. Hum Vaccin Immunother 2024; 20:2409502. [PMID: 39387286 PMCID: PMC11469366 DOI: 10.1080/21645515.2024.2409502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 09/12/2024] [Accepted: 09/24/2024] [Indexed: 10/15/2024] Open
Abstract
Neisseria meningitidis serogroups A, B, C, W, X, and Y cause invasive meningococcal disease (IMD) worldwide. Factor H binding protein (FHbp), a key meningococcal virulence factor, is an antigen included in both licensed meningococcal serogroup B (MenB) vaccines. This review examines the biology and epidemiology of FHbp and assesses the ability and potential of FHbp vaccine antigens to protect against IMD. Using evidence from the literature and the contemporary PubMLST database, we discuss analyses of MenB genotypes on the representation of the most prevalent multilocus sequence typing (MLST)/clonal complexes, FHbp subfamily distribution, and FHbp and porin A (PorA) variants. We further discuss that the similar genotypes, distribution, and diversity of FHbp variant types have remained stable over long time periods, supporting the potential for FHbp-containing, protein-based vaccines to protect against IMD, including MenB-FHbp (Trumenba®), which contains two lipidated FHbp antigens (one each from both FHbp subfamilies: A and B).
Collapse
Affiliation(s)
- Zhenghui Li
- Pfizer Vaccine Research and Development, Pfizer Inc, Pearl River, NY, USA
| | | | - Li Hao
- Pfizer Vaccine Research and Development, Pfizer Inc, Pearl River, NY, USA
| | - Lubomira Andrew
- Pfizer Vaccine Research and Development, Pfizer Inc, Pearl River, NY, USA
| | | |
Collapse
|
2
|
Veggi D, Chesterman CC, Santini L, Huang Y, Pacchiani N, Sierra J, Chen L, Laliberte J, Bianchi F, Cozzi R, Frigimelica E, Maione D, Finco O, Bottomley MJ. Bactericidal human monoclonal antibody 1B1 shows specificity for meningococcal factor H binding protein variant 2 and displaces human factor H. FASEB Bioadv 2024; 6:235-248. [PMID: 39114449 PMCID: PMC11301264 DOI: 10.1096/fba.2023-00077] [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: 08/03/2023] [Revised: 04/29/2024] [Accepted: 05/17/2024] [Indexed: 08/10/2024] Open
Abstract
Thousands of disease cases and hundreds of deaths occur globally each year due to invasive meningococcal disease. Neisseria meningitidis serogroup B (MenB) is the leading cause of such disease in developed countries. Two vaccines, 4CMenB and MenB-fHbp, that protect against MenB are available and include one or two forms respectively of factor H binding protein (fHbp), a key protective antigen. Studies of circulating meningococci have identified over 1380 different fHbp amino acid sequences, which form three immunologically distinct clusters, termed variants 1, 2, and 3. Neither of the current vaccines contains a variant 2 antigen, which is less well characterized than fHbp variants 1 and 3. We characterized the interaction of fHbp variant 2 with humAb 1B1 using biochemical methods and live meningococcal assays. Further, we determined the crystal structure of the complex at 2.4 Å resolution, clearly revealing the epitope and providing the first detailed report of an antibody with distinct specificity for fHbp variant 2. Extensive mutagenesis and binding studies elucidated key hotspots in the interface. This combination of structural and functional studies provides a molecular explanation for the bactericidal potency and specificity of humAb 1B1 for fHbp variant 2. Our studies, focused on fHbp variant 2, expand the understanding of this previously under characterized group of the vast family of variants of fHbp, a virulence factor present on all meningococci. Moreover, the definition of a protective conformational epitope on fHbp variant 2 may support the design and development of novel variant 2-containing MenB vaccines affording greater breadth of protection.
Collapse
|
3
|
Borrow R, Findlow J. The important lessons lurking in the history of meningococcal epidemiology. Expert Rev Vaccines 2024; 23:445-462. [PMID: 38517733 DOI: 10.1080/14760584.2024.2329618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 03/08/2024] [Indexed: 03/24/2024]
Abstract
INTRODUCTION The epidemiology of invasive meningococcal disease (IMD), a rare but potentially fatal illness, is typically described as unpredictable and subject to sporadic outbreaks. AREAS COVERED Meningococcal epidemiology and vaccine use during the last ~ 200 years are examined within the context of meningococcal characterization and classification to guide future IMD prevention efforts. EXPERT OPINION Historical and contemporary data highlight the dynamic nature of meningococcal epidemiology, with continued emergence of hyperinvasive clones and affected regions. Recent shifts include global increases in serogroup W disease, meningococcal antimicrobial resistance (AMR), and meningococcal urethritis; additionally, unvaccinated populations have experienced disease resurgences following lifting of COVID-19 restrictions. Despite these changes, a close analysis of meningococcal epidemiology indicates consistent dominance of serogroups A, B, C, W, and Y and elevated IMD rates among infants and young children, adolescents/young adults, and older adults. Demonstrably effective vaccines against all 5 major disease-causing serogroups are available, and their prophylactic use represents a powerful weapon against IMD, including AMR. The World Health Organization's goal of defeating meningitis by the year 2030 demands broad protection against IMD, which in turn indicates an urgent need to expand meningococcal vaccination programs across major disease-causing serogroups and age-related risk groups.
Collapse
Affiliation(s)
- Ray Borrow
- Meningococcal Reference Unit, UKHSA, Manchester Royal Infirmary, Manchester, UK
| | - Jamie Findlow
- Global Medical Affairs, Vaccines and Antivirals, Pfizer Ltd, Tadworth, UK
| |
Collapse
|
4
|
Presa J, Burman C, Tort MJ, Cane A, Bocchini JA. Serum bactericidal activity against circulating and reference strains of meningococcal serogroup B in the United States: A review of the strain coverage of meningococcal serogroup B (MenB) vaccines in adolescents and young adults. Hum Vaccin Immunother 2023; 19:2212570. [PMID: 37257838 DOI: 10.1080/21645515.2023.2212570] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 04/24/2023] [Accepted: 05/06/2023] [Indexed: 06/02/2023] Open
Abstract
Invasive meningococcal disease (IMD) is rare but associated with high morbidity and mortality. In the United States, the most vulnerable age groups are infants and adolescents/young adults, and the most common type of IMD is caused by serogroup B (MenB). MenB is preventable among adolescents and young adults with the use of two licensed vaccines, MenB-FHbp (Trumenba®, bivalent rLP2086; Pfizer Inc, Collegeville, PA) and MenB-4C (Bexsero®; GSK Vaccines, Srl, Italy). Because the effectiveness of MenB vaccination is dependent on broad vaccine coverage across circulating disease-causing strains, we reviewed the available clinical and real-world evidence regarding breadth of coverage of the two licensed vaccines in adolescents and young adults in the United States. Both vaccines protect against various MenB strains. More controlled data regarding breadth of coverage across MenB strains are available for MenB-FHbp compared with MenB-4C, whereas more observational data regarding US outbreak strain susceptibility are available for MenB-4C.
Collapse
Affiliation(s)
- Jessica Presa
- Vaccines Medical Development & Scientific/Clinical Affairs, Pfizer Inc, Collegeville, PA, USA
| | - Cynthia Burman
- Vaccines Medical Development & Scientific/Clinical Affairs, Pfizer Inc, Collegeville, PA, USA
| | - Maria J Tort
- Vaccines Medical Development & Scientific/Clinical Affairs, Pfizer Inc, Collegeville, PA, USA
| | - Alejandro Cane
- Vaccines Medical Development & Scientific/Clinical Affairs, Pfizer Inc, Collegeville, PA, USA
| | - Joseph A Bocchini
- Pediatric Infectious Disease, Willis-Knighton Health System, Shreveport, LA, USA
| |
Collapse
|
5
|
Ispasanie E, Muri L, Schmid M, Schubart A, Thorburn C, Zamurovic N, Holbro T, Kammüller M, Pluschke G. In vaccinated individuals serum bactericidal activity against B meningococci is abrogated by C5 inhibition but not by inhibition of the alternative complement pathway. Front Immunol 2023; 14:1180833. [PMID: 37457736 PMCID: PMC10349132 DOI: 10.3389/fimmu.2023.1180833] [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: 03/06/2023] [Accepted: 06/05/2023] [Indexed: 07/18/2023] Open
Abstract
Introduction Several diseases caused by the dysregulation of complement activation can be treated with inhibitors of the complement components C5 and/or C3. However, complement is required for serum bactericidal activity (SBA) against encapsulated Gram-negative bacteria. Therefore, C3 and C5 inhibition increases the risk of invasive disease, in particular by Neisseria meningitidis. As inhibitors against complement components other than C3 and C5 may carry a reduced risk of infection, we compared the effect of inhibitors targeting the terminal pathway (C5), the central complement component C3, the alternative pathway (FB and FD), and the lectin pathway (MASP-2) on SBA against serogroup B meningococci. Methods Serum from adults was collected before and after vaccination with the meningococcal serogroup B vaccine 4CMenB and tested for meningococcal killing. Since the B capsular polysaccharide is structurally similar to certain human polysaccharides, 4CMenB was designed to elicit antibodies against meningococcal outer membrane proteins. Results While only a few pre-vaccination sera showed SBA against the tested B meningococcal isolates, 4CMenB vaccination induced potent complement-activating IgG titers against isolates expressing a matching allele of the bacterial cell surface-exposed factor H-binding protein (fHbp). SBA triggered by these cell surface protein-specific antibodies was blocked by C5 and reduced by C3 inhibition, whereas alternative (factor B and D) and lectin (MASP-2) pathway inhibitors had no effect on the SBA of post-4CMenB vaccination sera. Discussion Compared to the SBA triggered by A,C,W,Y capsule polysaccharide conjugate vaccination, SBA against B meningococci expressing a matching fHbp allele was remarkably resilient against the alternative pathway inhibition.
Collapse
Affiliation(s)
- Emma Ispasanie
- Swiss Tropical and Public Health Institute, Molecular Immunology Unit, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Lukas Muri
- Swiss Tropical and Public Health Institute, Molecular Immunology Unit, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Marc Schmid
- Swiss Tropical and Public Health Institute, Molecular Immunology Unit, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Anna Schubart
- Novartis Institutes for Biomedical Research, Department Autoimmunity, Transplantation and Inflammation, Basel, Switzerland
| | | | - Natasa Zamurovic
- Novartis Institutes for Biomedical Research, Translational Medicine-Preclinical Safety, Basel, Switzerland
| | - Thomas Holbro
- Global Drug Development, Novartis Pharma AG, Basel, Switzerland
| | - Michael Kammüller
- Novartis Institutes for Biomedical Research, Translational Medicine-Preclinical Safety, Basel, Switzerland
| | - Gerd Pluschke
- Swiss Tropical and Public Health Institute, Molecular Immunology Unit, Basel, Switzerland
- University of Basel, Basel, Switzerland
| |
Collapse
|
6
|
Cassiolato AP, Camargo CH, Piccoli Cecconi MC, Christakis S, Gonçalves CR, Rodrigues Campos K, Takenori Higa F, Andrade Pereira G, de Moraes C, Silva de Lemos AP. Genomic characterization of invasive meningococcal X isolates from Brazil, 1992-2022. Int Microbiol 2023:10.1007/s10123-023-00325-5. [PMID: 36626096 PMCID: PMC9838296 DOI: 10.1007/s10123-023-00325-5] [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: 12/09/2022] [Revised: 12/12/2022] [Accepted: 01/04/2023] [Indexed: 01/11/2023]
Abstract
INTRODUCTION Invasive meningococcal disease (IMD) is a major health problem. Given the post-COVID-19 pandemic scenario with the loosening of the non-pharmacological measures to control the virus transmission and considering the observed global reduction of meningococcal vaccination coverage, an increase in IMD cases can be expected. METHODOLOGY Using whole-genome sequencing, we characterized six Neisseria meningitidis serogroup X (MenX) isolates recovered from IMD cases in Brazil in the last 30 years. RESULTS The predominance (66.6%, 4/6) of ST2888 presenting fHbp 160, NHBA 129, NadA 21, and PorA 19,15 was found on isolates. Two novel STs, 15458 and 15477, were described. CONCLUSION This study describes the circulation of MenX lineage ST2888 in Brazil, previously reported only in Europe. Continuous universal surveillance is crucial to implement prompt public health measures aiming to prevent and control non-vaccine preventable serogroup X IMD cases.
Collapse
Affiliation(s)
- Ana Paula Cassiolato
- grid.417672.10000 0004 0620 4215Center of Bacteriology, Instituto Adolfo Lutz (IAL), São Paulo, State of São Paulo Brazil
| | - Carlos Henrique Camargo
- grid.417672.10000 0004 0620 4215Center of Bacteriology, Instituto Adolfo Lutz (IAL), São Paulo, State of São Paulo Brazil
| | | | - Sandra Christakis
- Public Health Laboratory, Florianópolis, State of Santa Catarina Brazil
| | - Claudia Regina Gonçalves
- grid.417672.10000 0004 0620 4215Strategic Laboratory, Instituto Adolfo Lutz (IAL), São Paulo, State of São Paulo Brazil
| | - Karoline Rodrigues Campos
- grid.417672.10000 0004 0620 4215Strategic Laboratory, Instituto Adolfo Lutz (IAL), São Paulo, State of São Paulo Brazil
| | - Fabio Takenori Higa
- Center of Immunology, Institute Adolfo Lutz (IAL), São Paulo, State of São Paulo Brazil
| | - Gabriela Andrade Pereira
- grid.414596.b0000 0004 0602 9808Secretariat of Health Surveillance, Ministry of Health, Brasília, Federal District Brazil
| | - Camile de Moraes
- grid.414596.b0000 0004 0602 9808Secretariat of Health Surveillance, Ministry of Health, Brasília, Federal District Brazil
| | - Ana Paula Silva de Lemos
- grid.417672.10000 0004 0620 4215Center of Bacteriology, Instituto Adolfo Lutz (IAL), São Paulo, State of São Paulo Brazil
| |
Collapse
|
7
|
Marshall HS, Vesikari T, Richmond PC, Wysocki J, Szenborn L, Beeslaar J, Maguire JD, Balmer P, O'Neill R, Anderson AS, Prégaldien JL, Maansson R, Jiang HQ, Perez JL. Safety and immunogenicity of a primary series and booster dose of the meningococcal serogroup B-factor H binding protein vaccine (MenB-FHbp) in healthy children aged 1-9 years: two phase 2 randomised, controlled, observer-blinded studies. THE LANCET. INFECTIOUS DISEASES 2023; 23:103-116. [PMID: 36087588 DOI: 10.1016/s1473-3099(22)00424-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 06/01/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND The meningococcal serogroup B-factor H binding protein vaccine (MenB-FHbp) is licensed for use in children aged 10 years or older for protection against invasive serogroup B meningococcal disease. Because young children are at increased risk of invasive meningococcal disease, MenB-FHbp clinical data in this population are needed. METHODS We conducted two phase 2 randomised, controlled, observer-blinded studies including healthy toddlers (age 12-23 months) across 26 Australian, Czech, Finnish, and Polish centres, and older children (age 2-9 years) across 14 Finnish and Polish centres. Exclusion criteria included previous vaccinations against serogroup B meningococcus or hepatitis A virus (HAV), and chronic antibiotic use. Toddlers were randomly allocated (2:1) via an interactive response technology system to receive either 60 μg or 120 μg MenB-FHbp or HAV vaccine and saline (control). Older children were randomly allocated (3:1) to receive 120 μg MenB-FHbp or control, with stratification by age group (2-3 years and 4-9 years). All vaccinations were administered as three doses (0, 2, and 6 months, with only saline given at 2 months in the control group). Toddlers who received 120 μg MenB-FHbp could receive a 120 μg booster dose 24 months after the end of the primary series. The percentages of participants with serum bactericidal activity using human complement (hSBA) titres at or above the lower limit of quantification (LLOQ; all greater than the 1:4 correlate of protection) against four test strains of serogroup B meningococcus 1 month after the third dose (primary immunogenicity endpoint) were measured in the evaluable immunogenicity populations (participants who received the vaccine as randomised, had available and determinate hSBA results, and had no major protocol violations). Not all participants were tested against all strains because of serum sample volume constraints. The frequencies of reactogenicity and adverse events after each dose were recorded in the safety population (all participants who received at least one dose and had safety data available). These studies are registered with ClinicalTrials.gov (NCT02534935 and NCT02531698) and are completed. FINDINGS Between Aug 31, 2015, and Aug 22, 2016, for the toddler study and between Aug 27, 2015, and March 7, 2016, for the older children study, we enrolled and randomly allocated 396 toddlers (60 μg MenB-FHbp group n=44; 120 μg MenB-FHbp group n=220; control group n=132) and 400 older children (120 μg MenB-FHbp group n=294; control group n=106). 1 month after the third dose, the proportions of participants with hSBA titres at or above the LLOQ ranged across test strains from 85·0% (95% CI 62·1-96·8; 17 of 20 participants) to 100·0% (82·4-100·0; 19 of 19) in toddlers receiving 60 μg MenB-FHbp, and from 71·6% (61·4-80·4; 68 of 95) to 100·0% (96·2-100·0; 95 of 95) in toddlers receiving 120 μg MenB-FHbp, and from 79·1% (71·2-85·6; 106 of 134) to 100·0% (97·4-100·0; 139 of 139) in children aged 2-9 years receiving 120 μg MenB-FHbp. hSBA titres peaked at 1 month after the third primary dose of MenB-FHbp and then declined over time. 24 months after the third dose in the toddler study, the proportions with hSBA titres at or above the LLOQ ranged from 0·0% (0·0-17·6; 0 of 19 participants) to 41·2% (18·4-67·1; seven of 17) in those who received 60 μg MenB-FHbp and from 3·7% (0·8-10·4; three of 81) to 22·8% (14·1-33·6; 18 of 79) in those who received 120 μg MenB-FHbp. 1 month after the booster dose in toddlers, the proportions with hSBA titres at or above the LLOQ were higher than at 1 month after the primary series. MenB-FHbp reactogenicity was mostly transient and of mild to moderate severity. Adverse event frequency was similar between the MenB-FHbp and control groups and less frequent following MenB-FHbp booster than following primary doses. Two participants from the toddler study (both from the 120 μg MenB-FHbp group) and four from the older children study (three from the 120 μg MenB-FHbp group and one from the control group) were withdrawn from the study because of adverse events. INTERPRETATION MenB-FHbp was well tolerated and induced protective immune responses in a high proportion of participants. These findings support a favourable MenB-FHbp immunogenicity and reactogenicity profile in young children, a population at increased risk of adverse invasive meningococcal disease outcomes. FUNDING Pfizer.
Collapse
Affiliation(s)
- Helen S Marshall
- Vaccinology and Immunology Research Trials Unit, Women's and Children's Health Network, North Adelaide, SA, Australia; Robinson Research Institute and Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia.
| | | | - Peter C Richmond
- University of Western Australia School of Medicine, Vaccine Trials Group, Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, WA, Australia; Perth Children's Hospital, Nedlands, WA, Australia
| | - Jacek Wysocki
- Poznań University of Medical Sciences, Poznań, Poland
| | - Leszek Szenborn
- Clinical Department of Pediatric Infectious Diseases, Wroclaw Medical University, Wroclaw, Poland
| | | | - Jason D Maguire
- Pfizer Vaccine Clinical Research and Development, Pearl River, NY, USA
| | - Paul Balmer
- Pfizer Vaccine Medical Development and Scientific/Clinical Affairs, Collegeville, PA, USA
| | - Robert O'Neill
- Pfizer Vaccine Research and Development, Pearl River, NY, USA
| | | | | | - Roger Maansson
- Pfizer Vaccine Clinical Research and Development, Collegeville, PA, USA
| | - Han-Qing Jiang
- Pfizer Vaccine Clinical Research and Development, Pearl River, NY, USA
| | - John L Perez
- Pfizer Vaccine Clinical Research and Development, Collegeville, PA, USA
| |
Collapse
|
8
|
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
|
9
|
Chang CM, Awanye AM, Marsay L, Dold C, Pollard AJ, Rollier CS, Feavers IM, Maiden MCJ, Derrick JP. Application of a Neisseria meningitidis antigen microarray to identify candidate vaccine proteins from a human Phase I clinical trial. Vaccine 2022; 40:3835-3842. [PMID: 35610106 PMCID: PMC7616631 DOI: 10.1016/j.vaccine.2022.05.032] [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: 03/17/2022] [Revised: 05/04/2022] [Accepted: 05/09/2022] [Indexed: 10/18/2022]
Abstract
Meningococcal meningitis is a rare but serious condition affecting mainly children and young adults. Outer membrane vesicles (OMV) from Neisseria meningitidis have been used successfully as vaccines against the disease, although they only provide protection against a limited number of the many existing variants. There have been many attempts to identify suitable protein antigens for use in defined vaccines that provide broad protection against the disease, such as that leading to the development of the four component 4CMenB vaccine. We previously reported the use of a protein antigen microarray to screen for IgG antibodies in sera derived from human recipients of an OMV-based vaccine, as part of a Phase I clinical trial. Here, we show that computational methods can be used to cluster antigens that elicit similar responses in the same individuals. Fitting of IgG antibody binding data to 4,005 linear regressions identified pairs of antigens that exhibited significant correlations. Some were from the same antigens in different quaternary states, whilst others might be correlated for functional or immunological reasons. We also conducted statistical analyses to examine correlations between individual serum bactericidal antibody (SBA) titres and IgG reactivity against specific antigens. Both Kendall's tau and Spearman's rank correlation coefficient statistics identified specific antigens that correlated with log(SBA) titre in five different isolates. The principal antigens identified were PorA and PorB, RmpM, OpcA, and the type IV pilus assembly secretin, PilQ. Other minor antigens identified included a lipoprotein, two proteins from the BAM complex and the efflux channel MtrE. Our results suggest that consideration of the entire antigen composition, and allowance for potential interaction between antigens, could be valuable in designing future meningococcal vaccines. Such an approach has the advantages that it uses data derived from human, rather than animal, immunization and that it avoids the need to screen individual antigens.
Collapse
Affiliation(s)
- Chun-Mien Chang
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PL, UK
| | - Amaka M Awanye
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PL, UK
| | - Leanne Marsay
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, and the NIHR Oxford Biomedical Research Centre, Oxford OX3 7LE, UK
| | - Christina Dold
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, and the NIHR Oxford Biomedical Research Centre, Oxford OX3 7LE, UK
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, and the NIHR Oxford Biomedical Research Centre, Oxford OX3 7LE, UK
| | - Christine S Rollier
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, and the NIHR Oxford Biomedical Research Centre, Oxford OX3 7LE, UK; School of Biosciences and Medicine, University of Surrey, Guildford, UK
| | - Ian M Feavers
- National Institute for Biological Standards and Control, Blanche Lane, South Mimms, Potters Bar, Hertfordshire, UK; Department of Zoology, University of Oxford, 11a Mansfield Road, Oxford OX1 3SZ, UK
| | - Martin C J Maiden
- Department of Zoology, University of Oxford, 11a Mansfield Road, Oxford OX1 3SZ, UK
| | - Jeremy P Derrick
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PL, UK.
| |
Collapse
|
10
|
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
|
11
|
Veggi D, Malito E, Lo Surdo P, Pansegrau W, Rippa V, Wahome N, Savino S, Masignani V, Pizza M, Bottomley MJ. Structural characterization of a cross-protective natural chimera of factor H binding protein from meningococcal serogroup B strain NL096. Comput Struct Biotechnol J 2022; 20:2070-2081. [PMID: 35601959 PMCID: PMC9079162 DOI: 10.1016/j.csbj.2022.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/11/2022] [Accepted: 04/11/2022] [Indexed: 11/25/2022] Open
Abstract
Invasive meningococcal disease can cause fatal sepsis and meningitis and is a global health threat. Factor H binding protein (fHbp) is a protective antigen included in the two currently available vaccines against serogroup B meningococcus (MenB). FHbp is a remarkably variable surface-exposed meningococcal virulence factor with over 1300 different amino acid sequences identified so far. Based on this variability, fHbp has been classified into three variants, two subfamilies or nine modular groups, with low degrees of cross-protective activity. Here, we report the crystal structure of a natural fHbp cross-variant chimera, named variant1-2,3.x expressed by the MenB clinical isolate NL096, at 1.2 Å resolution, the highest resolution of any fHbp structure reported to date. We combined biochemical, site-directed mutagenesis and computational biophysics studies to deeply characterize this rare chimera. We determined the structure to be composed of two adjacent domains deriving from the three variants and determined the molecular basis of its stability, ability to bind Factor H and to adopt the canonical three-dimensional fHbp structure. These studies guided the design of loss-of-function mutations with potential for even greater immunogenicity. Moreover, this study represents a further step in the understanding of the fHbp biological and immunological evolution in nature. The chimeric variant1-2,3.x fHbp protein emerges as an intriguing cross-protective immunogen and suggests that identification of such naturally occurring hybrid proteins may result in stable and cross-protective immunogens when seeking to design and develop vaccines against highly variable pathogens.
Collapse
Affiliation(s)
- Daniele Veggi
- Corresponding author at: GSK Vaccines srl, Via Fiorentina 1, Siena 53100, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Marshall HS, Maiden MCJ. Impact of a Meningococcal Protein-based Serogroup B Vaccine on Serogroup W Invasive Disease in Children. Clin Infect Dis 2021; 73:e1669-e1672. [PMID: 32845980 DOI: 10.1093/cid/ciaa1253] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 08/20/2020] [Indexed: 01/19/2023] Open
Affiliation(s)
- Helen S Marshall
- Robinson Research Institute and Department of Paediatrics, Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia.,Department of Paediatrics, Women's and Children's Health Network, Adelaide, South Australia, Australia
| | | |
Collapse
|
13
|
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
|
14
|
Safadi MAP, Martinón-Torres F, Serra L, Burman C, Presa J. Translating meningococcal serogroup B vaccines for healthcare professionals. Expert Rev Vaccines 2021; 20:401-414. [PMID: 34151699 DOI: 10.1080/14760584.2021.1899820] [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: 10/21/2022]
Abstract
INTRODUCTION Vaccination is an effective strategy to combat invasive meningococcal disease (IMD). Vaccines against the major disease-causing meningococcal serogroups are available; however, development of vaccines against serogroup B faced particular challenges, including the inability to target traditional meningococcal antigens (i.e. polysaccharide capsule) and limited alternative antigens due to serogroup B strain diversity. Two different recombinant, protein-based, serogroup B (MenB) vaccines that may address these challenges are currently available. These vaccines have been extensively evaluated in pre-licensure safety and immunogenicity trials, and recently in real-world studies on effectiveness, safety, and impact on disease burden. AREAS COVERED This review provides healthcare professionals, particularly pediatricians, an overview of currently available MenB vaccines, including development strategies and evaluation of coverage. EXPERT OPINION Overall, recombinant MenB vaccines are valuable tools for healthcare professionals to protect patients against IMD. Their development required innovative design approaches that overcame challenging hurdles and identified novel protein antigen targets; however, important distinctions in the approaches used in their development, evaluation, and administration exist and many unanswered questions remain. Healthcare providers frequently prescribing MenB vaccines are challenged to keep abreast of these differences to ensure patient protection against this serious disease.
Collapse
Affiliation(s)
- Marco Aurelio P Safadi
- Department of Pediatrics, Santa Casa De São Paulo School of Medical Sciences, São Paulo, Brazil
| | - Federico Martinón-Torres
- Translational Pediatrics and Infectious Diseases, Pediatrics Department, Hospital Clínico Universitario De Santiago De Compostela, Santiago De Compostela, Spain.,Genetics, Vaccines and Pediatrics Research Group, Universitario De Santiago De Compostela, Instituto De Investigación Sanitaria De Santiago De Compostela, Santiago De Compostela, Spain
| | - Lidia Serra
- Pfizer Vaccine Medical Development, Scientific and Clinical Affairs, Collegeville, PA, USA
| | - Cynthia Burman
- Pfizer Vaccine Medical Development, Scientific and Clinical Affairs, Collegeville, PA, USA
| | - Jessica Presa
- Pfizer Vaccines, Medical and Scientific Affairs, Collegeville, PA, USA
| |
Collapse
|
15
|
Genetic Diversity of Meningococcal Serogroup B Vaccine Antigens among Carriage Isolates Collected from Students at Three Universities in the United States, 2015-2016. mBio 2021; 12:mBio.00855-21. [PMID: 34006659 PMCID: PMC8262942 DOI: 10.1128/mbio.00855-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Carriage evaluations were conducted during 2015 to 2016 at two U.S. universities in conjunction with the response to disease outbreaks caused by Neisseria meningitidis serogroup B and at a university where outbreak and response activities had not occurred. All eligible students at the two universities received the serogroup B meningococcal factor H binding protein vaccine (MenB-FHbp); 5.2% of students (181/3,509) at one university received MenB-4C. A total of 1,514 meningococcal carriage isolates were obtained from 8,905 oropharyngeal swabs from 7,001 unique participants. Whole-genome sequencing data were analyzed to understand MenB-FHbp’s impact on carriage and antigen genetic diversity and distribution. Of 1,422 isolates from carriers with known vaccination status (726 [51.0%] from MenB-FHbp-vaccinated, 42 [3.0%] from MenB-4C-vaccinated, and 654 [46.0%] from unvaccinated participants), 1,406 (98.9%) had intact fHbp alleles (716 from MenB-FHbp-vaccinated participants). Of 726 isolates from MenB-FHbp-vaccinated participants, 250 (34.4%) harbored FHbp peptides that may be covered by MenB-FHbp. Genogroup B was detected in 122/1,422 (8.6%) and 112/1,422 (7.9%) isolates from MenB-FHbp-vaccinated and unvaccinated participants, respectively. FHbp subfamily and peptide distributions between MenB-FHbp-vaccinated and unvaccinated participants were not statistically different. Eighteen of 161 MenB-FHbp-vaccinated repeat carriers (11.2%) acquired a new strain containing one or more new vaccine antigen peptides during multiple rounds of sample collection, which was not statistically different (P = 0.3176) from the unvaccinated repeat carriers (1/30; 3.3%). Our findings suggest that lack of MenB vaccine impact on carriage was not due to missing the intact fHbp gene; MenB-FHbp did not affect antigen genetic diversity and distribution during the study period.
Collapse
|
16
|
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
|
17
|
de Lemos APS, Sacchi CT, Gonçalves CR, Camargo CH, Andrade AL. Genomic surveillance of Neisseria meningitidis serogroup B invasive strains: Diversity of vaccine antigen types, Brazil, 2016-2018. PLoS One 2020; 15:e0243375. [PMID: 33347452 PMCID: PMC7751880 DOI: 10.1371/journal.pone.0243375] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 11/19/2020] [Indexed: 11/19/2022] Open
Abstract
Background Neisseria meningitidis serogroup B remains a prominent cause of invasive meningococcal disease (IMD) in Brazil. Because two novel protein-based vaccines against serogroup B are available, the main purpose of this study was to provide data on the diversity and distribution of meningococcal vaccine antigen types circulating in Brazil. Methodology Genetic lineages, vaccine antigen types, and allele types of antimicrobial-associated resistance genes based on whole-genome sequencing of a collection of 145 Neisseria meningitidis serogroup B invasive strains recovered in Brazil from 2016 to 2018 were collected. Results A total of 11 clonal complexes (ccs) were identified among the 145 isolates, four of which were predominant, namely, cc461, cc35, cc32, and cc213, accounting for 72.0% of isolates. The most prevalent fHbp peptides were 24 (subfamily A/variant 2), 47 (subfamily A/variant 3), 1 (subfamily B/variant 1) and 45 (subfamily A/variant 3), which were predominantly associated with cc35, cc461, cc32, and cc213, respectively. The NadA peptide was detected in only 26.2% of the isolates. The most frequent NadA peptide 1 was found almost exclusively in cc32. We found seven NHBA peptides that accounted for 74.5% of isolates, and the newly described peptide 1390 was the most prevalent peptide exclusively associated with cc461. Mutated penA alleles were detected in 56.5% of the isolates, whereas no rpoB and gyrA mutant alleles were found. Conclusion During the study period, changes in the clonal structure of circulating strains were observed, without a predominance of a single hyperinvasive lineage, indicating that an epidemiologic shift has occurred that led to a diversity of vaccine antigen types in recent years in Brazil.
Collapse
Affiliation(s)
| | | | | | | | - Ana Lúcia Andrade
- Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia, Brazil
| |
Collapse
|
18
|
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
|
19
|
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
|
20
|
Alderfer J, Isturiz RE, Srivastava A. Lessons from mass vaccination response to meningococcal B outbreaks at US universities. Postgrad Med 2020; 132:614-623. [DOI: 10.1080/00325481.2020.1766265] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- Justine Alderfer
- Medical Development, Scientific & Clinical Affairs, Pfizer Vaccines, Pfizer Inc, Collegeville, PA, USA
| | - Raul E. Isturiz
- Medical Development, Scientific & Clinical Affairs, Pfizer Vaccines, Pfizer Inc, Collegeville, PA, USA
| | - Amit Srivastava
- Medical Development, Scientific & Clinical Affairs, Pfizer Vaccines, Pfizer Inc, Collegeville, PA, USA
| |
Collapse
|
21
|
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
|
22
|
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
|
23
|
Bettinger JA, Liberator P, Halperin SA, Vaudry W, Sadarangani M, Hao L, Lambert N, Jansen KU, Anderson AS, Tsang R. Estimated susceptibility of Canadian meningococcal B isolates to a meningococcal serogroup B vaccine (MenB-FHbp). Vaccine 2020; 38:2026-2033. [PMID: 31983586 DOI: 10.1016/j.vaccine.2019.12.051] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 12/19/2019] [Accepted: 12/20/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Invasive meningococcal disease caused by Neisseria meningitidis serogroup B (MenB) remains a health risk in Canada and globally. Two MenB vaccines are now approved for use. An understanding of the genotype of Canadian strains and the potential strain coverage conferred by the MenB-FHbp vaccine is needed to inform immunization policies. METHODS Serogroup B Neisseria meningitidis strains responsible for meningococcal disease in Canada from 2006 to 2012 were collected as part of the Canadian Immunization Monitoring Program Active surveillance network. Genotypic analysis was done on MenB isolates from 2006 to 2012 with determination of fHbp surface expression for a subset of isolates: those occurring from 2010 to 2012. RESULTS Two clonal complexes (cc269 and cc41/44) were observed in 68.8% of the 276 isolates. A total of 50 different fHbp peptides were identified among isolates from 2006 to 2012. Surface expression of fHbp was detected on 95% of MenB isolates from 2010 to 2012 and 91% of isolates expressed fHbp at levels that are predicted to be susceptible to the bactericidal immune response elicited by the MenB-FHbp vaccine. Some regional differences were observed, particularly in isolates from British Columbia and Quebec. CONCLUSION The majority of MenB isolates responsible for meningococcal disease in Canada expressed fHbp at levels predicted to be sufficient for complement mediated bactericidal activity in the presence of MenB-FHbp induced serum antibodies.
Collapse
Affiliation(s)
- Julie A Bettinger
- Vaccine Evaluation Center, BC Children's Hospital and the University of British Columbia, Vancouver V5Z4H4, Canada.
| | - Paul Liberator
- Vaccine Research and Development, Pfizer, Inc., Pearl River, NY 10965, USA
| | - Scott A Halperin
- Canadian Center for Vaccinology, IWK Health Centre and Dalhousie University, Halifax B3K6R8, Canada
| | - Wendy Vaudry
- Stollery Children's Hospital and University of Alberta, Edmonton T6G1C9, Canada
| | - Manish Sadarangani
- Vaccine Evaluation Center, BC Children's Hospital and the University of British Columbia, Vancouver V5Z4H4, Canada
| | - Li Hao
- Vaccine Research and Development, Pfizer, Inc., Pearl River, NY 10965, USA
| | - Nathaniel Lambert
- Vaccine Research and Development, Pfizer, Inc., Pearl River, NY 10965, USA; Juno Therapeutics, A Celgene Company, Seattle, WA 98102, USA
| | - Kathrin U Jansen
- Vaccine Research and Development, Pfizer, Inc., Pearl River, NY 10965, USA
| | | | - Raymond Tsang
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg R3E3R2, Canada
| |
Collapse
|
24
|
da Silva RAG, Karlyshev AV, Oldfield NJ, Wooldridge KG, Bayliss CD, Ryan A, Griffin R. Variant Signal Peptides of Vaccine Antigen, FHbp, Impair Processing Affecting Surface Localization and Antibody-Mediated Killing in Most Meningococcal Isolates. Front Microbiol 2019; 10:2847. [PMID: 31921030 PMCID: PMC6930937 DOI: 10.3389/fmicb.2019.02847] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 11/25/2019] [Indexed: 11/24/2022] Open
Abstract
Meningococcal lipoprotein, Factor H binding protein (FHbp), is the sole antigen of the Trumenba vaccine (Pfizer) and one of four antigens of the Bexsero vaccine (GSK) targeting Neisseria meningitidis serogroup B isolates. Lipidation of FHbp is assumed to occur for all isolates. We show in the majority of a collection of United Kingdom isolates (1742/1895) non-synonymous single nucleotide polymorphisms (SNPs) in the signal peptide (SP) of FHbp. A single SNP, common to all, alters a polar amino acid that abolishes processing: lipidation and SP cleavage. Whilst some of the FHbp precursor is retained in the cytoplasm due to reduced binding to SecA, remarkably some is translocated and further surface-localized by Slam. Thus we show Slam is not lipoprotein-specific. In a panel of isolates tested, the overall reduced surface localization of the precursor FHbp, compared to isolates with an intact SP, corresponded with decreased susceptibility to antibody-mediated killing. Our findings shed new light on the canonical pathway for lipoprotein processing and translocation of important relevance for lipoprotein-based vaccines in development and in particular for Trumenba.
Collapse
Affiliation(s)
- Ronni A G da Silva
- Centre for Biomolecular Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Andrey V Karlyshev
- School of Life Sciences, Pharmacy and Chemistry, Kingston University, Kingston upon Thames, United Kingdom
| | - Neil J Oldfield
- Centre for Biomolecular Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Karl G Wooldridge
- Centre for Biomolecular Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Christopher D Bayliss
- Department of Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom
| | - Ali Ryan
- School of Life Sciences, Pharmacy and Chemistry, Kingston University, Kingston upon Thames, United Kingdom
| | - Ruth Griffin
- Centre for Biomolecular Sciences, University of Nottingham, Nottingham, United Kingdom
| |
Collapse
|
25
|
Burman C, Alderfer J, Snow VT. A review of the immunogenicity, safety and current recommendations for the meningococcal serogroup B vaccine, MenB-FHbp. J Clin Pharm Ther 2019; 45:270-281. [PMID: 31820483 DOI: 10.1111/jcpt.13083] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 10/14/2019] [Accepted: 11/04/2019] [Indexed: 12/27/2022]
Abstract
WHAT IS KNOWN AND OBJECTIVE This review describes invasive meningococcal disease (IMD) epidemiology in the United States, provides a brief overview of available meningococcal vaccines and discusses meningococcal serogroup B (MenB) vaccine development. Particular focus is given to the recombinant protein MenB vaccine, MenB-FHbp (Trumenba® , bivalent rLP2086) in light of recent publication of phase 3 data; the other MenB vaccine (Bexsero® , MenB-4C) has been recently reviewed. Current recommendations of the US Advisory Committee on Immunization Practices (ACIP) for MenB vaccination and potential barriers to immunization are also discussed. METHODS Using the published literature, this article reviews the development and use of MenB-FHbp to date, with a focus on the United States. RESULTS AND DISCUSSION Despite the availability of medical treatment, IMD is associated with significant mortality and frequently occurring serious permanent sequelae in surviving individuals. Worldwide, most IMD is caused by six serogroups (A, B, C, W, X and Y). MenB is the most common disease-causing meningococcal serogroup in the United States and has caused several recent university-based IMD outbreaks. MenB vaccines, including MenB-FHbp, are available in the United States. ACIP recommends that all individuals ≥10 years of age at increased risk for meningococcal disease receive MenB vaccination; healthy individuals 16-23 years of age are recommended MenB vaccines based on individual clinical decision-making. MenB-FHbp is used on a 2-dose schedule (0, 6 months) when vaccinating healthy individuals and on a tailored 3-dose schedule (0, 1-2, 6 months) in cases of increased risk. WHAT IS NEW AND CONCLUSION Because vaccination provides the most effective protection against IMD, pharmacists are in an excellent position to offer evidence-based vaccine information, as well as to encourage and provide meningococcal immunizations to adolescents and young adults.
Collapse
Affiliation(s)
- Cynthia Burman
- Pfizer Vaccines Medical Development & Scientific and Clinical Affairs, Pfizer Inc, Collegeville, PA, USA
| | - Justine Alderfer
- Pfizer Vaccines Medical Development & Scientific and Clinical Affairs, Pfizer Inc, Collegeville, PA, USA
| | - Vincenza T Snow
- Pfizer Vaccines Medical Development & Scientific and Clinical Affairs, Pfizer Inc, Collegeville, PA, USA
| |
Collapse
|
26
|
Brik A, Terrade A, Hong E, Deghmane A, Taha MK, Bouafsoun A, Khmiri M, Boussetta K, Boukhir S, Jaballah NB, Kechrid A, Smaoui H. Phenotypic and genotypic characterization of meningococcal isolates in Tunis, Tunisia: High diversity and impact on vaccination strategies. Int J Infect Dis 2019; 91:73-78. [PMID: 31756567 DOI: 10.1016/j.ijid.2019.11.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 11/07/2019] [Accepted: 11/12/2019] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVES The aim of this study was to characterize Neisseria meningitidis (Men) isolates in Tunisian paediatric patients with invasive meningococcal disease (IMD) in order to target therapeutic and preventive strategies. METHODS Fifty-nine isolates of Men and four cerebrospinal fluid samples that were culture-negative but Men-positive by PCR (NC-MenPPCR) (2009-2016) were collected from IMD patients. Isolates were analysed for their antimicrobial susceptibility. Whole-genome sequencing (WGS) was used to characterize isolates and multilocus sequence typing for NC-MenPPCR. Coverage of Men serogroup B (MenB) was determined by Genetic Meningococcal Antigen Typing System (gMATS) and fHbp expression by ELISA. RESULTS MenB was the predominant type (88.9%). The majority of isolates (81%) had reduced susceptibility to penicillin G with altered penA alleles. The clonal complex CC461 (27.1%) was the most frequent. Among the MenB vaccine targets neisserial heparin binding antigen (NHBA) and fHbp, the predominant variants were NHBA118 (30.8%) and fHbp peptide 47 (25%), respectively. The nadA gene was present in 17.3% of isolates. Using gMATS, 36.5% of MenB were predicted to be covered by the 4CMenB vaccine. ELISA showed that 92.4% of the MenB were expected to be killed by anti-fHbp antibodies. CONCLUSIONS MenB was the leading serogroup in IMD, and more than 90% had a sufficient level of fHbp expression for vaccine coverage. The study results will be useful for the Tunisian vaccination programme.
Collapse
Affiliation(s)
- A Brik
- University of Tunis El Manar, Children's Hospital of Tunis, Laboratory of Microbiology, LR18ES39, Beb Saadoun, 1007 Tunis, Tunisia; Institut Pasteur, Invasive Bacterial Infections Unit, 25-28 Rue du Docteur Roux, 75015 Paris, France
| | - A Terrade
- Institut Pasteur, Invasive Bacterial Infections Unit, 25-28 Rue du Docteur Roux, 75015 Paris, France
| | - E Hong
- Institut Pasteur, Invasive Bacterial Infections Unit, 25-28 Rue du Docteur Roux, 75015 Paris, France
| | - A Deghmane
- Institut Pasteur, Invasive Bacterial Infections Unit, 25-28 Rue du Docteur Roux, 75015 Paris, France
| | - M K Taha
- Institut Pasteur, Invasive Bacterial Infections Unit, 25-28 Rue du Docteur Roux, 75015 Paris, France
| | - A Bouafsoun
- University of Tunis El Manar, Children's Hospital of Tunis, Laboratory of Microbiology, LR18ES39, Beb Saadoun, 1007 Tunis, Tunisia
| | - M Khmiri
- Department of Paediatrics A, Children's Hospital of Tunis, Beb Saadoun, 1007 Tunis, Tunisia
| | - K Boussetta
- Department of Paediatrics B, Children's Hospital of Tunis, Beb Saadoun, 1007 Tunis, Tunisia
| | - S Boukhir
- Department of Paediatrics C, Children's Hospital of Tunis, Beb Saadoun, 1007 Tunis, Tunisia
| | - N Ben Jaballah
- Intensive Care Unit, Béchir Hamza Children's Hospital of Tunis, Beb Saadoun, 1007 Tunis, Tunisia
| | - A Kechrid
- University of Tunis El Manar, Children's Hospital of Tunis, Laboratory of Microbiology, LR18ES39, Beb Saadoun, 1007 Tunis, Tunisia
| | - H Smaoui
- University of Tunis El Manar, Children's Hospital of Tunis, Laboratory of Microbiology, LR18ES39, Beb Saadoun, 1007 Tunis, Tunisia.
| |
Collapse
|
27
|
Rivero-Calle I, Raguindin PF, Gómez-Rial J, Rodriguez-Tenreiro C, Martinón-Torres F. Meningococcal Group B Vaccine For The Prevention Of Invasive Meningococcal Disease Caused By Neisseria meningitidis Serogroup B. Infect Drug Resist 2019; 12:3169-3188. [PMID: 31632103 PMCID: PMC6793463 DOI: 10.2147/idr.s159952] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Accepted: 09/12/2019] [Indexed: 12/13/2022] Open
Abstract
Invasive meningococcal disease (IMD) is a major public health concern because of its high case fatality, long-term morbidity, and potential to course with outbreaks. IMD caused by Nesseira meningitidis serogroup B has been predominant in different regions of the world like Europe and only recently broadly protective vaccines against B serogroup have become available. Two protein-based vaccines, namely 4CMenB (Bexsero®) and rLP2086 (Trumenba®) are currently licensed for use in different countries against MenB disease. These vaccines came from a novel technology on vaccine design (or antigen selection) using highly specific antigen targets identified through whole-genome sequence analysis. Moreover, it has the potential to confer protection against non-B meningococcus and against other Neisserial species such as gonococcus. Real-world data on the vaccine-use are rapidly accumulating from the UK and other countries which used the vaccine for control of outbreak or as part of routine immunization program, reiterating its safety and efficacy. Additional data on real-life effectiveness, long-term immunity, and eventual herd effects, including estimates on vaccine impact for cost-effectiveness assessment are further needed. Given the predominance of MenB in Europe and other parts of the world, these new vaccines are crucial for the prevention and public health control of the disease, and should be considered.
Collapse
Affiliation(s)
- Irene Rivero-Calle
- Translational Pediatrics and Infectious Diseases, Department of Pediatrics, Hospital Clínico Universitario de Santiago de Compostela, Galicia, Spain
- Genetics, Vaccines and Pediatric Infectious Diseases Research Group (GENVIP), Hospital Clínico Universitario and Universidad de Santiago de Compostela (USC), Galicia, Spain
| | - Peter Francis Raguindin
- Genetics, Vaccines and Pediatric Infectious Diseases Research Group (GENVIP), Hospital Clínico Universitario and Universidad de Santiago de Compostela (USC), Galicia, Spain
| | - Jose Gómez-Rial
- Genetics, Vaccines and Pediatric Infectious Diseases Research Group (GENVIP), Hospital Clínico Universitario and Universidad de Santiago de Compostela (USC), Galicia, Spain
| | - Carmen Rodriguez-Tenreiro
- Genetics, Vaccines and Pediatric Infectious Diseases Research Group (GENVIP), Hospital Clínico Universitario and Universidad de Santiago de Compostela (USC), Galicia, Spain
| | - Federico Martinón-Torres
- Translational Pediatrics and Infectious Diseases, Department of Pediatrics, Hospital Clínico Universitario de Santiago de Compostela, Galicia, Spain
- Genetics, Vaccines and Pediatric Infectious Diseases Research Group (GENVIP), Hospital Clínico Universitario and Universidad de Santiago de Compostela (USC), Galicia, Spain
| |
Collapse
|
28
|
Genetic Similarity of Gonococcal Homologs to Meningococcal Outer Membrane Proteins of Serogroup B Vaccine. mBio 2019; 10:mBio.01668-19. [PMID: 31506309 PMCID: PMC6737241 DOI: 10.1128/mbio.01668-19] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The human pathogens Neisseria gonorrhoeae and Neisseria meningitidis share high genome identity. Retrospective analysis of surveillance data from New Zealand indicates the potential cross-protective effect of outer membrane vesicle (OMV) meningococcal serogroup B vaccine (MeNZB) against N. gonorrhoeae A licensed OMV-based MenB vaccine, MenB-4C, consists of a recombinant FHbp, NhbA, NadA, and the MeNZB OMV. Previous work has identified several abundantly expressed outer membrane proteins (OMPs) as major components of the MenB-4C OMV with high sequence similarity between N. gonorrhoeae and N. meningitidis, suggesting a mechanism for cross-protection. To build off these findings, we performed comparative genomic analysis on 970 recent N. gonorrhoeae isolates collected through a U.S surveillance system against N. meningitidis serogroup B (NmB) reference sequences. We identified 1,525 proteins that were common to both Neisseria species, of which 57 proteins were predicted to be OMPs using in silico methods. Among the MenB-4C antigens, NhbA showed moderate sequence identity (73%) to the respective gonococcal homolog, was highly conserved within N. gonorrhoeae, and was predicted to be surface expressed. In contrast, the gonococcal FHbp was predicted not to be surface expressed, while NadA was absent in all N. gonorrhoeae isolates. Our work confirmed recent observations (E. A. Semchenko, A. Tan, R. Borrow, and K. L. Seib, Clin Infect Dis, 2018, https://doi.org/10.1093/cid/ciy1061) and describes homologous OMPs from a large panel of epidemiologically relevant N. gonorrhoeae strains in the United States against NmB reference strains. Based on our results, we report a set of OMPs that may contribute to the previously observed cross-protection and provide potential antigen targets to guide the next steps in gonorrhea vaccine development.IMPORTANCE Gonorrhea, a sexually transmitted disease, causes substantial global morbidity and economic burden. New prevention and control measures for this disease are urgently needed, as strains resistant to almost all classes of antibiotics available for treatment have emerged. Previous reports demonstrate that cross-protection from gonococcal infections may be conferred by meningococcal serogroup B (MenB) outer membrane vesicle (OMV)-based vaccines. Among 1,525 common proteins shared across the genomes of both N. gonorrhoeae and N. meningitidis, 57 proteins were predicted to be surface expressed (outer membrane proteins [OMPs]) and thus preferred targets for vaccine development. The majority of these OMPs showed high sequence identity between the 2 bacterial species. Our results provide valuable insight into the meningococcal antigens present in the current OMV-containing MenB-4C vaccine that may contribute to cross-protection against gonorrhea and may inform next steps in gonorrhea vaccine development.
Collapse
|
29
|
Distribution of Neisseria meningitidis serogroup b (NmB) vaccine antigens in meningococcal disease causing isolates in the United States during 2009-2014, prior to NmB vaccine licensure. J Infect 2019; 79:426-434. [PMID: 31505201 DOI: 10.1016/j.jinf.2019.09.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 09/02/2019] [Accepted: 09/04/2019] [Indexed: 12/17/2022]
Abstract
OBJECTIVES Two Neisseria meningitidis serogroup B (NmB) vaccines are licensed in the United States. To estimate their potential coverage, we examined the vaccine antigen diversity among meningococcal isolates prior to vaccine licensure. METHODS NmB vaccine antigen genes of invasive isolates collected in the U.S. from 2009 to 2014 were characterized by Sanger or whole-genome sequencing. RESULTS During 2009-2014, the predominant antigen types have remained similar to those reported in 2000-2008 for NmB and 2006-2008 for NmC, NmY, with the emergence of a few new types. FHbp of subfamily B or variant 1 (B/v1) remained prevalent among NmB whereas FHbp of subfamily A or variant 2 and 3 (A/v2-3) were more prevalent among non-NmB. FHbp peptide 1 (B24/1.1) remains the most prevalent type in NmB. Full-length NadA peptide was detected in 26% of isolates, primarily in NmB and NmW. The greatest diversity of NhbA peptides was detected among NmB, with p0005 as the most prevalent type. CONCLUSIONS The prevalence and diversity of the NmB vaccine antigens have remained stable with common antigen types persisting over time. The data collected prior to NmB vaccine licensure provide the baseline to understand the potential impact of NmB vaccines on antigen diversity and strain coverage.
Collapse
|
30
|
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
|
31
|
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
|
32
|
Rodrigues CMC, Chan H, Vipond C, Jolley K, Harrison OB, Wheeler J, Whiting G, Feavers IM, Maiden MCJ. Typing complex meningococcal vaccines to understand diversity and population structure of key vaccine antigens. Wellcome Open Res 2019; 3:151. [PMID: 30687793 DOI: 10.12688/wellcomeopenres.14859.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/13/2018] [Indexed: 11/20/2022] Open
Abstract
Background: Protein-conjugate capsular polysaccharide vaccines can potentially control invasive meningococcal disease (IMD) caused by five (A, C, W, X, Y) of the six IMD-associated serogroups. Concerns raised by immunological similarity of the serogroup B capsule to human neural cell carbohydrates, meant that 'serogroup B substitute' vaccines target more variable subcapsular protein antigens. A successful approach using outer membrane vesicles (OMVs) as major vaccine components had limited strain coverage. In 4CMenB (Bexsero ®), recombinant proteins have been added to ameliorate this problem. Methods: Scalable, portable, genomic techniques were used to investigate the Bexsero ® OMV protein diversity in meningococcal populations. Shotgun proteomics identified 461 proteins in the OMV, defining a complex proteome. Amino acid sequences for the 24 proteins most likely to be involved in cross-protective immune responses were catalogued within the PubMLST.org/neisseria database using a novel OMV peptide Typing (OMVT) scheme. Results: Among these proteins there was variation in the extent of diversity and association with meningococcal lineages, identified as clonal complexes (ccs), ranging from the most conserved peptides (FbpA, NEISp0578, and putative periplasmic protein, NEISp1063) to the most diverse (TbpA, NEISp1690). There were 1752 unique OMVTs identified amongst 2492/3506 isolates examined by whole-genome sequencing (WGS). These OMVTs were grouped into clusters (sharing ≥18 identical OMVT peptides), with 45.3% of isolates assigned to one of 27 OMVT clusters. OMVTs and OMVT clusters were strongly associated with cc, genogroup, and Bexsero ® antigen variants, demonstrating that combinations of OMV proteins exist in discrete, non-overlapping combinations associated with genogroup and Bexsero ® Antigen Sequence Type. This highly structured population of IMD-associated meningococci is consistent with strain structure models invoking host immune and/or metabolic selection. Conclusions: The OMVT scheme facilitates region-specific WGS investigation of meningococcal diversity and is an open-access, portable tool with applications for vaccine development, especially in the choice of antigen combinations, assessment and implementation.
Collapse
Affiliation(s)
| | - Hannah Chan
- Division of Bacteriology, National Institute for Biological Standards and Control, Potters Bar, EN6 3QG, UK
| | - Caroline Vipond
- Division of Bacteriology, National Institute for Biological Standards and Control, Potters Bar, EN6 3QG, UK
| | - Keith Jolley
- Department of Zoology, University of Oxford, Oxford, OX1 3SY, UK
| | - Odile B Harrison
- Department of Zoology, University of Oxford, Oxford, OX1 3SY, UK
| | - Jun Wheeler
- Division of Bacteriology, National Institute for Biological Standards and Control, Potters Bar, EN6 3QG, UK
| | - Gail Whiting
- Division of Bacteriology, National Institute for Biological Standards and Control, Potters Bar, EN6 3QG, UK
| | - Ian M Feavers
- Division of Bacteriology, National Institute for Biological Standards and Control, Potters Bar, EN6 3QG, UK
| | | |
Collapse
|
33
|
Rodrigues CMC, Chan H, Vipond C, Jolley K, Harrison OB, Wheeler J, Whiting G, Feavers IM, Maiden MCJ. Typing complex meningococcal vaccines to understand diversity and population structure of key vaccine antigens. Wellcome Open Res 2019; 3:151. [PMID: 30687793 PMCID: PMC6338130 DOI: 10.12688/wellcomeopenres.14859.2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/07/2019] [Indexed: 01/09/2023] Open
Abstract
Background: Protein-conjugate capsular polysaccharide vaccines can potentially control invasive meningococcal disease (IMD) caused by five (A, C, W, X, Y) of the six IMD-associated serogroups. Concerns raised by immunological similarity of the serogroup B capsule to human neural cell carbohydrates, meant that ‘serogroup B substitute’ vaccines target more variable subcapsular protein antigens. A successful approach using outer membrane vesicles (OMVs) as major vaccine components had limited strain coverage. In 4CMenB (Bexsero
®), recombinant proteins have been added to ameliorate this problem. Methods: Scalable, portable, genomic techniques were used to investigate the Bexsero
® OMV protein diversity in meningococcal populations. Shotgun proteomics identified 461 proteins in the OMV, defining a complex proteome. Amino acid sequences for the 24 proteins most likely to be involved in cross-protective immune responses were catalogued within the
PubMLST.org/neisseria database using a novel OMV peptide Typing (OMVT) scheme. Results: Among these proteins there was variation in the extent of diversity and association with meningococcal lineages, identified as clonal complexes (ccs), ranging from the most conserved peptides (FbpA, NEISp0578, and putative periplasmic protein, NEISp1063) to the most diverse (TbpA, NEISp1690). There were 1752 unique OMVTs identified amongst 2492/3506 isolates examined by whole-genome sequencing (WGS). These OMVTs were grouped into clusters (sharing ≥18 identical OMVT peptides), with 45.3% of isolates assigned to one of 27 OMVT clusters. OMVTs and OMVT clusters were strongly associated with cc, genogroup, and Bexsero
® antigen variants, demonstrating that combinations of OMV proteins exist in discrete, non-overlapping combinations associated with genogroup and Bexsero
® Antigen Sequence Type. This highly structured population of IMD-associated meningococci is consistent with strain structure models invoking host immune and/or metabolic selection. Conclusions:The OMVT scheme facilitates region-specific WGS investigation of meningococcal diversity and is an open-access, portable tool with applications for vaccine development, especially in the choice of antigen combinations, assessment and implementation.
Collapse
Affiliation(s)
| | - Hannah Chan
- Division of Bacteriology, National Institute for Biological Standards and Control, Potters Bar, EN6 3QG, UK
| | - Caroline Vipond
- Division of Bacteriology, National Institute for Biological Standards and Control, Potters Bar, EN6 3QG, UK
| | - Keith Jolley
- Department of Zoology, University of Oxford, Oxford, OX1 3SY, UK
| | - Odile B Harrison
- Department of Zoology, University of Oxford, Oxford, OX1 3SY, UK
| | - Jun Wheeler
- Division of Bacteriology, National Institute for Biological Standards and Control, Potters Bar, EN6 3QG, UK
| | - Gail Whiting
- Division of Bacteriology, National Institute for Biological Standards and Control, Potters Bar, EN6 3QG, UK
| | - Ian M Feavers
- Division of Bacteriology, National Institute for Biological Standards and Control, Potters Bar, EN6 3QG, UK
| | | |
Collapse
|
34
|
Tenenbaum T, Hellenbrand W, Schroten H. Impfstoffe gegen Meningokokken für das Kindesalter. Monatsschr Kinderheilkd 2019. [DOI: 10.1007/s00112-018-0635-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
35
|
Findlow J, Nuttens C, Kriz P. Introduction of a second MenB vaccine into Europe – needs and opportunities for public health. Expert Rev Vaccines 2019; 18:225-239. [DOI: 10.1080/14760584.2019.1578217] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Jamie Findlow
- Medical & Scientific Affairs – International Developed Markets, Pfizer Limited, Tadworth, UK
| | - Charles Nuttens
- Medical & Scientific Affairs – International Developed Markets, Pfizer, Paris, France
| | - Paula Kriz
- Centre for Epidemiology and Microbiology – National Institute of Public Health, Prague, Czech Republic
| |
Collapse
|
36
|
Vesikari T, Østergaard L, Beeslaar J, Absalon J, Eiden JJ, Jansen KU, Jones TR, Harris SL, Maansson R, Munson S, O'Neill RE, York LJ, Perez JL. Persistence and 4-year boosting of the bactericidal response elicited by two- and three-dose schedules of MenB-FHbp: A phase 3 extension study in adolescents. Vaccine 2019; 37:1710-1719. [PMID: 30770221 DOI: 10.1016/j.vaccine.2018.11.073] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 07/25/2018] [Accepted: 11/26/2018] [Indexed: 01/11/2023]
Abstract
BACKGROUND The period of heightened risk of invasive meningococcal disease in adolescence extends for >10 years. This study aimed to evaluate persistence of the immune response to the serogroup B meningococcal (MenB) vaccine MenB-FHbp (Trumenba®, Bivalent rLP2086) under two- and three-dose primary vaccination schedules, both of which are approved in the United States and the European Union, and to assess safety and immunogenicity of a booster dose. METHODS This was an open-label extension study of a phase 2 randomized MenB-FHbp study (primary study). This interim analysis includes data through 1 month after booster vaccination. In the primary study, adolescents 11-18 years of age were randomized using an interactive voice or web-based response system to receive 120 μg MenB-FHbp under 0-, 1-, 6-month; 0-, 2-, 6-month; 0-, 6-month; 0-, 2-month; or 0-, 4-month schedules (termed study groups for the current analysis). For the primary study, participants were blinded to their vaccine study group allocation, but investigators and the study sponsor were unblinded. Immune responses in subjects from the primary study were evaluated through 48 months after primary vaccination (persistence stage; 17 sites in Czech Republic, Denmark, Germany, and Sweden). Safety and immunogenicity of a booster dose given at 48 months after primary vaccination (booster stage; 14 sites in Czech Republic, Denmark, and Sweden) were also assessed. Immune responses were evaluated in serum bactericidal assays with human complement (hSBAs) using four MenB test strains representative of disease-causing MenB strains in the United States and Europe and expressing factor H binding proteins (FHbps) heterologous to the vaccine antigens. The primary immunogenicity endpoints were the proportions of subjects with hSBA titers greater than or equal to the assays' lower limit of quantitation (LLOQ; 1:8 or 1:16 depending on strain) at 12, 18, 24, 36, and 48 months after primary vaccination (persistence stage) and 1 and 48 months after the primary vaccination series and 1 month after receipt of the booster dose (booster stage). Safety evaluations during the booster stage included local reactions and systemic events by severity, antipyretic use, adverse events (AEs), immediate AEs, serious AEs (SAEs), medically attended AEs (MAEs), newly diagnosed chronic medical conditions (NDCMCs), and missed days of school and work because of AEs. The modified intent-to-treat (mITT) population was used for immunogenicity evaluations in the persistence stage. The booster stage immunogenicity evaluations used the evaluable immunogenicity population; analyses were also performed in the mITT population. For the persistence stage, safety evaluations included subjects with at least one blood draw, whereas for the booster stage, they included subjects who received the booster dose and had available safety data. This trial is registered at ClinicalTrials.gov number NCT01543087. FINDINGS A total of 465 subjects were enrolled in the persistence stage, and 271 subjects were enrolled in the booster stage. Sera for the extension phase of this interim analysis were collected from September 7, 2012 to December 7, 2015. One month after primary vaccination, 73.8-100.0% of subjects depending on study group responded with hSBA titers ≥LLOQ. Response rates declined during the 12 months after last primary vaccination and then remained stable through 48 months, with 18.0-61.3% of subjects depending on study group having hSBA titers ≥LLOQ at this time point. One month after receipt of the booster dose, 91.9-100.0% of subjects depending on study group had hSBA titers ≥LLOQ against the four primary strains individually and 91.8-98.2% had hSBA titers ≥LLOQ against all four strains combined (composite response). Geometric mean titers were higher after booster vaccination than at 1 month after primary vaccination. Immune responses were generally similar across study groups, regardless of whether a two- or three-dose primary series was received. None of the AEs (2.2-6.9% of subjects depending on study group) or NDCMCs (1.8-5.0%) that were reported during the persistence stage were considered related to the investigational product. Local reactions and systemic events were reported by 84.4-93.8% and 68.8-76.6% of subjects depending on study group, respectively, in the booster stage; these were generally similar across study groups, transient, and less frequent than after any primary vaccination. Additionally, there was no general progressive worsening in severity of reactogenicity events (ie, potentiation; ≤3 subjects per group), and reactogenicity events did not lead to any study withdrawals. No NDCMCs or immediate AEs were reported during the booster stage. AEs were reported by 3.7-12.5% of subjects depending on study group during the booster stage. The two possibly related AEs included a mild worsening of psoriasis and a severe influenza-like illness that resolved in 10 days. INTERPRETATION Immune responses declined after the primary vaccination series; however, a substantially greater number of subjects retained protective responses at 48 months after primary vaccination compared with subjects having protective responses before vaccination. Persistence trends were similar across all 5 study groups regardless of whether a two- or three-dose primary schedule was received. Furthermore, a booster dose given 48 months after primary vaccination was safe, well-tolerated, and elicited robust immune responses indicative of immunologic memory; these responses were similar between two- and three-dose primary schedule study groups. Use of a booster dose may help further extend protection against MenB disease in adolescents. FUNDING Pfizer Inc.
Collapse
Affiliation(s)
- Timo Vesikari
- Vaccine Research Center, University of Tampere Medical School, Biokatu 10, 33520 Tampere, Finland
| | - Lars Østergaard
- Department of Infectious Diseases, Aarhus University Hospital, Skejby, Palle Juul-Jensens Blvd 99, 8200 Aarhus N, Denmark
| | - Johannes Beeslaar
- Pfizer UK Vaccine Research and Development, Horizon Building, Honey Lane, Hurley SL6 6RJ, UK.
| | - Judith Absalon
- Pfizer Vaccine Research and Development, 401 North Middletown Road, Pearl River, NY 10965, USA
| | - Joseph J Eiden
- Pfizer Vaccine Research and Development, 401 North Middletown Road, Pearl River, NY 10965, USA
| | - Kathrin U Jansen
- Pfizer Vaccine Research and Development, 401 North Middletown Road, Pearl River, NY 10965, USA
| | - Thomas R Jones
- Pfizer Vaccine Research and Development, 401 North Middletown Road, Pearl River, NY 10965, USA
| | - Shannon L Harris
- Pfizer Vaccine Research and Development, 401 North Middletown Road, Pearl River, NY 10965, USA
| | - Roger Maansson
- Pfizer Vaccine Research and Development, 500 Arcola Road, Collegeville, PA 19426, USA
| | - Samantha Munson
- Pfizer Vaccine Research and Development, 500 Arcola Road, Collegeville, PA 19426, USA
| | - Robert E O'Neill
- Pfizer Vaccine Research and Development, 401 North Middletown Road, Pearl River, NY 10965, USA
| | - Laura J York
- Pfizer Vaccine Medical Development, Scientific & Clinical Affairs, 500 Arcola Road, Collegeville, PA 19426, USA
| | - John L Perez
- Pfizer Vaccine Research and Development, 500 Arcola Road, Collegeville, PA 19426, USA
| |
Collapse
|
37
|
Maiden MCJ. The Impact of Nucleotide Sequence Analysis on Meningococcal Vaccine Development and Assessment. Front Immunol 2019; 9:3151. [PMID: 30697213 PMCID: PMC6340965 DOI: 10.3389/fimmu.2018.03151] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 12/20/2018] [Indexed: 12/21/2022] Open
Abstract
Since it became available as a routine tool in biology, the determination and analysis of nucleotide sequences has been applied to the design of vaccines and the investigation of their effectiveness. As vaccination is primarily concerned with the interaction of biological molecules with the immune system, the utility of sequence data is not immediately obvious and, indeed, nucleotide sequence data are most effective when used to complement more conventional immunological approaches. Here, the impact of sequencing on the field of vaccinology will be illustrated with reference to the development and implementation of vaccines against Neisseria meningitidis (the meningococcus) over the 30-year period from the late-1980s to the late-2010s. Nucleotide sequence-based studies have been important in the fight against this aggressive pathogen largely because of its high genetic and antigenic diversity, properties that were only fully appreciated because of sequence-based studies. Five aspects will be considered, the use of sequence data to: (i) discover vaccine antigens; (ii) assess the diversity and distribution of vaccine antigens; (iii) determine the evolutionary and population biology of the organism and their implications for immunization; and (iv) develop molecular approaches to investigate pre- and post-vaccine pathogen populations to assess vaccine impact. One of the great advantages of nucleotide sequence data has been its scalability, which has meant that increasingly large data sets have been available, which has proved invaluable in the investigation of an organism as diverse and enigmatic as the meningococcus.
Collapse
|
38
|
AZZARI C, BONANNI P. A new meningococcal B vaccine for adolescents and adults: characteristics and methods of use. JOURNAL OF PREVENTIVE MEDICINE AND HYGIENE 2018; 59:E257-E260. [PMID: 30656227 PMCID: PMC6319125 DOI: 10.15167/2421-4248/jpmh2018.59.4.1096] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 11/30/2018] [Indexed: 01/24/2023]
Abstract
The invasive disease from Neisseria meningitidis is one of the leading causes of death for meningitis and sepsis at all ages. The highest incidence of cases occurs at paediatric and adolescent age, but no age of life is considered protected from this infection and disease. Prevention against the five main serogroups is possible using the combined conjugated polysaccharide vaccine against the ACWY (anti-MenACWY) serogroups and the meningococcal B (anti-MenB) protein vaccines. Trumenba® vaccine, approved by the EMA (European Medicine Agency) for use in individuals aged ≥ 10 years, protects against serogroup B invasive disease. This bivalent, recombinant vaccine is able, when given with a 0-6 month schedule, to induce a protective response in adolescents and young adults, comparable with a 3-doses schedule. For this reason, the Trumenba® vaccine should be used routinely with the 2-dose schedule (0-6 months). The 3-doses use could be considered in particular situations, like an occurring epidemic or particular individual risk factors such as asplenia or complement deficit, but is not needed for underlying conditions like diabetes or heart diseases.
Collapse
Affiliation(s)
- C. AZZARI
- Pediatric Immunology Unit “Anna Meyer” Hospital, University of Florence, Italy
- Department of Health Sciences, University of Florence, Italy
| | - P. BONANNI
- Department of Health Sciences, University of Florence, Italy
- * Correspondence: Paolo Bonanni, Department of Health Sciences, University of Florence, Italy - E-mail:
| |
Collapse
|
39
|
Harris SL, Tan C, Andrew L, Hao L, Liberator PA, Absalon J, Anderson AS, Jones TR. The bivalent factor H binding protein meningococcal serogroup B vaccine elicits bactericidal antibodies against representative non-serogroup B meningococci. Vaccine 2018; 36:6867-6874. [DOI: 10.1016/j.vaccine.2018.05.081] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 05/21/2018] [Accepted: 05/22/2018] [Indexed: 11/26/2022]
|
40
|
Perez JL, Absalon J, Beeslaar J, Balmer P, Jansen KU, Jones TR, Harris S, York LJ, Jiang Q, Radley D, Anderson AS, Crowther G, Eiden JJ. From research to licensure and beyond: clinical development of MenB-FHbp, a broadly protective meningococcal B vaccine. Expert Rev Vaccines 2018; 17:461-477. [DOI: 10.1080/14760584.2018.1483726] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- John L. Perez
- Pfizer Vaccines Research and Development, Collegeville, PA, USA
| | - Judith Absalon
- Pfizer Vaccines Research and Development, Pearl River, NY, USA
| | | | - Paul Balmer
- Pfizer Vaccines Research and Development, Collegeville, PA, USA
| | | | - Thomas R. Jones
- Pfizer Vaccines Research and Development, Pearl River, NY, USA
| | - Shannon Harris
- Pfizer Vaccines Research and Development, Pearl River, NY, USA
| | - Laura J. York
- Pfizer Vaccines Medical Development, Scientific & Clinical Affairs, Collegeville, PA, USA
| | - Qin Jiang
- Pfizer Vaccines Research and Development, Collegeville, PA, USA
| | - David Radley
- Pfizer Vaccines Research and Development, Collegeville, PA, USA
| | | | | | - Joseph J. Eiden
- Pfizer Vaccines Research and Development, Pearl River, NY, USA
| |
Collapse
|
41
|
Balmer P, York LJ. Optimal use of meningococcal serogroup B vaccines: moving beyond outbreak control. Ther Adv Vaccines Immunother 2018; 6:49-60. [PMID: 30182092 DOI: 10.1177/2515135518781757] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 04/26/2018] [Indexed: 11/16/2022] Open
Abstract
Neisseria meningitidis is a major cause of meningitis and septicemia globally. Vaccines directed against N. meningitidis serogroup B (MenB) have been used to control sporadic and sustained disease in industrialized and non-industrialized countries. Early outer membrane vesicle (OMV) vaccines effectively reduced MenB disease in countries such as Norway, New Zealand, and France; however, these vaccines were highly specific for their targeted outbreak strain, did not elicit a durable immune response, and were ineffective for widespread use due to the diversity of MenB-disease-causing isolates. Recently developed recombinant protein-based MenB vaccines that target conserved surface proteins have the potential to induce a broader immune response against the diversity of disease-causing strains. Given the deleterious consequences and sporadic nature of MenB disease, the use of optimal vaccination strategies is crucial for prevention. Reactive vaccination strategies used in the past have significant limitations, including delayed implementation, substantial use of resources, and time constraints. The broad coverage potential of recombinant protein-based MenB vaccines suggests that routine use could result in a reduced burden of disease. Despite this, routine use of MenB vaccines is currently limited in practice.
Collapse
Affiliation(s)
- Paul Balmer
- Senior Medical Director, Pfizer Vaccines Medical Development and Scientific/Clinical Affairs, Pfizer Inc., 500 Arcola Road, Collegeville, PA, USA
| | - Laura J York
- Vice President, Global Meningococcal Vaccines, Pfizer Vaccines Medical Development and Scientific/Clinical Affairs, Pfizer Inc., 500 Arcola Road, Collegeville, PA, USA
| |
Collapse
|
42
|
Beeslaar J, Absalon J, Balmer P, Srivastava A, Maansson R, York LJ, Perez JL. Clinical data supporting a 2-dose schedule of MenB-FHbp, a bivalent meningococcal serogroup B vaccine, in adolescents and young adults. Vaccine 2018; 36:4004-4013. [PMID: 29861182 DOI: 10.1016/j.vaccine.2018.05.060] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 05/04/2018] [Accepted: 05/08/2018] [Indexed: 12/19/2022]
Abstract
Invasive meningococcal disease (IMD) caused by Neisseria meningitidis is a potentially devastating condition that can result in death and is associated with serious long-term sequelae in survivors. Vaccination is the preferred preventative strategy. Quadrivalent polysaccharide-based vaccines that protect against infection caused by meningococcal serogroups A, C, W, and Y are not effective against meningococcal serogroup B (MenB), which was responsible for approximately 60% and 35% of confirmed IMD cases in the European Union and the United States in 2016, respectively. A recombinant protein MenB vaccine (MenB-FHbp [bivalent rLP2086; Trumenba®]) has been approved for protection against MenB infection in persons 10-25 years of age in the United States and Canada and for individuals ≥10 years of age in the European Union and Australia. In these regions, MenB-FHbp is approved as a 2- or 3-dose primary vaccination schedule. This report will review the current evidence supporting administration of MenB-FHbp as a 2-dose primary vaccination schedule. Different contexts in which a 2- or 3-dose primary vaccination schedule might be preferred (eg, routine prospective vaccination vs outbreak control) are reviewed.
Collapse
Affiliation(s)
| | - Judith Absalon
- Pfizer Vaccine Clinical Research and Development, Pearl River, NY, USA
| | - Paul Balmer
- Pfizer Vaccine Clinical Research and Development, Collegeville, PA, USA
| | | | - Roger Maansson
- Pfizer Vaccine Clinical Research and Development, Collegeville, PA, USA
| | - Laura J York
- Pfizer Vaccines Medical Development, Scientific & Clinical Affairs, Collegeville, PA, USA
| | - John L Perez
- Pfizer Vaccine Clinical Research and Development, Collegeville, PA, USA
| |
Collapse
|
43
|
Hao L, Holden MTG, Wang X, Andrew L, Wellnitz S, Hu F, Whaley M, Sammons S, Knipe K, Frace M, McNamara LA, Liberator P, Anderson AS. Distinct evolutionary patterns of Neisseria meningitidis serogroup B disease outbreaks at two universities in the USA. Microb Genom 2018; 4. [PMID: 29616896 PMCID: PMC5989579 DOI: 10.1099/mgen.0.000155] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Neisseria meningitidis serogroup B (MnB) was responsible for two independent meningococcal disease outbreaks at universities in the USA during 2013. The first at University A in New Jersey included nine confirmed cases reported between March 2013 and March 2014. The second outbreak occurred at University B in California, with four confirmed cases during November 2013. The public health response to these outbreaks included the approval and deployment of a serogroup B meningococcal vaccine that was not yet licensed in the USA. This study investigated the use of whole-genome sequencing(WGS) to examine the genetic profile of the disease-causing outbreak isolates at each university. Comparative WGS revealed differences in evolutionary patterns between the two disease outbreaks. The University A outbreak isolates were very closely related, with differences primarily attributed to single nucleotide polymorphisms/insertion-deletion (SNP/indel) events. In contrast, the University B outbreak isolates segregated into two phylogenetic clades, differing in large part due to recombination events covering extensive regions (>30 kb) of the genome including virulence factors. This high-resolution comparison of two meningococcal disease outbreaks further demonstrates the genetic complexity of meningococcal bacteria as related to evolution and disease virulence.
Collapse
Affiliation(s)
- Li Hao
- 1Vaccine Research & Development, Pfizer Inc, 401 N. Middletown Rd, Pearl River, NY 10965, USA
| | | | - Xin Wang
- 3Division of Bacterial Diseases, Centers for Diseases Control and Prevention, Atlanta, Georgia, USA
| | - Lubomira Andrew
- 1Vaccine Research & Development, Pfizer Inc, 401 N. Middletown Rd, Pearl River, NY 10965, USA
| | - Sabine Wellnitz
- 1Vaccine Research & Development, Pfizer Inc, 401 N. Middletown Rd, Pearl River, NY 10965, USA
| | - Fang Hu
- 3Division of Bacterial Diseases, Centers for Diseases Control and Prevention, Atlanta, Georgia, USA
| | - Melissa Whaley
- 3Division of Bacterial Diseases, Centers for Diseases Control and Prevention, Atlanta, Georgia, USA
| | - Scott Sammons
- 3Division of Bacterial Diseases, Centers for Diseases Control and Prevention, Atlanta, Georgia, USA
| | - Kristen Knipe
- 3Division of Bacterial Diseases, Centers for Diseases Control and Prevention, Atlanta, Georgia, USA
| | - Mike Frace
- 3Division of Bacterial Diseases, Centers for Diseases Control and Prevention, Atlanta, Georgia, USA
| | - Lucy A McNamara
- 3Division of Bacterial Diseases, Centers for Diseases Control and Prevention, Atlanta, Georgia, USA
| | - Paul Liberator
- 1Vaccine Research & Development, Pfizer Inc, 401 N. Middletown Rd, Pearl River, NY 10965, USA
| | - Annaliesa S Anderson
- 1Vaccine Research & Development, Pfizer Inc, 401 N. Middletown Rd, Pearl River, NY 10965, USA
| |
Collapse
|
44
|
Predicting the Susceptibility of Meningococcal Serogroup B Isolates to Bactericidal Antibodies Elicited by Bivalent rLP2086, a Novel Prophylactic Vaccine. mBio 2018. [PMID: 29535195 PMCID: PMC5850321 DOI: 10.1128/mbio.00036-18] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Bivalent rLP2086 (Trumenba), a vaccine for prevention of Neisseria meningitidis serogroup B (NmB) disease, was licensed for use in adolescents and young adults after it was demonstrated that it elicits antibodies that initiate complement-mediated killing of invasive NmB isolates in a serum bactericidal assay with human complement (hSBA). The vaccine consists of two factor H binding proteins (fHBPs) representing divergent subfamilies to ensure broad coverage. Although it is the surrogate of efficacy, an hSBA is not suitable for testing large numbers of strains in local laboratories. Previously, an association between the in vitro fHBP surface expression level and the susceptibility of NmB isolates to killing was observed. Therefore, a flow cytometric meningococcal antigen surface expression (MEASURE) assay was developed and validated by using an antibody that binds to all fHBP variants from both fHBP subfamilies and accurately quantitates the level of fHBP expressed on the cell surface of NmB isolates with mean fluorescence intensity as the readout. Two collections of invasive NmB isolates (n = 1,814, n = 109) were evaluated in the assay, with the smaller set also tested in hSBAs using individual and pooled human serum samples from young adults vaccinated with bivalent rLP2086. From these data, an analysis based on fHBP variant prevalence in the larger 1,814-isolate set showed that >91% of all meningococcal serogroup B isolates expressed sufficient levels of fHBP to be susceptible to bactericidal killing by vaccine-induced antibodies.IMPORTANCE Bivalent rLP2086 (Trumenba) vaccine, composed of two factor H binding proteins (fHBPs), was recently licensed for the prevention of N. meningitidis serogroup B (NmB) disease in individuals 10 to 25 years old in the United States. This study evaluated a large collection of NmB isolates from the United States and Europe by using a flow cytometric MEASURE assay to quantitate the surface expression of the vaccine antigen fHBP. We find that expression levels and the proportion of strains above the level associated with susceptibility in an hSBA are generally consistent across these geographic regions. Thus, the assay can be used to predict which NmB isolates are susceptible to killing in the hSBA and therefore is able to demonstrate an fHBP vaccine-induced bactericidal response. This work significantly advances our understanding of the potential for bivalent rLP2086 to provide broad coverage against diverse invasive-disease-causing NmB isolates.
Collapse
|
45
|
Ostergaard L, Vesikari T, Absalon J, Beeslaar J, Ward BJ, Senders S, Eiden JJ, Jansen KU, Anderson AS, York LJ, Jones TR, Harris SL, O'Neill R, Radley D, Maansson R, Prégaldien JL, Ginis J, Staerke NB, Perez JL. A Bivalent Meningococcal B Vaccine in Adolescents and Young Adults. N Engl J Med 2017; 377:2349-2362. [PMID: 29236639 DOI: 10.1056/nejmoa1614474] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND MenB-FHbp is a licensed meningococcal B vaccine targeting factor H-binding protein. Two phase 3 studies assessed the safety of the vaccine and its immunogenicity against diverse strains of group B meningococcus. METHODS We randomly assigned 3596 adolescents (10 to 18 years of age) to receive MenB-FHbp or hepatitis A virus vaccine and saline and assigned 3304 young adults (18 to 25 years of age) to receive MenB-FHbp or saline at baseline, 2 months, and 6 months. Immunogenicity was assessed in serum bactericidal assays that included human complement (hSBAs). We used 14 meningococcal B test strains that expressed vaccine-heterologous factor H-binding proteins representative of meningococcal B epidemiologic diversity; an hSBA titer of at least 1:4 is the accepted correlate of protection. The five primary end points were the proportion of participants who had an increase in their hSBA titer for each of 4 primary strains by a factor of 4 or more and the proportion of those who had an hSBA titer at least as high as the lower limit of quantitation (1:8 or 1:16) for all 4 strains combined after dose 3. We also assessed the hSBA responses to the primary strains after dose 2; hSBA responses to the 10 additional strains after doses 2 and 3 were assessed in a subgroup of participants only. Safety was assessed in participants who received at least one dose. RESULTS In the modified intention-to-treat population, the percentage of adolescents who had an increase in the hSBA titer by a factor of 4 or more against each primary strain ranged from 56.0 to 85.3% after dose 2 and from 78.8 to 90.2% after dose 3; the percentages of young adults ranged from 54.6 to 85.6% and 78.9 to 89.7%, after doses 2 and 3, respectively. Composite responses after doses 2 and 3 in adolescents were 53.7% and 82.7%, respectively, and those in young adults were 63.3% and 84.5%, respectively. Responses to the 4 primary strains were predictive of responses to the 10 additional strains. Most of those who received MenB-FHbp reported mild or moderate pain at the vaccination site. CONCLUSIONS MenB-FHbp elicited bactericidal responses against diverse meningococcal B strains after doses 2 and 3 and was associated with more reactions at the injection site than the hepatitis A virus vaccine and saline. (Funded by Pfizer; ClinicalTrials.gov numbers, NCT01830855 and NCT01352845 ).
Collapse
Affiliation(s)
- Lars Ostergaard
- From Aarhus University Hospital, Aarhus, Denmark (L.O., N.B.S.); Vaccine Research Center, University of Tampere Medical School, Tampere, Finland (T.V.); Pfizer Vaccine Clinical Research and Development (J.A., J.J.E.) and Pfizer Vaccine Research and Development (K.U.J., A.S.A., T.R.J., S.L.H., R.O.), Pearl River, NY; Pfizer Vaccine Clinical Research and Development, Hurley, United Kingdom (J.B.); Research Institute of the McGill University Health Center, Montreal (B.J.W.); Senders Pediatrics, South Euclid, OH (S.S.); Pfizer Vaccine Medical Development, Scientific and Clinical Affairs (L.J.Y.) and Pfizer Vaccine Clinical Research and Development (D.R., R.M., J.G., J.L.P.), Collegeville, PA; and Pfizer Vaccine Clinical Research and Development, Brussels (J.-L.P.)
| | - Timo Vesikari
- From Aarhus University Hospital, Aarhus, Denmark (L.O., N.B.S.); Vaccine Research Center, University of Tampere Medical School, Tampere, Finland (T.V.); Pfizer Vaccine Clinical Research and Development (J.A., J.J.E.) and Pfizer Vaccine Research and Development (K.U.J., A.S.A., T.R.J., S.L.H., R.O.), Pearl River, NY; Pfizer Vaccine Clinical Research and Development, Hurley, United Kingdom (J.B.); Research Institute of the McGill University Health Center, Montreal (B.J.W.); Senders Pediatrics, South Euclid, OH (S.S.); Pfizer Vaccine Medical Development, Scientific and Clinical Affairs (L.J.Y.) and Pfizer Vaccine Clinical Research and Development (D.R., R.M., J.G., J.L.P.), Collegeville, PA; and Pfizer Vaccine Clinical Research and Development, Brussels (J.-L.P.)
| | - Judith Absalon
- From Aarhus University Hospital, Aarhus, Denmark (L.O., N.B.S.); Vaccine Research Center, University of Tampere Medical School, Tampere, Finland (T.V.); Pfizer Vaccine Clinical Research and Development (J.A., J.J.E.) and Pfizer Vaccine Research and Development (K.U.J., A.S.A., T.R.J., S.L.H., R.O.), Pearl River, NY; Pfizer Vaccine Clinical Research and Development, Hurley, United Kingdom (J.B.); Research Institute of the McGill University Health Center, Montreal (B.J.W.); Senders Pediatrics, South Euclid, OH (S.S.); Pfizer Vaccine Medical Development, Scientific and Clinical Affairs (L.J.Y.) and Pfizer Vaccine Clinical Research and Development (D.R., R.M., J.G., J.L.P.), Collegeville, PA; and Pfizer Vaccine Clinical Research and Development, Brussels (J.-L.P.)
| | - Johannes Beeslaar
- From Aarhus University Hospital, Aarhus, Denmark (L.O., N.B.S.); Vaccine Research Center, University of Tampere Medical School, Tampere, Finland (T.V.); Pfizer Vaccine Clinical Research and Development (J.A., J.J.E.) and Pfizer Vaccine Research and Development (K.U.J., A.S.A., T.R.J., S.L.H., R.O.), Pearl River, NY; Pfizer Vaccine Clinical Research and Development, Hurley, United Kingdom (J.B.); Research Institute of the McGill University Health Center, Montreal (B.J.W.); Senders Pediatrics, South Euclid, OH (S.S.); Pfizer Vaccine Medical Development, Scientific and Clinical Affairs (L.J.Y.) and Pfizer Vaccine Clinical Research and Development (D.R., R.M., J.G., J.L.P.), Collegeville, PA; and Pfizer Vaccine Clinical Research and Development, Brussels (J.-L.P.)
| | - Brian J Ward
- From Aarhus University Hospital, Aarhus, Denmark (L.O., N.B.S.); Vaccine Research Center, University of Tampere Medical School, Tampere, Finland (T.V.); Pfizer Vaccine Clinical Research and Development (J.A., J.J.E.) and Pfizer Vaccine Research and Development (K.U.J., A.S.A., T.R.J., S.L.H., R.O.), Pearl River, NY; Pfizer Vaccine Clinical Research and Development, Hurley, United Kingdom (J.B.); Research Institute of the McGill University Health Center, Montreal (B.J.W.); Senders Pediatrics, South Euclid, OH (S.S.); Pfizer Vaccine Medical Development, Scientific and Clinical Affairs (L.J.Y.) and Pfizer Vaccine Clinical Research and Development (D.R., R.M., J.G., J.L.P.), Collegeville, PA; and Pfizer Vaccine Clinical Research and Development, Brussels (J.-L.P.)
| | - Shelly Senders
- From Aarhus University Hospital, Aarhus, Denmark (L.O., N.B.S.); Vaccine Research Center, University of Tampere Medical School, Tampere, Finland (T.V.); Pfizer Vaccine Clinical Research and Development (J.A., J.J.E.) and Pfizer Vaccine Research and Development (K.U.J., A.S.A., T.R.J., S.L.H., R.O.), Pearl River, NY; Pfizer Vaccine Clinical Research and Development, Hurley, United Kingdom (J.B.); Research Institute of the McGill University Health Center, Montreal (B.J.W.); Senders Pediatrics, South Euclid, OH (S.S.); Pfizer Vaccine Medical Development, Scientific and Clinical Affairs (L.J.Y.) and Pfizer Vaccine Clinical Research and Development (D.R., R.M., J.G., J.L.P.), Collegeville, PA; and Pfizer Vaccine Clinical Research and Development, Brussels (J.-L.P.)
| | - Joseph J Eiden
- From Aarhus University Hospital, Aarhus, Denmark (L.O., N.B.S.); Vaccine Research Center, University of Tampere Medical School, Tampere, Finland (T.V.); Pfizer Vaccine Clinical Research and Development (J.A., J.J.E.) and Pfizer Vaccine Research and Development (K.U.J., A.S.A., T.R.J., S.L.H., R.O.), Pearl River, NY; Pfizer Vaccine Clinical Research and Development, Hurley, United Kingdom (J.B.); Research Institute of the McGill University Health Center, Montreal (B.J.W.); Senders Pediatrics, South Euclid, OH (S.S.); Pfizer Vaccine Medical Development, Scientific and Clinical Affairs (L.J.Y.) and Pfizer Vaccine Clinical Research and Development (D.R., R.M., J.G., J.L.P.), Collegeville, PA; and Pfizer Vaccine Clinical Research and Development, Brussels (J.-L.P.)
| | - Kathrin U Jansen
- From Aarhus University Hospital, Aarhus, Denmark (L.O., N.B.S.); Vaccine Research Center, University of Tampere Medical School, Tampere, Finland (T.V.); Pfizer Vaccine Clinical Research and Development (J.A., J.J.E.) and Pfizer Vaccine Research and Development (K.U.J., A.S.A., T.R.J., S.L.H., R.O.), Pearl River, NY; Pfizer Vaccine Clinical Research and Development, Hurley, United Kingdom (J.B.); Research Institute of the McGill University Health Center, Montreal (B.J.W.); Senders Pediatrics, South Euclid, OH (S.S.); Pfizer Vaccine Medical Development, Scientific and Clinical Affairs (L.J.Y.) and Pfizer Vaccine Clinical Research and Development (D.R., R.M., J.G., J.L.P.), Collegeville, PA; and Pfizer Vaccine Clinical Research and Development, Brussels (J.-L.P.)
| | - Annaliesa S Anderson
- From Aarhus University Hospital, Aarhus, Denmark (L.O., N.B.S.); Vaccine Research Center, University of Tampere Medical School, Tampere, Finland (T.V.); Pfizer Vaccine Clinical Research and Development (J.A., J.J.E.) and Pfizer Vaccine Research and Development (K.U.J., A.S.A., T.R.J., S.L.H., R.O.), Pearl River, NY; Pfizer Vaccine Clinical Research and Development, Hurley, United Kingdom (J.B.); Research Institute of the McGill University Health Center, Montreal (B.J.W.); Senders Pediatrics, South Euclid, OH (S.S.); Pfizer Vaccine Medical Development, Scientific and Clinical Affairs (L.J.Y.) and Pfizer Vaccine Clinical Research and Development (D.R., R.M., J.G., J.L.P.), Collegeville, PA; and Pfizer Vaccine Clinical Research and Development, Brussels (J.-L.P.)
| | - Laura J York
- From Aarhus University Hospital, Aarhus, Denmark (L.O., N.B.S.); Vaccine Research Center, University of Tampere Medical School, Tampere, Finland (T.V.); Pfizer Vaccine Clinical Research and Development (J.A., J.J.E.) and Pfizer Vaccine Research and Development (K.U.J., A.S.A., T.R.J., S.L.H., R.O.), Pearl River, NY; Pfizer Vaccine Clinical Research and Development, Hurley, United Kingdom (J.B.); Research Institute of the McGill University Health Center, Montreal (B.J.W.); Senders Pediatrics, South Euclid, OH (S.S.); Pfizer Vaccine Medical Development, Scientific and Clinical Affairs (L.J.Y.) and Pfizer Vaccine Clinical Research and Development (D.R., R.M., J.G., J.L.P.), Collegeville, PA; and Pfizer Vaccine Clinical Research and Development, Brussels (J.-L.P.)
| | - Thomas R Jones
- From Aarhus University Hospital, Aarhus, Denmark (L.O., N.B.S.); Vaccine Research Center, University of Tampere Medical School, Tampere, Finland (T.V.); Pfizer Vaccine Clinical Research and Development (J.A., J.J.E.) and Pfizer Vaccine Research and Development (K.U.J., A.S.A., T.R.J., S.L.H., R.O.), Pearl River, NY; Pfizer Vaccine Clinical Research and Development, Hurley, United Kingdom (J.B.); Research Institute of the McGill University Health Center, Montreal (B.J.W.); Senders Pediatrics, South Euclid, OH (S.S.); Pfizer Vaccine Medical Development, Scientific and Clinical Affairs (L.J.Y.) and Pfizer Vaccine Clinical Research and Development (D.R., R.M., J.G., J.L.P.), Collegeville, PA; and Pfizer Vaccine Clinical Research and Development, Brussels (J.-L.P.)
| | - Shannon L Harris
- From Aarhus University Hospital, Aarhus, Denmark (L.O., N.B.S.); Vaccine Research Center, University of Tampere Medical School, Tampere, Finland (T.V.); Pfizer Vaccine Clinical Research and Development (J.A., J.J.E.) and Pfizer Vaccine Research and Development (K.U.J., A.S.A., T.R.J., S.L.H., R.O.), Pearl River, NY; Pfizer Vaccine Clinical Research and Development, Hurley, United Kingdom (J.B.); Research Institute of the McGill University Health Center, Montreal (B.J.W.); Senders Pediatrics, South Euclid, OH (S.S.); Pfizer Vaccine Medical Development, Scientific and Clinical Affairs (L.J.Y.) and Pfizer Vaccine Clinical Research and Development (D.R., R.M., J.G., J.L.P.), Collegeville, PA; and Pfizer Vaccine Clinical Research and Development, Brussels (J.-L.P.)
| | - Robert O'Neill
- From Aarhus University Hospital, Aarhus, Denmark (L.O., N.B.S.); Vaccine Research Center, University of Tampere Medical School, Tampere, Finland (T.V.); Pfizer Vaccine Clinical Research and Development (J.A., J.J.E.) and Pfizer Vaccine Research and Development (K.U.J., A.S.A., T.R.J., S.L.H., R.O.), Pearl River, NY; Pfizer Vaccine Clinical Research and Development, Hurley, United Kingdom (J.B.); Research Institute of the McGill University Health Center, Montreal (B.J.W.); Senders Pediatrics, South Euclid, OH (S.S.); Pfizer Vaccine Medical Development, Scientific and Clinical Affairs (L.J.Y.) and Pfizer Vaccine Clinical Research and Development (D.R., R.M., J.G., J.L.P.), Collegeville, PA; and Pfizer Vaccine Clinical Research and Development, Brussels (J.-L.P.)
| | - David Radley
- From Aarhus University Hospital, Aarhus, Denmark (L.O., N.B.S.); Vaccine Research Center, University of Tampere Medical School, Tampere, Finland (T.V.); Pfizer Vaccine Clinical Research and Development (J.A., J.J.E.) and Pfizer Vaccine Research and Development (K.U.J., A.S.A., T.R.J., S.L.H., R.O.), Pearl River, NY; Pfizer Vaccine Clinical Research and Development, Hurley, United Kingdom (J.B.); Research Institute of the McGill University Health Center, Montreal (B.J.W.); Senders Pediatrics, South Euclid, OH (S.S.); Pfizer Vaccine Medical Development, Scientific and Clinical Affairs (L.J.Y.) and Pfizer Vaccine Clinical Research and Development (D.R., R.M., J.G., J.L.P.), Collegeville, PA; and Pfizer Vaccine Clinical Research and Development, Brussels (J.-L.P.)
| | - Roger Maansson
- From Aarhus University Hospital, Aarhus, Denmark (L.O., N.B.S.); Vaccine Research Center, University of Tampere Medical School, Tampere, Finland (T.V.); Pfizer Vaccine Clinical Research and Development (J.A., J.J.E.) and Pfizer Vaccine Research and Development (K.U.J., A.S.A., T.R.J., S.L.H., R.O.), Pearl River, NY; Pfizer Vaccine Clinical Research and Development, Hurley, United Kingdom (J.B.); Research Institute of the McGill University Health Center, Montreal (B.J.W.); Senders Pediatrics, South Euclid, OH (S.S.); Pfizer Vaccine Medical Development, Scientific and Clinical Affairs (L.J.Y.) and Pfizer Vaccine Clinical Research and Development (D.R., R.M., J.G., J.L.P.), Collegeville, PA; and Pfizer Vaccine Clinical Research and Development, Brussels (J.-L.P.)
| | - Jean-Louis Prégaldien
- From Aarhus University Hospital, Aarhus, Denmark (L.O., N.B.S.); Vaccine Research Center, University of Tampere Medical School, Tampere, Finland (T.V.); Pfizer Vaccine Clinical Research and Development (J.A., J.J.E.) and Pfizer Vaccine Research and Development (K.U.J., A.S.A., T.R.J., S.L.H., R.O.), Pearl River, NY; Pfizer Vaccine Clinical Research and Development, Hurley, United Kingdom (J.B.); Research Institute of the McGill University Health Center, Montreal (B.J.W.); Senders Pediatrics, South Euclid, OH (S.S.); Pfizer Vaccine Medical Development, Scientific and Clinical Affairs (L.J.Y.) and Pfizer Vaccine Clinical Research and Development (D.R., R.M., J.G., J.L.P.), Collegeville, PA; and Pfizer Vaccine Clinical Research and Development, Brussels (J.-L.P.)
| | - John Ginis
- From Aarhus University Hospital, Aarhus, Denmark (L.O., N.B.S.); Vaccine Research Center, University of Tampere Medical School, Tampere, Finland (T.V.); Pfizer Vaccine Clinical Research and Development (J.A., J.J.E.) and Pfizer Vaccine Research and Development (K.U.J., A.S.A., T.R.J., S.L.H., R.O.), Pearl River, NY; Pfizer Vaccine Clinical Research and Development, Hurley, United Kingdom (J.B.); Research Institute of the McGill University Health Center, Montreal (B.J.W.); Senders Pediatrics, South Euclid, OH (S.S.); Pfizer Vaccine Medical Development, Scientific and Clinical Affairs (L.J.Y.) and Pfizer Vaccine Clinical Research and Development (D.R., R.M., J.G., J.L.P.), Collegeville, PA; and Pfizer Vaccine Clinical Research and Development, Brussels (J.-L.P.)
| | - Nina B Staerke
- From Aarhus University Hospital, Aarhus, Denmark (L.O., N.B.S.); Vaccine Research Center, University of Tampere Medical School, Tampere, Finland (T.V.); Pfizer Vaccine Clinical Research and Development (J.A., J.J.E.) and Pfizer Vaccine Research and Development (K.U.J., A.S.A., T.R.J., S.L.H., R.O.), Pearl River, NY; Pfizer Vaccine Clinical Research and Development, Hurley, United Kingdom (J.B.); Research Institute of the McGill University Health Center, Montreal (B.J.W.); Senders Pediatrics, South Euclid, OH (S.S.); Pfizer Vaccine Medical Development, Scientific and Clinical Affairs (L.J.Y.) and Pfizer Vaccine Clinical Research and Development (D.R., R.M., J.G., J.L.P.), Collegeville, PA; and Pfizer Vaccine Clinical Research and Development, Brussels (J.-L.P.)
| | - John L Perez
- From Aarhus University Hospital, Aarhus, Denmark (L.O., N.B.S.); Vaccine Research Center, University of Tampere Medical School, Tampere, Finland (T.V.); Pfizer Vaccine Clinical Research and Development (J.A., J.J.E.) and Pfizer Vaccine Research and Development (K.U.J., A.S.A., T.R.J., S.L.H., R.O.), Pearl River, NY; Pfizer Vaccine Clinical Research and Development, Hurley, United Kingdom (J.B.); Research Institute of the McGill University Health Center, Montreal (B.J.W.); Senders Pediatrics, South Euclid, OH (S.S.); Pfizer Vaccine Medical Development, Scientific and Clinical Affairs (L.J.Y.) and Pfizer Vaccine Clinical Research and Development (D.R., R.M., J.G., J.L.P.), Collegeville, PA; and Pfizer Vaccine Clinical Research and Development, Brussels (J.-L.P.)
| |
Collapse
|
46
|
Breadth and Duration of Meningococcal Serum Bactericidal Activity in Health Care Workers and Microbiologists Immunized with the MenB-FHbp Vaccine. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2017; 24:CVI.00121-17. [PMID: 28566335 DOI: 10.1128/cvi.00121-17] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Accepted: 05/23/2017] [Indexed: 12/17/2022]
Abstract
MenB-FHbp is a meningococcal serogroup B vaccine with two factor H binding protein (FHbp) antigens from subfamilies A and B. For licensure, efficacy was inferred from serum bactericidal antibody (SBA) responses to four reference strains. Only limited information is available on the breadth or duration of protective SBA responses to genetically diverse disease-causing strains. Seventeen health care or laboratory workers were immunized with two (n = 2) or three (n = 15) doses of MenB-FHbp at 0, 2, and 6 months. SBA levels were measured against 14 serogroup B case isolates, including 6 from U.S. college outbreaks and 2 from Quebec during hyperendemic disease. Compared with preimmunization titers, the proportion of subjects with ≥4-fold increases in SBA titer 1 month after 2 doses of vaccine ranged from 35% to 94% for six isolates with FHbp subfamily A and from 24% to 76% for eight isolates with subfamily B FHbp. The respective proportions with ≥4-fold titer increases at 1 month after dose 3 were 73% to 100% and 67% to 100%. At that time point, the proportion of subjects with titers of ≥1:4 (presumed sufficient for short-term protection) ranged from 93% to 100% for all 14 isolates. By 9 to 11 months after dose 3, 50% or fewer of the subjects with follow-up sera had protective titers of ≥1:4 for 4 of 9 isolates tested. Three doses of MenB-FHbp elicited short-term protective SBA responses to diverse disease-causing serogroup B strains. For some strains, serum titers declined to <1:4 by 9 to 11 months, which raises concerns about the duration of broad, long-term protection. (This study has been registered at ClinicalTrials.gov under registration no. NCT02569632.).
Collapse
|
47
|
Predicted vs observed effectiveness of outer membrane vesicle (OMV) vaccines against meningococcal serogroup B disease: Systematic review. J Infect 2017; 75:81-94. [DOI: 10.1016/j.jinf.2017.05.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 03/27/2017] [Accepted: 05/03/2017] [Indexed: 11/18/2022]
|
48
|
Abstract
The majority of invasive meningococcal disease (IMD) in the developed world is caused by capsular group B Neisseria meningitidis, however success with vaccination against organisms bearing this capsule has previously been restricted to control of geographically limited clonal outbreaks. As we enter a new era, with the first routine program underway to control endemic group B meningococcal disease for infants in the UK, it is timely to review the key landmarks in group B vaccine development, and discuss the issues determining whether control of endemic group B disease will be achieved. Evidence of a reduction in carriage acquisition of invasive group B meningococcal strains, after vaccination among adolescents, is imperative if routine immunization is to drive population control of disease beyond those who are vaccinated (i.e. through herd immunity). The need for multiple doses to generate a sufficiently protective response and reactogenicity remain significant problems with the new generation of vaccines. Despite these limitations, early data from the UK indicate that new group B meningococcal vaccines have the potential to have a major impact on meningococcal disease, and to provide new insight into how we might do better in the future.
Collapse
Affiliation(s)
- N Y Wang
- a School of Medicine , Monash University , Melbourne , Australia.,b Department of Paediatrics , Oxford Vaccine Group , Oxford , UK
| | - A J Pollard
- b Department of Paediatrics , Oxford Vaccine Group , Oxford , UK.,c NIHR Oxford Biomedical Research Centre, University of Oxford , Oxford , UK
| |
Collapse
|
49
|
Costoya L, Marzoa J, Ferreirós C, Criado MT. Liposomes or traditional adjuvants: induction of bactericidal activity by the macrophage infectivity potentiator protein (Mip) of Neisseria meningitidis. APMIS 2017; 125:725-731. [PMID: 28543600 DOI: 10.1111/apm.12709] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 03/20/2017] [Indexed: 01/10/2023]
Abstract
Currently, one of the main approaches to achieve a vaccine for serogroup B Neisseria meningitidis is based on outer membrane proteins with low antigenic variability among strains. Since these proteins tend to be minor components of the outer membrane, recombinant production is required to obtain them in sufficient amounts for evaluation and development of vaccines. In this study, we analysed the ability of recombinant macrophage infectivity potentiator (rMip) protein to induce protective bactericidal activity in mice. The rMip protein was cloned from N. meningitidis strain H44/76 and was used to immunise mice, and the sera obtained were tested against the homologous and several heterologous N. meningitidis strains. The sera were obtained using the rMip alone, with adjuvant Al(OH)3 , or after inclusion into liposomes. Bactericidal activity was variable depending on the strain, although high titres were seen against strains H44/76 and NmP27. Liposomes enhanced fourfold the reactivity against the homologous strain. The results presented suggest that the rMip protein should be considered a promising candidate for the improvement of future protein-based vaccines.
Collapse
Affiliation(s)
- Liliana Costoya
- Departamento de Microbioloxía e Parasitoloxía, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Juan Marzoa
- Departamento de Microbioloxía e Parasitoloxía, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Carlos Ferreirós
- Departamento de Microbioloxía e Parasitoloxía, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Maria Teresa Criado
- Departamento de Microbioloxía e Parasitoloxía, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| |
Collapse
|
50
|
Toneatto D, Pizza M, Masignani V, Rappuoli R. Emerging experience with meningococcal serogroup B protein vaccines. Expert Rev Vaccines 2017; 16:433-451. [PMID: 28375029 DOI: 10.1080/14760584.2017.1308828] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION The successful development of two broadly protective vaccines targeting Neisseria meningitidis serogroup B (MenB); 4CMenB and rLP2086, is the most significant recent advance in meningococcal disease prevention. Areas covered: Here we review the principles underlying the development of each vaccine and the novel methods used to estimate vaccine coverage. We update clinical and post-licensure experience with 4CMenB and rLP2086. Expert commentary: The immunogenicity and acceptable safety profile of 4CMenB and rLP2086 has been demonstrated in clinical trials. Continuing uncertainties exist around the appropriate age groups to be immunized, the degree and duration of efficacy, and the impact on nasopharyngeal carriage which has implications for strategies to interrupt transmission and maximize herd protection effects. Universal vaccination programs such as those undertaken in Quebec and the United Kingdom are providing important information on these issues. The potential for MenB vaccines to prevent infection by other serogroups appears promising, and the impact of MenB vaccines on other pathogenic neisserial species with similar surface proteins warrants further investigation.
Collapse
|