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
|
Treviño MA, Amankwah KA, Fernandez D, Weston SA, Stewart CJ, Gallardo JM, Shahgholi M, Sharaf NG. Expression, purification, and characterization of diacylated Lipo-YcjN from Escherichia coli. J Biol Chem 2024; 300:107853. [PMID: 39362470 PMCID: PMC11543891 DOI: 10.1016/j.jbc.2024.107853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/17/2024] [Accepted: 09/25/2024] [Indexed: 10/05/2024] Open
Abstract
YcjN is a putative substrate binding protein expressed from a cluster of genes involved in carbohydrate import and metabolism in Escherichia coli. Here, we determine the crystal structure of YcjN to a resolution of 1.95 Å, revealing that its three-dimensional structure is similar to substrate binding proteins in subcluster D-I, which includes the well-characterized maltose binding protein. Furthermore, we found that recombinant overexpression of YcjN results in the formation of a lipidated form of YcjN that is posttranslationally diacylated at cysteine 21. Comparisons of size-exclusion chromatography profiles and dynamic light scattering measurements of lipidated and nonlipidated YcjN proteins suggest that lipidated YcjN aggregates in solution via its lipid moiety. Additionally, bioinformatic analysis indicates that YcjN-like proteins may exist in both Bacteria and Archaea, potentially in both lipidated and nonlipidated forms. Together, our results provide a better understanding of the aggregation properties of recombinantly expressed bacterial lipoproteins in solution and establish a foundation for future studies that aim to elucidate the role of these proteins in bacterial physiology.
Collapse
Affiliation(s)
- Matthew A Treviño
- Department of Biology, Stanford University, Stanford, California, USA
| | - Kofi A Amankwah
- Department of Biology, Stanford University, Stanford, California, USA
| | - Daniel Fernandez
- Macromolecular Structure Knowledge Center (MSKC) at Sarafan ChEM-H, Stanford University, Stanford, California, USA; Sarafan ChEM-H Institute, Stanford University, Stanford, California, USA
| | - Scott A Weston
- Department of Biology, Stanford University, Stanford, California, USA
| | - Claire J Stewart
- Department of Biology, Stanford University, Stanford, California, USA
| | | | - Mona Shahgholi
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena California, USA
| | - Naima G Sharaf
- Department of Biology, Stanford University, Stanford, California, USA.
| |
Collapse
|
3
|
Treviño MA, Amankwah K, Fernandez D, Weston S, Stewart CJ, Gallardo JM, Shahgholi M, Sharaf NG. Expression, purification, and characterization of diacylated Lipo-YcjN from Escherichia coli. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.05.611266. [PMID: 39282304 PMCID: PMC11398462 DOI: 10.1101/2024.09.05.611266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/09/2024]
Abstract
YcjN is a putative substrate-binding protein expressed from a cluster of genes involved in carbohydrate import and metabolism in Escherichia coli. Here, we determine the crystal structure of YcjN to a resolution of 1.95 Å, revealing that its three-dimensional structure is similar to substrate binding proteins in subcluster D-I, which includes the well-characterized maltose binding protein (MBP). Furthermore, we found that recombinant overexpression of YcjN results in the formation of a lipidated form of YcjN that is posttranslationally diacylated at cysteine 21. Comparisons of size-exclusion chromatography profiles and dynamic light scattering measurements of lipidated and non-lipidated YcjN proteins suggest that lipidated YcjN aggregates in solution via its lipid moiety. Additionally, bioinformatic analysis indicates that YcjN-like proteins may exist in both Bacteria and Archaea, potentially in both lipidated and non-lipidated forms. Together, our results provide a better understanding of the aggregation properties of recombinantly expressed bacterial lipoproteins in solution and establish a foundation for future studies that aim to elucidate the role of these proteins in bacterial physiology.
Collapse
Affiliation(s)
| | - Kofi Amankwah
- Department of Biology, Stanford University, Stanford CA 94305
| | - Daniel Fernandez
- Macromolecular Structure Knowledge Center (MSKC) at Sarafan ChEM-H
- Sarafan ChEM-H, Stanford ChEM-H Building, Stanford University, Stanford CA 94305
| | - Scott Weston
- Department of Biology, Stanford University, Stanford CA 94305
| | | | | | - Mona Shahgholi
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena CA
| | - Naima G. Sharaf
- Department of Biology, Stanford University, Stanford CA 94305
| |
Collapse
|
4
|
Croucher NJ. Immune interface interference vaccines: An evolution-informed approach to anti-bacterial vaccine design. Microb Biotechnol 2024; 17:e14446. [PMID: 38536702 PMCID: PMC10970203 DOI: 10.1111/1751-7915.14446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 03/01/2024] [Indexed: 10/17/2024] Open
Abstract
Developing protein-based vaccines against bacteria has proved much more challenging than producing similar immunisations against viruses. Currently, anti-bacterial vaccines are designed using methods based on reverse vaccinology. These identify broadly conserved, immunogenic proteins using a combination of genomic and high-throughput laboratory data. While this approach has successfully generated multiple rationally designed formulations that show promising immunogenicity in animal models, few have been licensed. The difficulty of inducing protective immunity in humans with such vaccines mirrors the ability of many bacteria to recolonise individuals despite recognition by natural polyvalent antibody repertoires. As bacteria express too many antigens to evade all adaptive immune responses through mutation, they must instead inhibit the efficacy of such host defences through expressing surface structures that interface with the immune system. Therefore, 'immune interface interference' (I3) vaccines that target these features should synergistically directly target bacteria and prevent them from inhibiting responses to other surface antigens. This approach may help us understand the efficacy of the two recently introduced immunisations against serotype B meningococci, which both target the Factor H-binding protein (fHbp) that inhibits complement deposition on the bacterial surface. Therefore, I3 vaccine designs may help overcome the current challenges of developing protein-based vaccines to prevent bacterial infections.
Collapse
Affiliation(s)
- Nicholas J. Croucher
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, School of Public HealthImperial College LondonLondonUK
| |
Collapse
|
5
|
Savitskaya VY, Dolinnaya NG, Strekalovskikh VV, Peskovatskova ES, Snyga VG, Trefilov VS, Monakhova MV, Kubareva EA. Bioinformatics Analysis of Global Diversity in Meningococcal Vaccine Antigens over the Past 10 Years: Vaccine Efficacy Prognosis. Med Sci (Basel) 2023; 11:76. [PMID: 38132917 PMCID: PMC10744425 DOI: 10.3390/medsci11040076] [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: 09/30/2023] [Revised: 11/17/2023] [Accepted: 11/27/2023] [Indexed: 12/23/2023] Open
Abstract
Neisseria meningitidis (N. meningitidis) serogroup B (MenB) is the leading cause of invasive meningococcal disease worldwide. The pathogen has a wide range of virulence factors, which are potential vaccine components. Studying the genetic variability of antigens within a population, especially their long-term persistence, is necessary to develop new vaccines and predict the effectiveness of existing ones. The multicomponent 4CMenB vaccine (Bexsero), used since 2014, contains three major genome-derived recombinant proteins: factor H-binding protein (fHbp), Neisserial Heparin-Binding Antigen (NHBA) and Neisserial adhesin A (NadA). Here, we assessed the prevalence and sequence variations of these vaccine antigens in a panel of 5667 meningococcal isolates collected worldwide over the past 10 years and deposited in the PubMLST database. Using multiple amino acid sequence alignments and Random Forest Classifier machine learning methods, we estimated the potential strain coverage of fHbp and NHBA vaccine variants (51 and about 25%, respectively); the NadA antigen sequence was found in only 18% of MenB genomes analyzed, but cross-reactive variants were present in less than 1% of isolates. Based on our findings, we proposed various strategies to improve the 4CMenB vaccine and broaden the coverage of N. meningitidis strains.
Collapse
Affiliation(s)
- Viktoriia Yu. Savitskaya
- Department of Chemistry, Lomonosov Moscow State University, Leninskie Gory 1, Moscow 119991, Russia
| | - Nina G. Dolinnaya
- Department of Chemistry, Lomonosov Moscow State University, Leninskie Gory 1, Moscow 119991, Russia
| | - Vadim V. Strekalovskikh
- Department of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Leninskie Gory 1, Moscow 119234, Russia; (V.V.S.); (E.S.P.)
| | - Elizaveta S. Peskovatskova
- Department of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Leninskie Gory 1, Moscow 119234, Russia; (V.V.S.); (E.S.P.)
| | - Viktoriia G. Snyga
- Department of Chemistry, Lomonosov Moscow State University, Leninskie Gory 1, Moscow 119991, Russia
| | - Vadim S. Trefilov
- Department of Chemistry, Lomonosov Moscow State University, Leninskie Gory 1, Moscow 119991, Russia
| | - Mayya V. Monakhova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Leninskie Gory 1, Moscow 119992, Russia
| | - Elena A. Kubareva
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Leninskie Gory 1, Moscow 119992, Russia
| |
Collapse
|
6
|
Yee WX, Barnes G, Lavender H, Tang CM. Meningococcal factor H-binding protein: implications for disease susceptibility, virulence, and vaccines. Trends Microbiol 2023; 31:805-815. [PMID: 36941192 PMCID: PMC10914675 DOI: 10.1016/j.tim.2023.02.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/23/2023] [Accepted: 02/24/2023] [Indexed: 03/23/2023]
Abstract
Neisseria meningitidis is a human-adapted pathogen that causes meningitis and sepsis worldwide. N. meningitidis factor H-binding protein (fHbp) provides a mechanism for immune evasion by binding human complement factor H (CFH) to protect it from complement-mediated killing. Here, we discuss features of fHbp which enable it to engage human CFH (hCFH), and the regulation of fHbp expression. Studies of host susceptibility and bacterial genome-wide association studies (GWAS) highlight the importance of the interaction between fHbp and CFH and other complement factors, such as CFHR3, on the development of invasive meningococcal disease (IMD). Understanding the basis of fHbp:CFH interactions has also informed the design of next-generation vaccines as fHbp is a protective antigen. Structure-informed refinement of fHbp vaccines will help to combat the threat posed by the meningococcus, and accelerate the elimination of IMD.
Collapse
Affiliation(s)
- Wearn-Xin Yee
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
| | - Grace Barnes
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
| | - Hayley Lavender
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK.
| | - Christoph M Tang
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK.
| |
Collapse
|
7
|
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
|
8
|
Dold C, Marsay L, Wang N, Silva-Reyes L, Clutterbuck E, Paterson GK, Sharkey K, Wyllie D, Beernink PT, Hill AV, Pollard AJ, Rollier CS. An adenoviral-vectored vaccine confers seroprotection against capsular group B meningococcal disease. Sci Transl Med 2023; 15:eade3901. [PMID: 37343082 DOI: 10.1126/scitranslmed.ade3901] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 05/30/2023] [Indexed: 06/23/2023]
Abstract
Adenoviral-vectored vaccines are licensed for prevention of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and Ebola virus, but, for bacterial proteins, expression in a eukaryotic cell may affect the antigen's localization and conformation or lead to unwanted glycosylation. Here, we investigated the potential use of an adenoviral-vectored vaccine platform for capsular group B meningococcus (MenB). Vector-based candidate vaccines expressing MenB antigen factor H binding protein (fHbp) were generated, and immunogenicity was assessed in mouse models, including the functional antibody response by serum bactericidal assay (SBA) using human complement. All adenovirus-based vaccine candidates induced high antigen-specific antibody and T cell responses. A single dose induced functional serum bactericidal responses with titers superior or equal to those induced by two doses of protein-based comparators, as well as longer persistence and a similar breadth. The fHbp transgene was further optimized for human use by incorporating a mutation abrogating binding to the human complement inhibitor factor H. The resulting vaccine candidate induced high and persistent SBA responses in transgenic mice expressing human factor H. The optimized transgene was inserted into the clinically relevant ChAdOx1 backbone, and this vaccine has now progressed to clinical development. The results of this preclinical vaccine development study underline the potential of vaccines based on genetic material to induce functional antibody responses against bacterial outer membrane proteins.
Collapse
Affiliation(s)
- Christina Dold
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, CCVTM, Churchill Lane, Oxford OX3 7LE, UK
| | - Leanne Marsay
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, CCVTM, Churchill Lane, Oxford OX3 7LE, UK
| | - Nelson Wang
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, CCVTM, Churchill Lane, Oxford OX3 7LE, UK
| | - Laura Silva-Reyes
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, CCVTM, Churchill Lane, Oxford OX3 7LE, UK
| | - Elizabeth Clutterbuck
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, CCVTM, Churchill Lane, Oxford OX3 7LE, UK
| | - Gavin K Paterson
- Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Kelsey Sharkey
- Division of Infectious Diseases and Global Health, Department of Pediatrics, University of California, San Francisco, San Francisco, CA 94143, USA
| | - David Wyllie
- Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Peter T Beernink
- Division of Infectious Diseases and Global Health, Department of Pediatrics, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Adrian V Hill
- Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, CCVTM, Churchill Lane, Oxford OX3 7LE, UK
| | - Christine S Rollier
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, CCVTM, Churchill Lane, Oxford OX3 7LE, UK
- School of Biosciences and Medicine, University of Surrey, Guildford GU2 7XH, UK
| |
Collapse
|
9
|
Zografaki I, Detsis M, Del Amo M, Iantomasi R, Maia A, Montuori EA, Mendez C. Invasive Meningococcal Disease epidemiology and vaccination strategies in four Southern European countries: a review of the available data. Expert Rev Vaccines 2023. [PMID: 37316234 DOI: 10.1080/14760584.2023.2225596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 06/12/2023] [Indexed: 06/16/2023]
Abstract
INTRODUCTION Invasive meningococcal disease (IMD) is a major health concern which can be prevented through vaccination. Conjugate vaccines against serogroups A, C, W and Y and two protein-based vaccines against serogroup B are currently available in the European Union. AREAS COVERED We present epidemiologic data for Italy, Portugal, Greece and Spain using publicly available reports from national reference laboratories and national or regional immunization programs (1999-2019), aiming to confirm risk groups, and describe time trends in overall incidence and serogroup distribution, as well as impact of immunization. Analysis of circulating MenB isolates in terms of the surface factor H binding protein (fHbp) using PubMLST is discussed as fHbp represents an important MenB vaccine antigen. Predictions of potential reactivity of the two available MenB vaccines (MenB-fHbp and 4CMenB) with circulating MenB isolates are also provided as assessed using the recently developed MenDeVAR tool. EXPERT OPINION Understanding dynamics of IMD and continued genomic surveillance are essential for evaluating vaccine effectiveness, but also prompting proactive immunization programs to prevent future outbreaks. Importantly, the successful design of further effective meningococcal vaccines to fight IMD relies on considering the unpredictable epidemiology of the disease and combining lessons learnt from capsule polysaccharide vaccines and protein-based vaccines.
Collapse
Affiliation(s)
| | | | | | | | - Ana Maia
- Vaccines Department, Pfizer Portugal, Lisbon, Portugal
| | | | | |
Collapse
|
10
|
Khan MA, Amin A, Farid A, Ullah A, Waris A, Shinwari K, Hussain Y, Alsharif KF, Alzahrani KJ, Khan H. Recent Advances in Genomics-Based Approaches for the Development of Intracellular Bacterial Pathogen Vaccines. Pharmaceutics 2022; 15:pharmaceutics15010152. [PMID: 36678781 PMCID: PMC9863128 DOI: 10.3390/pharmaceutics15010152] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/12/2022] [Accepted: 12/19/2022] [Indexed: 01/04/2023] Open
Abstract
Infectious diseases continue to be a leading cause of morbidity and mortality worldwide. The majority of infectious diseases are caused by intracellular pathogenic bacteria (IPB). Historically, conventional vaccination drives have helped control the pathogenesis of intracellular bacteria and the emergence of antimicrobial resistance, saving millions of lives. However, in light of various limitations, many diseases that involve IPB still do not have adequate vaccines. In response to increasing demand for novel vaccine development strategies, a new area of vaccine research emerged following the advent of genomics technology, which changed the paradigm of vaccine development by utilizing the complete genomic data of microorganisms against them. It became possible to identify genes related to disease virulence, genetic patterns linked to disease virulence, as well as the genetic components that supported immunity and favorable vaccine responses. Complete genomic databases, and advancements in transcriptomics, metabolomics, structural genomics, proteomics, immunomics, pan-genomics, synthetic genomics, and population biology have allowed researchers to identify potential vaccine candidates and predict their effects in patients. New vaccines have been created against diseases for which previously there were no vaccines available, and existing vaccines have been improved. This review highlights the key issues and explores the evolution of vaccines. The increasing volume of IPB genomic data, and their application in novel genome-based techniques for vaccine development, were also examined, along with their characteristics, and the opportunities and obstacles involved. Critically, the application of genomics technology has helped researchers rapidly select and evaluate candidate antigens. Novel vaccines capable of addressing the limitations associated with conventional vaccines have been developed and pressing healthcare issues are being addressed.
Collapse
Affiliation(s)
- Muhammad Ajmal Khan
- Division of Life Science, Center for Cancer Research, and State Key Lab of Molecular Neuroscience, Hong Kong University of Science and Technology, Hong Kong, China
- Correspondence: (M.A.K.); or (H.K.)
| | - Aftab Amin
- Division of Life Science, Center for Cancer Research, and State Key Lab of Molecular Neuroscience, Hong Kong University of Science and Technology, Hong Kong, China
| | - Awais Farid
- Division of Environment and Sustainability, Hong Kong University of Science and Technology, Hong Kong, China
| | - Amin Ullah
- Molecular Virology Laboratory, Department of Microbiology and Biotechnology, Abasyn University, Peshawar 25000, Pakistan
| | - Abdul Waris
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Khyber Shinwari
- Institute of Chemical Engineering, Department Immuno-Chemistry, Ural Federal University, Yekaterinbiurg 620002, Russia
| | - Yaseen Hussain
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan
| | - Khalaf F. Alsharif
- Department of Clinical Laboratory, College of Applied Medical Science, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Khalid J. Alzahrani
- Department of Clinical Laboratory, College of Applied Medical Science, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Haroon Khan
- Department of Clinical Laboratory, College of Applied Medical Science, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
- Correspondence: (M.A.K.); or (H.K.)
| |
Collapse
|
11
|
Stefanizzi P, Bianchi FP, Martinelli A, Di Lorenzo A, De Petro P, Graziano G, Lattanzio S, Diella G, Stella P, Ancona D, Tafuri S. Safety profile of MenB-FHBp vaccine among adolescents: data from surveillance of Adverse Events Following Immunization in Puglia (Italy), 2018-2020. Hum Vaccin Immunother 2022; 18:2041359. [PMID: 35201942 PMCID: PMC9009947 DOI: 10.1080/21645515.2022.2041359] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
MenB-FHBp was licensed in Europe in 2017 from the age of 10. In the “postmarketing life” of a new vaccine, surveillance of Adverse Events Following Immunization (AEFI) is crucial, to better understand the pattern of safety and the effectiveness. This paper describes the MenB-FHBp AEFIs notified in Puglia in 2018–2021, to take a picture of the safety profile of this vaccine in the real life, four years after its introduction in Italy. This is a retrospective observational study. Data were collected from the list of AEFIs notified after MenB-FHBp vaccine administration in Puglia in 2018–2020, and the number of doses of this vaccine administered in the same period. AEFIs were classified according to WHOʻs algorithm, and causality assessment was carried out for serious AEFIs. From January 2018 to December 2020, in Puglia, 43,061 doses of MenB-FHBp were administered and 42 MenB-FHBp AEFIs (reporting rate: 97.5 per 100,000 doses administered) were reported. Among these, 12 were classified as severe (28.6%; reporting rate 27.9 per 100,000 doses). Overall, the male/female ratio in AEFIs was 1:1. The median age of people who suffered from AEFIs was 12 years (range 11–13). For the 11 serious AEFIs for which the classification was “consistent causal association,” the diagnosis was hyperpyrexia (reporting rate 13.9 per 100,000 doses), fainting (rate 4.6 per 100,000 doses), urticaria (rate 2,3 per 100,000 doses), convulsions (rate 2,3 per 100,000 doses), and vomit (rate 2,3 per 100,000 doses). No deaths or impairment were notified in studied AEFIs. The picture of MenB-FHBp vaccine supports that the risk of AEFIs is in line with previous published data and in general acceptable.
Collapse
Affiliation(s)
- Pasquale Stefanizzi
- Department of Biomedical Science and Human Oncology, Aldo Moro University of Bari, Bari, Italy
| | - Francesco Paolo Bianchi
- Department of Biomedical Science and Human Oncology, Aldo Moro University of Bari, Bari, Italy
| | - Andrea Martinelli
- Department of Biomedical Science and Human Oncology, Aldo Moro University of Bari, Bari, Italy
| | - Antonio Di Lorenzo
- Department of Biomedical Science and Human Oncology, Aldo Moro University of Bari, Bari, Italy
| | - Paola De Petro
- Department of Biomedical Science and Human Oncology, Aldo Moro University of Bari, Bari, Italy
| | - Giusi Graziano
- Department of Biomedical Science and Human Oncology, Aldo Moro University of Bari, Bari, Italy
| | - Sabrina Lattanzio
- Department of Biomedical Science and Human Oncology, Aldo Moro University of Bari, Bari, Italy
| | - Giusy Diella
- Department of Biomedical Science and Human Oncology, Aldo Moro University of Bari, Bari, Italy
| | - Paolo Stella
- Regional Pharmacovigilance Center, Puglia Region, Italy
| | | | - Silvio Tafuri
- Department of Biomedical Science and Human Oncology, Aldo Moro University of Bari, Bari, Italy
| |
Collapse
|
12
|
Alfini R, Brunelli B, Bartolini E, Carducci M, Luzzi E, Ferlicca F, Buccato S, Galli B, Lo Surdo P, Scarselli M, Romagnoli G, Cartocci E, Maione D, Savino S, Necchi F, Delany I, Micoli F. Investigating the Role of Antigen Orientation on the Immune Response Elicited by Neisseria meningitidis Factor H Binding Protein on GMMA. Vaccines (Basel) 2022; 10:1182. [PMID: 35893831 PMCID: PMC9331691 DOI: 10.3390/vaccines10081182] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/15/2022] [Accepted: 07/16/2022] [Indexed: 11/16/2022] Open
Abstract
GMMA are outer membrane vesicles (OMVs) released from Gram-negative bacteria genetically modified to enhance OMVs formation that have been shown to be optimal systems to enhance immunogenicity of protein antigens. Here, we selected Neisseria meningitidis factor H binding protein (fHbp) and used the conjugation chemistry as a tool to alter antigen orientation on GMMA. Indeed, fHbp was randomly linked to GMMA or selectively attached via the N-terminus to mimic native presentation of the protein on the bacterial surface. Interestingly, protein and peptide array analyses confirmed that antibodies induced by the selective and the random conjugates showed a pattern very similar to fHbp natively expressed on bacterial surfaces or to the recombinant protein mixed with GMMA, respectively. However, the two conjugates elicited antibodies with similar serum bactericidal activity against meningococcal strains, superior to the protein alone or physically mixed with GMMA. Presentation of fHbp on GMMA strongly enhances the functional immune response elicited by the protein but its orientation on the bacterial surface does not have an impact. This study demonstrates the flexibility of the GMMA platform as a display and delivery system for enhancing antigen immunogenicity and further supports the use of such promising technology for the development of effective vaccines.
Collapse
Affiliation(s)
- Renzo Alfini
- GSK Vaccines Institute for Global Health (GVGH), 53100 Siena, Italy; (R.A.); (M.C.); (F.N.)
| | - Brunella Brunelli
- GSK, 53100 Siena, Italy; (B.B.); (E.B.); (E.L.); (F.F.); (S.B.); (B.G.); (P.L.S.); (M.S.); (G.R.); (E.C.); (D.M.); (S.S.); (I.D.)
| | - Erika Bartolini
- GSK, 53100 Siena, Italy; (B.B.); (E.B.); (E.L.); (F.F.); (S.B.); (B.G.); (P.L.S.); (M.S.); (G.R.); (E.C.); (D.M.); (S.S.); (I.D.)
| | - Martina Carducci
- GSK Vaccines Institute for Global Health (GVGH), 53100 Siena, Italy; (R.A.); (M.C.); (F.N.)
| | - Enrico Luzzi
- GSK, 53100 Siena, Italy; (B.B.); (E.B.); (E.L.); (F.F.); (S.B.); (B.G.); (P.L.S.); (M.S.); (G.R.); (E.C.); (D.M.); (S.S.); (I.D.)
| | - Francesca Ferlicca
- GSK, 53100 Siena, Italy; (B.B.); (E.B.); (E.L.); (F.F.); (S.B.); (B.G.); (P.L.S.); (M.S.); (G.R.); (E.C.); (D.M.); (S.S.); (I.D.)
| | - Scilla Buccato
- GSK, 53100 Siena, Italy; (B.B.); (E.B.); (E.L.); (F.F.); (S.B.); (B.G.); (P.L.S.); (M.S.); (G.R.); (E.C.); (D.M.); (S.S.); (I.D.)
| | - Barbara Galli
- GSK, 53100 Siena, Italy; (B.B.); (E.B.); (E.L.); (F.F.); (S.B.); (B.G.); (P.L.S.); (M.S.); (G.R.); (E.C.); (D.M.); (S.S.); (I.D.)
| | - Paola Lo Surdo
- GSK, 53100 Siena, Italy; (B.B.); (E.B.); (E.L.); (F.F.); (S.B.); (B.G.); (P.L.S.); (M.S.); (G.R.); (E.C.); (D.M.); (S.S.); (I.D.)
| | - Maria Scarselli
- GSK, 53100 Siena, Italy; (B.B.); (E.B.); (E.L.); (F.F.); (S.B.); (B.G.); (P.L.S.); (M.S.); (G.R.); (E.C.); (D.M.); (S.S.); (I.D.)
| | - Giacomo Romagnoli
- GSK, 53100 Siena, Italy; (B.B.); (E.B.); (E.L.); (F.F.); (S.B.); (B.G.); (P.L.S.); (M.S.); (G.R.); (E.C.); (D.M.); (S.S.); (I.D.)
| | - Elena Cartocci
- GSK, 53100 Siena, Italy; (B.B.); (E.B.); (E.L.); (F.F.); (S.B.); (B.G.); (P.L.S.); (M.S.); (G.R.); (E.C.); (D.M.); (S.S.); (I.D.)
| | - Domenico Maione
- GSK, 53100 Siena, Italy; (B.B.); (E.B.); (E.L.); (F.F.); (S.B.); (B.G.); (P.L.S.); (M.S.); (G.R.); (E.C.); (D.M.); (S.S.); (I.D.)
| | - Silvana Savino
- GSK, 53100 Siena, Italy; (B.B.); (E.B.); (E.L.); (F.F.); (S.B.); (B.G.); (P.L.S.); (M.S.); (G.R.); (E.C.); (D.M.); (S.S.); (I.D.)
| | - Francesca Necchi
- GSK Vaccines Institute for Global Health (GVGH), 53100 Siena, Italy; (R.A.); (M.C.); (F.N.)
| | - Isabel Delany
- GSK, 53100 Siena, Italy; (B.B.); (E.B.); (E.L.); (F.F.); (S.B.); (B.G.); (P.L.S.); (M.S.); (G.R.); (E.C.); (D.M.); (S.S.); (I.D.)
| | - Francesca Micoli
- GSK Vaccines Institute for Global Health (GVGH), 53100 Siena, Italy; (R.A.); (M.C.); (F.N.)
| |
Collapse
|
13
|
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
|
14
|
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
|
15
|
Mimicking Native Display of CD0873 on Liposomes Augments Its Potency as an Oral Vaccine against Clostridioides difficile. Vaccines (Basel) 2021; 9:vaccines9121453. [PMID: 34960199 PMCID: PMC8708880 DOI: 10.3390/vaccines9121453] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/02/2021] [Accepted: 12/03/2021] [Indexed: 11/23/2022] Open
Abstract
Mucosal vaccination aims to prevent infection mainly by inducing secretory IgA (sIgA) antibody, which neutralises pathogens and enterotoxins by blocking their attachment to epithelial cells. We previously demonstrated that encapsulated protein antigen CD0873 given orally to hamsters induces neutralising antibodies locally as well as systemically, affording partial protection against Clostridioides difficile infection. The aim of this study was to determine whether displaying CD0873 on liposomes, mimicking native presentation, would drive a stronger antibody response. The recombinant form we previously tested resembles the naturally cleaved lipoprotein commencing with a cysteine but lacking lipid modification. A synthetic lipid (DHPPA-Mal) was designed for conjugation of this protein via its N-terminal cysteine to the maleimide headgroup. DHPPA-Mal was first formulated with liposomes to produce MalLipo; then, CD0873 was conjugated to headgroups protruding from the outer envelope to generate CD0873-MalLipo. The immunogenicity of CD0873-MalLipo was compared to CD0873 in hamsters. Intestinal sIgA and CD0873-specific serum IgG were induced in all vaccinated animals; however, neutralising activity was greatest for the CD0873-MalLipo group. Our data hold great promise for development of a novel oral vaccine platform driving intestinal and systemic immune responses.
Collapse
|
16
|
Liu Y, Maciel M, O’Dowd A, Poole ST, Rollenhagen JE, Etobayeva IV, Savarino SJ. Development and Comparison of a Panel of Modified CS17 Fimbrial Tip Adhesin Proteins as Components for an Adhesin-Based Vaccine against Enterotoxigenic Escherichia coli. Microorganisms 2021; 9:microorganisms9081646. [PMID: 34442726 PMCID: PMC8401227 DOI: 10.3390/microorganisms9081646] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 07/25/2021] [Accepted: 07/27/2021] [Indexed: 11/23/2022] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) is a leading cause of diarrhea in travelers and children in resource-limited countries. ETEC colonization factors, fimbrial tip adhesins and enterotoxins are key virulence factors, and thus have been studied as vaccine candidates. Some prevalent colonization factors, including CFA/I and CS17, belong to the class 5 family. We previously found that passive oral administration of hyperimmune bovine colostral IgG (bIgG) raised against dscCfaE (donor strand complemented CFA/I tip adhesin) protected volunteers against CFA/I+ ETEC challenge, while anti-dscCsbD bIgG (CS17 tip adhesin) did not confer protection. These findings led us to develop and optimize a panel of alternative CsbD-based vaccine candidates based on allele matching and in silico protein engineering. Physicochemical characterizations revealed that an optimized vaccine candidate dscCsbDLSN139(P218A/G3) had the greatest thermal stability among the six tested dscCsbD adhesins, whereas the overall secondary structures and solubility of these adhesins had no obvious differences. Importantly, dscCsbDLSN139(P218A/G3) elicited significantly higher CS17+ ETEC hemagglutination inhibition titers in sera from mice intranasally immunized with the panel of dscCsbD adhesins, while no significant difference was observed among heterologous neutralizing titers. Our results strongly advocate for the incorporation of these modifications into a new generation of CsbD-based ETEC vaccine candidates.
Collapse
Affiliation(s)
- Yang Liu
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817, USA; (M.M.J.); (A.O.); (S.T.P.); (J.E.R.)
- Correspondence:
| | - Milton Maciel
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817, USA; (M.M.J.); (A.O.); (S.T.P.); (J.E.R.)
| | - Aisling O’Dowd
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817, USA; (M.M.J.); (A.O.); (S.T.P.); (J.E.R.)
| | - Steven T. Poole
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817, USA; (M.M.J.); (A.O.); (S.T.P.); (J.E.R.)
| | - Julianne E. Rollenhagen
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817, USA; (M.M.J.); (A.O.); (S.T.P.); (J.E.R.)
| | - Irina V. Etobayeva
- Enteric Diseases Department, Naval Medical Research Center, Silver Spring, MD 20910, USA; (I.V.E.); (S.J.S.)
| | - Stephen J. Savarino
- Enteric Diseases Department, Naval Medical Research Center, Silver Spring, MD 20910, USA; (I.V.E.); (S.J.S.)
| |
Collapse
|
17
|
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
|
18
|
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
|
19
|
Clark SA, Gray S, Finn A, Borrow R. Colistin Sensitivity and Factor H-Binding Protein Expression among Commensal Neisseria Species. mSphere 2021; 6:e0017521. [PMID: 34133203 PMCID: PMC8265630 DOI: 10.1128/msphere.00175-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 05/28/2021] [Indexed: 11/20/2022] Open
Abstract
Many bacterial carriage studies utilize colistin-containing media to select for Neisseria meningitidis among the diverse human pharyngeal milieu. These studies commonly report the isolation of Neisseria commensal species, with carriage rates of around 1% or less typically observed. Here, we describe the isolation of N. cinerea and N. polysaccharea from pharyngeal swabs using nonselective agar and confirm they are unable to grow on colistin-containing media. We also demonstrated colistin sensitivity among archived Neisseria commensal strains, including N. cinerea, N. polysaccharea, N. mucosa, and N. subflava. The distribution of lptA among these strains indicated that, while the phosphoethanolamine (PEA) transferase encoded by this gene confers colistin resistance, other mechanisms may lead to reduced susceptibility in some lptA-deficient strains. The majority of the N. cinerea and N. polysaccharea isolates expressed medium to very high levels of factor H-binding protein (fHbp), an important meningococcal vaccine antigen. Sequence analysis showed that the commensal fHbp peptide variants were similar in sequence to fHbp variants typically observed among invasive meningococci. Altogether, these results not only suggest that Neisseria commensal strains could be carried at much higher rates than previously reported but also raise questions about the impact of protein-based meningococcal vaccines on these unencapsulated commensals. IMPORTANCE This study highlights the need for further work to accurately determine the pharyngeal carriage prevalence of Neisseria commensal bacteria (e.g., N. cinerea and N. polysaccharea) among the general population. Previous studies have clearly demonstrated the suppressive effect these commensal species can have on meningococcal colonization, and so the carriage prevalence of these species could be an important factor in the spread of meningococci through the population. Furthermore, the surface expression of the meningococcal vaccine antigen factor H-binding protein by many of these commensal strains could have important implications for the use of fHbp-containing vaccines. Carriage of these commensal species may influence the immune response to these vaccines, or conversely, the immune response elicited by vaccination may induce clearance of these potentially important members of the pharyngeal niche.
Collapse
Affiliation(s)
- Stephen A. Clark
- Meningococcal Reference Unit (MRU), Public Health England (PHE), Manchester, United Kingdom
| | - Steve Gray
- Meningococcal Reference Unit (MRU), Public Health England (PHE), Manchester, United Kingdom
| | - Adam Finn
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | - Ray Borrow
- Meningococcal Reference Unit (MRU), Public Health England (PHE), Manchester, United Kingdom
| |
Collapse
|
20
|
Sands NA, Beernink PT. Two human antibodies to a meningococcal serogroup B vaccine antigen enhance binding of complement Factor H by stabilizing the Factor H binding site. PLoS Pathog 2021; 17:e1009655. [PMID: 34125873 PMCID: PMC8224966 DOI: 10.1371/journal.ppat.1009655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 06/24/2021] [Accepted: 05/20/2021] [Indexed: 11/19/2022] Open
Abstract
Microbial pathogens bind host complement regulatory proteins to evade the immune system. The bacterial pathogen Neisseria meningitidis, or meningococcus, binds several complement regulators, including human Factor H (FH). FH binding protein (FHbp) is a component of two licensed meningococcal vaccines and in mice FHbp elicits antibodies that inhibit binding of FH to FHbp, which defeat the bacterial evasion mechanism. However, humans vaccinated with FHbp develop antibodies that enhance binding of FH to the bacteria, which could limit the effectiveness of the vaccines. In the present study, we show that two vaccine-elicited antibody fragments (Fabs) isolated from different human subjects increase binding of complement FH to meningococcal FHbp by ELISA. The two Fabs have different effects on the kinetics of FH binding to immobilized FHbp as measured by surface plasmon resonance. The 1.7- and 2.0-Å resolution X-ray crystal structures of the Fabs in complexes with FHbp illustrate that the two Fabs bind to similar epitopes on the amino-terminal domain of FHbp, adjacent to the FH binding site. Superposition models of ternary complexes of each Fab with FHbp and FH show that there is likely minimal contact between the Fabs and FH. Collectively, the structures reveal that the Fabs enhance binding of FH to FHbp by altering the conformations and mobilities of two loops adjacent to the FH binding site of FHbp. In addition, the 1.5 Å-resolution structure of one of the isolated Fabs defines the structural rearrangements associated with binding to FHbp. The FH-enhancing human Fabs, which are mirrored in the human polyclonal antibody responses, have important implications for tuning the effectiveness of FHbp-based vaccines.
Collapse
Affiliation(s)
- Nathaniel A. Sands
- Division of Infectious Diseases and Global Health, Department of Pediatrics, School of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Peter T. Beernink
- Division of Infectious Diseases and Global Health, Department of Pediatrics, School of Medicine, University of California San Francisco, San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
21
|
Vaccination with meningococcal outer membrane vesicles carrying Borrelia OspA protects against experimental Lyme borreliosis. Vaccine 2021; 39:2561-2567. [PMID: 33812741 DOI: 10.1016/j.vaccine.2021.03.059] [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] [Received: 12/14/2020] [Revised: 03/05/2021] [Accepted: 03/18/2021] [Indexed: 11/21/2022]
Abstract
Currently there is no human vaccine against Lyme borreliosis, and most research focuses on recombinant protein vaccines, as such a vaccine has been proven to be successful in the past. The expression of recombinant antigens in meningococcal Outer Membrane Vesicles (OMVs), with the OMV functioning both as adjuvant and delivery vehicle, greatly enhances their potential. Immunization studies in mice have shown that OMV-based vaccines can protect against various pathogens and an OMV-based meningococcal vaccine is approved and available for human use. Because of its surface localization in Borrelia and the detailed knowledge regarding its immunogenicity and structure, OspA was chosen as a suitable lipoprotein to be tested as an OMV-based vaccine against Lyme borreliosis. We have previously shown that the OMV-OspA vaccine was immunogenic in mice and here we assessed the efficacy of OMV-OspA. We generated a second-generation OMV-OspA vaccine and vaccinated C3H/HeN mice with (EDTA extracted) meningococcal OMVs expressing OspA from B. burgdorferi strain B31. The adjuvant effect of empty OMVs on recombinant OspA was tested as well. We subsequently challenged mice with a subcutaneous injection of B. burgdorferi. Average antibody end-point titers against the OspA-OMV construct were high, although lower compared to the antibodies raised against recombinant OspA. Interestingly, antibody titers between recombinant OspA adjuvanted with aluminum hydroxide and recombinant OspA with OMV as adjuvant were comparable. Finally, qPCR and culture data show that both the OspA-OMV and the vaccine based on recombinant OspA with OMV as adjuvant provided significant, yet partial protection, against Borrelia infection. OMV-based vaccines using Borrelia (lipo)proteins are an easy and feasible vaccination method protecting against B. burgdorferi infection and could be a promising strategy in humans.
Collapse
|
22
|
Eichner H, Karlsson J, Spelmink L, Pathak A, Sham LT, Henriques-Normark B, Loh E. RNA thermosensors facilitate Streptococcus pneumoniae and Haemophilus influenzae immune evasion. PLoS Pathog 2021; 17:e1009513. [PMID: 33914847 PMCID: PMC8084184 DOI: 10.1371/journal.ppat.1009513] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 03/29/2021] [Indexed: 11/18/2022] Open
Abstract
Bacterial meningitis is a major cause of death and disability in children worldwide. Two human restricted respiratory pathogens, Streptococcus pneumoniae and Haemophilus influenzae, are the major causative agents of bacterial meningitis, attributing to 200,000 deaths annually. These pathogens are often part of the nasopharyngeal microflora of healthy carriers. However, what factors elicit them to disseminate and cause invasive diseases, remain unknown. Elevated temperature and fever are hallmarks of inflammation triggered by infections and can act as warning signals to pathogens. Here, we investigate whether these respiratory pathogens can sense environmental temperature to evade host complement-mediated killing. We show that productions of two vital virulence factors and vaccine components, the polysaccharide capsules and factor H binding proteins, are temperature dependent, thus influencing serum/opsonophagocytic killing of the bacteria. We identify and characterise four novel RNA thermosensors in S. pneumoniae and H. influenzae, responsible for capsular biosynthesis and production of factor H binding proteins. Our data suggest that these bacteria might have independently co-evolved thermosensing abilities with different RNA sequences but distinct secondary structures to evade the immune system.
Collapse
Affiliation(s)
- Hannes Eichner
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solna, Sweden
| | - Jens Karlsson
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solna, Sweden
| | - Laura Spelmink
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solna, Sweden
| | - Anuj Pathak
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solna, Sweden
| | - Lok-To Sham
- Infectious Disease Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Birgitta Henriques-Normark
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solna, Sweden
- Clinical Microbiology, Bioclinicum, Karolinska University Hospital, Solna, Sweden
- Lee Kong Chian School of Medicine and Singapore Centre on Environmental Life Sciences Engineering, Nanyang Technological University, Singapore, Singapore
| | - Edmund Loh
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solna, Sweden
- Clinical Microbiology, Bioclinicum, Karolinska University Hospital, Solna, Sweden
- Lee Kong Chian School of Medicine and Singapore Centre on Environmental Life Sciences Engineering, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
23
|
Spinsanti M, Brignoli T, Bodini M, Fontana LE, De Chiara M, Biolchi A, Muzzi A, Scarlato V, Delany I. Deconvolution of intergenic polymorphisms determining high expression of Factor H binding protein in meningococcus and their association with invasive disease. PLoS Pathog 2021; 17:e1009461. [PMID: 33770146 PMCID: PMC8026042 DOI: 10.1371/journal.ppat.1009461] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 04/07/2021] [Accepted: 03/09/2021] [Indexed: 12/11/2022] Open
Abstract
Neisseria meningitidis is a strictly human pathogen and is the major cause of septicemia and meningitis worldwide. Factor H binding protein (fHbp) is a meningococcal surface-exposed lipoprotein that binds the human Complement factor H allowing the bacterium to evade the host innate immune response. FHbp is also a key antigen in two vaccines against N. meningitidis serogroup B. Although the fHbp gene is present in most circulating meningococcal strains, level of fHbp expression varies among isolates and has been correlated to differences in promoter sequences upstream of the gene. Here we elucidated the sequence determinants that control fHbp expression in globally circulating strains. We analyzed the upstream fHbpintergenic region (fIR) of more than 5800 strains representative of the UK circulating isolates and we identified eleven fIR sequence alleles which represent 88% of meningococcal strains. By engineering isogenic recombinant strains where fHbp expression was under the control of each of the eleven fIR alleles, we confirmed that the fIR sequence determines a specific and distinct level of expression. Moreover, we identified the molecular basis for variation in expression through polymorphisms within key regulatory regions that are known to affect fHbp expression. We experimentally established three expression groups, high–medium–low, that correlated directly with the susceptibility to killing mediated by anti-fHbp antibodies and the ability of the meningococcal strain to survive within human serum. By using this sequence classification and information about the variant, we predicted fHbp expression in the panel of UK strains and we observed that strains with higher expressing fIR alleles are more likely associated with invasive disease. Overall, our findings can contribute to understand and predict vaccine coverage mediated by fHbp as well as to shed light on the role of this virulence factor in determining an invasive phenotype. Complement plays a key role in the immunity against Neisseria meningitidis. The meningococcus uses the Factor H binding protein (fHbp), to bind a negative regulator of the alternative complement pathway, factor H, to its surface thus preventing complement deposition and lysis. The use of fHbp as an antigen in two licensed vaccines highlights its public health relevance. Therefore the levels of this antigen produced by the bacterium are pivotal on the one hand for the survival of N. meningitidis in blood and on the other hand for the susceptibility to vaccine-induced killing antibodies. Here, we identify the predominant nucleotide sequences that drive distinct levels of the fHbp antigen in circulating meningococcal strains. We cluster them into distinct groups with increasing levels and observe that strains expressing higher fHbp amounts are associated with invasive disease. Our findings show that the nucleotide sequence of the fHbp promoter can be used for the prediction of antigen levels of any given strain and consequently for both the assessment of its sensitivity to killing by fHbp antibodies and its likelihood to cause invasive disease.
Collapse
Affiliation(s)
| | - Tarcisio Brignoli
- GSK, Siena, Italy
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Bologna, Italy
| | | | | | | | | | | | - Vincenzo Scarlato
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Bologna, Italy
| | | |
Collapse
|
24
|
Moore SR, Menon SS, Cortes C, Ferreira VP. Hijacking Factor H for Complement Immune Evasion. Front Immunol 2021; 12:602277. [PMID: 33717083 PMCID: PMC7947212 DOI: 10.3389/fimmu.2021.602277] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 01/15/2021] [Indexed: 12/15/2022] Open
Abstract
The complement system is an essential player in innate and adaptive immunity. It consists of three pathways (alternative, classical, and lectin) that initiate either spontaneously (alternative) or in response to danger (all pathways). Complement leads to numerous outcomes detrimental to invaders, including direct killing by formation of the pore-forming membrane attack complex, recruitment of immune cells to sites of invasion, facilitation of phagocytosis, and enhancement of cellular immune responses. Pathogens must overcome the complement system to survive in the host. A common strategy used by pathogens to evade complement is hijacking host complement regulators. Complement regulators prevent attack of host cells and include a collection of membrane-bound and fluid phase proteins. Factor H (FH), a fluid phase complement regulatory protein, controls the alternative pathway (AP) both in the fluid phase of the human body and on cell surfaces. In order to prevent complement activation and amplification on host cells and tissues, FH recognizes host cell-specific polyanionic markers in combination with complement C3 fragments. FH suppresses AP complement-mediated attack by accelerating decay of convertases and by helping to inactivate C3 fragments on host cells. Pathogens, most of which do not have polyanionic markers, are not recognized by FH. Numerous pathogens, including certain bacteria, viruses, protozoa, helminths, and fungi, can recruit FH to protect themselves against host-mediated complement attack, using either specific receptors and/or molecular mimicry to appear more like a host cell. This review will explore pathogen complement evasion mechanisms involving FH recruitment with an emphasis on: (a) characterizing the structural properties and expression patterns of pathogen FH binding proteins, as well as other strategies used by pathogens to capture FH; (b) classifying domains of FH important in pathogen interaction; and (c) discussing existing and potential treatment strategies that target FH interactions with pathogens. Overall, many pathogens use FH to avoid complement attack and appreciating the commonalities across these diverse microorganisms deepens the understanding of complement in microbiology.
Collapse
Affiliation(s)
- Sara R Moore
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
| | - Smrithi S Menon
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
| | - Claudio Cortes
- Department of Foundational Medical Sciences, Oakland University William Beaumont School of Medicine, Rochester, MI, United States
| | - Viviana P Ferreira
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
| |
Collapse
|
25
|
Lodi L, Moriondo M, Nieddu F, Ricci S, Guiducci S, Lippi F, Canessa C, Calistri E, Citera F, Giovannini M, Indolfi G, Resti M, Azzari C. Molecular typing of group B Neisseria meningitidis'subcapsular antigens directly on biological samples demonstrates epidemiological congruence between culture-positive and -negative cases: A surveillance study of invasive disease over a 13-year period. J Infect 2021; 82:28-36. [PMID: 33610687 DOI: 10.1016/j.jinf.2020.12.034] [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] [Received: 08/01/2020] [Revised: 12/07/2020] [Accepted: 12/22/2020] [Indexed: 10/22/2022]
Abstract
BACKGROUND Surveillance of serogroup B Neisseria meningitidis (MenB) subcapsular antigen variant distribution in invasive disease (IMD) is fundamental for multicomponent vaccine coverage prediction. IMD incidence in Tuscany in 2018 was 0.37/100,000 inhabitants, with MenB representing 57% of cases. More than 50% of MenB responsible for IMD cannot be grown in culture, and molecular characterization of these cases is often lacking. The aim of the present study was to describe the distribution of MenB subcapsular antigens, comparing their distribution in culture-positive and culture-negative cases. METHODS Molecular data regarding clonal complexes and subcapsular antigen variants of the 55 MenB-IMD occurring in Tuscany from 2007 to 2019 were made available, and their distribution between culture-positive and culture-negative cases was compared. Genetic-MATS and MenDeVAR prediction systems were used to assess multicomponent vaccine coverage predictions. RESULTS Culture-positive and culture-negative cases presented a similar percentage representation of fHbp subfamilies. Clonal complex 162 was almost constantly associated with fHbp B231/v1.390, Neisserial-heparin-binding-antigen (NHBA) peptide 20, and PorinA P1.22,14 (BAST-3033): these were the most represented antigenic variants, both in culture-positive and culture-negative groups. Point-estimate 4CMenB coverage prediction was 88.5% (84.6%-92.3%). CONCLUSIONS Our data demonstrate that non-cultivable meningococci, responsible for IMD, possess genetic variants of subcapsular antigens that are representative of what has been observed in culture. The vaccine-related antigenic epidemiology of MenB is thus similar in both groups. One of the first on-field applications of gMATS and MenDeVAR identifies their major advantage in their accessibility and in the possibility of dynamic data implementation that must be pursued continuously in the future.
Collapse
Affiliation(s)
- Lorenzo Lodi
- Department of Health Sciences, University of Florence and Meyer Children's University Hospital, viale Pieraccini 24, 50139 Florence, Italy
| | - Maria Moriondo
- Department of Health Sciences, University of Florence and Meyer Children's University Hospital, viale Pieraccini 24, 50139 Florence, Italy
| | - Francesco Nieddu
- Department of Health Sciences, University of Florence and Meyer Children's University Hospital, viale Pieraccini 24, 50139 Florence, Italy
| | - Silvia Ricci
- Department of Health Sciences, University of Florence and Meyer Children's University Hospital, viale Pieraccini 24, 50139 Florence, Italy.
| | - Sara Guiducci
- Department of Health Sciences, University of Florence and Meyer Children's University Hospital, viale Pieraccini 24, 50139 Florence, Italy
| | - Francesca Lippi
- Department of Health Sciences, University of Florence and Meyer Children's University Hospital, viale Pieraccini 24, 50139 Florence, Italy
| | - Clementina Canessa
- Department of Health Sciences, University of Florence and Meyer Children's University Hospital, viale Pieraccini 24, 50139 Florence, Italy
| | - Elisa Calistri
- Department of Health Sciences, University of Florence and Meyer Children's University Hospital, viale Pieraccini 24, 50139 Florence, Italy
| | - Francesco Citera
- Department of Health Sciences, University of Florence and Meyer Children's University Hospital, viale Pieraccini 24, 50139 Florence, Italy
| | - Mattia Giovannini
- Department of Health Sciences, University of Florence and Meyer Children's University Hospital, viale Pieraccini 24, 50139 Florence, Italy
| | - Giuseppe Indolfi
- Department of Pediatrics, Meyer Children's University Hospital, viale Pieraccini 24, 50139 Florence, Italy
| | - Massimo Resti
- Department of Pediatrics, Meyer Children's University Hospital, viale Pieraccini 24, 50139 Florence, Italy
| | - Chiara Azzari
- Department of Health Sciences, University of Florence and Meyer Children's University Hospital, viale Pieraccini 24, 50139 Florence, Italy
| |
Collapse
|
26
|
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
|
27
|
Comprehensive Bioinformatic Assessments of the Variability of Neisseria gonorrhoeae Vaccine Candidates. mSphere 2021; 6:6/1/e00977-20. [PMID: 33536323 PMCID: PMC7860988 DOI: 10.1128/msphere.00977-20] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
A protective vaccine is the only viable way to stop the spread of gonorrhea in the face of rising antibiotic resistance. However, the notorious phase and antigenic variation of Neisseria gonorrhoeae surface proteins remains one of the challenges in vaccine development. To facilitate vaccine advancement efforts, we carried out comprehensive bioinformatic analyses of sequence variation by comparing 34 gonorrhea antigen candidates among >5,000 clinical N. gonorrhoeae isolates deposited in the Neisseria PubMLST database. Eight protein antigens showed exceptional conservation by having a single allele variant distributed in >80% of isolates. An additional 18 vaccine candidates were represented by ≤3 alleles in >50% of N. gonorrhoeae isolates globally. Phylogenetic analyses highlighted closely related antigen variants and additionally showed that AniA and FetB were the closest between N. gonorrhoeae and N. meningitidis Up to 44% of N. meningitidis alleles for both antigens have premature stop codons, suggesting differential expression. Mapping polymorphisms to the available three-dimensional structures of 12 antigens revealed low-frequency surface polymorphisms. PorB and TbpB possessed numerous high-prevalence polymorphic sites. While TbpA was also highly variable, conserved loops were nonetheless identified. A high degree of sequence conservation, the distribution of a single antigen variant among N. gonorrhoeae strains globally, or low-frequency sequence polymorphisms in surface loops make ACP, AniA, BamA, BamE, MtrE, NspA, NGO0778, NGO1251, NGO1985, OpcA, PldA, Slam2, and ZnuD promising candidates for a gonorrhea vaccine. Finally, the commonly used N. gonorrhoeae FA1090 strain emerges as a vaccine prototype, as it carries antigen sequence types identical to the most broadly distributed antigen variants.IMPORTANCE Neisseria gonorrhoeae, the Gram-negative bacterium responsible for the sexually transmitted infection gonorrhea, is categorized as a high-priority pathogen for research and development efforts. N. gonorrhoeae's "superbug" status, its high morbidity, and the serious health impact associated with gonorrhea highlight the importance of vaccine development. One of the longstanding barriers to developing an effective vaccine against N. gonorrhoeae is the remarkable variability of surface-exposed antigens. In this report, we addressed this roadblock by applying extensive bioinformatic analyses to 34 gonorrhea antigen candidates among >5,000 clinical N. gonorrhoeae isolates. Our studies are important, as they reveal promising, conserved gonorrhea vaccine candidates and aid structural vaccinology. Moreover, these approaches are broadly applicable to other infectious diseases where surface antigen variability impedes successful vaccine design.
Collapse
|
28
|
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
|
29
|
Meningococcal Deduced Vaccine Antigen Reactivity (MenDeVAR) Index: a Rapid and Accessible Tool That Exploits Genomic Data in Public Health and Clinical Microbiology Applications. J Clin Microbiol 2020; 59:JCM.02161-20. [PMID: 33055180 PMCID: PMC7771438 DOI: 10.1128/jcm.02161-20] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 10/09/2020] [Indexed: 12/24/2022] Open
Abstract
As microbial genomics makes increasingly important contributions to clinical and public health microbiology, the interpretation of whole-genome sequence data by nonspecialists becomes essential. In the absence of capsule-based vaccines, two protein-based vaccines have been used for the prevention of invasive serogroup B meningococcal disease (IMD) since their licensure in 2013 and 2014. These vaccines have different components and different levels of coverage of meningococcal variants. Hence, decisions regarding which vaccine to use in managing serogroup B IMD outbreaks require information about the index case isolate, including (i) the presence of particular vaccine antigen variants, (ii) the expression of vaccine antigens, and (iii) the likely susceptibility of its antigen variants to antibody-dependent bactericidal killing. As microbial genomics makes increasingly important contributions to clinical and public health microbiology, the interpretation of whole-genome sequence data by nonspecialists becomes essential. In the absence of capsule-based vaccines, two protein-based vaccines have been used for the prevention of invasive serogroup B meningococcal disease (IMD) since their licensure in 2013 and 2014. These vaccines have different components and different levels of coverage of meningococcal variants. Hence, decisions regarding which vaccine to use in managing serogroup B IMD outbreaks require information about the index case isolate, including (i) the presence of particular vaccine antigen variants, (ii) the expression of vaccine antigens, and (iii) the likely susceptibility of its antigen variants to antibody-dependent bactericidal killing. To obtain this information requires a multitude of laboratory assays, impractical in real-time clinical settings, where the information is most urgently needed. To facilitate assessment for public health and clinical purposes, we synthesized genomic and experimental data from published sources to develop and implement the Meningococcal Deduced Vaccine Antigen Reactivity (MenDeVAR) Index, which is publicly available on PubMLST (https://pubmlst.org). Using whole-genome sequences or individual gene sequences obtained from IMD isolates or clinical specimens, the MenDeVAR Index provides rapid evidence-based information on the presence and possible immunological cross-reactivity of different meningococcal vaccine antigen variants. The MenDeVAR Index enables practitioners who are not genomics specialists to assess the likely reactivity of vaccines for individual cases, outbreak management, or the assessment of public health vaccine programs. The MenDeVAR Index has been developed in consultation with, but independently of, both the 4CMenB (Bexsero; GSK) and rLP2086 (Trumenba; Pfizer, Inc.) vaccine manufacturers.
Collapse
|
30
|
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
|
31
|
Neisseria meningitidis Urethritis Outbreak Isolates Express a Novel Factor H Binding Protein Variant That Is a Potential Target of Group B-Directed Meningococcal (MenB) Vaccines. Infect Immun 2020; 88:IAI.00462-20. [PMID: 32958529 DOI: 10.1128/iai.00462-20] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 09/17/2020] [Indexed: 12/12/2022] Open
Abstract
Factor H binding protein (FHbp) is an important Neisseria meningitidis virulence factor that binds a negative regulator of the alternative complement pathway, human factor H (FH). Binding of FH increases meningococcal resistance to complement-mediated killing. FHbp also is reported to prevent interaction of the antimicrobial peptide (AMP) LL-37 with the meningococcal surface and meningococcal killing. FHbp is a target of two licensed group B-directed meningococcal (MenB) vaccines. We found a new FHbp variant, peptide allele identification no. 896 (ID 896), was highly expressed by an emerging meningococcal pathotype, the nonencapsulated urethritis clade (US_NmUC). This clade has been responsible for outbreaks of urethritis in multiple U.S. cities since 2015, other mucosal infections, and cases of invasive meningococcal disease. FHbp ID 896 is a member of the variant group 1 (subfamily B), bound protective anti-FHbp monoclonal antibodies, bound high levels of human FH, and enhanced the resistance of the clade to complement-mediated killing in low levels of human complement likely present at human mucosal surfaces. Interestingly, expression of FHbp ID 896 resulted in augmented killing of the clade by LL-37. FHbp ID 896 of the clade was recognized by antibodies elicited by FHbp in MenB vaccines.
Collapse
|
32
|
Carannante A, Fazio C, Neri A, Lista F, Fillo S, Ciammaruconi A, Vacca P, Stefanelli P. Meningococcal B vaccine antigen FHbp variants among disease-causing Neisseria meningitidis B isolates, Italy, 2014-2017. PLoS One 2020; 15:e0241793. [PMID: 33176334 PMCID: PMC7657669 DOI: 10.1371/journal.pone.0241793] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 10/20/2020] [Indexed: 11/19/2022] Open
Abstract
Background Typing of Neisseria meningitidis isolates is crucial for the surveillance of invasive meningococcal disease (IMD). We performed a molecular epidemiology study of N. meningitidis serogroup B (MenB) causing IMD in Italy between 2014 and 2017 to describe circulating strains belonging to this serogroup, with particular regards to the two factor H-binding protein (FHbp) subfamilies present in the bivalent MenB vaccine. Materials and methods A total of 109 culture positive and 46 culture negative MenB samples were collected within the National Surveillance System (NSS) of IMD in Italy and molecularly analyzed by conventional methods. Results Overall, 71 MenB samples showed the FHbp subfamily A and 83 the subfamily B. The subfamily variants were differently distributed by age. The most frequent variants, A05 and B231, were associated with cc213 and cc162, respectively. All MenB with the FHbp A05 variant displayed the PorA P1.22,14 and 85.7% of them the FetA F5-5. The majority of MenB with the FHbp B231 variant showed the PorA P1.22,14 (65.4%) and 84.6%, the FetA F3-6. Conclusion MenB circulating in Italy were characterized by a remarkable association between clonal complex and FHbp variants, although a high degree of genetic diversity observed over time. A dynamic trend in clonal complexes distribution within MenB was detected. Our results stress the importance of continued meningococcal molecular surveillance to evaluate the potential vaccine coverage of the available MenB vaccines.
Collapse
Affiliation(s)
- Anna Carannante
- Department Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Cecilia Fazio
- Department Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Arianna Neri
- Department Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | | | - Silvia Fillo
- Scientific Department, Army Medical Center, Rome, Italy
| | | | - Paola Vacca
- Department Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Paola Stefanelli
- Department Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
- * E-mail:
| |
Collapse
|
33
|
Immunological fingerprint of 4CMenB recombinant antigens via protein microarray reveals key immunosignatures correlating with bactericidal activity. Nat Commun 2020; 11:4994. [PMID: 33020485 PMCID: PMC7536418 DOI: 10.1038/s41467-020-18791-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 09/10/2020] [Indexed: 11/08/2022] Open
Abstract
Serogroup B meningococcus (MenB) is a leading cause of meningitis and sepsis across the world and vaccination is the most effective way to protect against this disease. 4CMenB is a multi-component vaccine against MenB, which is now licensed for use in subjects >2 months of age in several countries. In this study, we describe the development and use of an ad hoc protein microarray to study the immune response induced by the three major 4CMenB antigenic components (fHbp, NHBA and NadA) in individual sera from vaccinated infants, adolescents and adults. The resulting 4CMenB protein antigen fingerprinting allowed the identification of specific human antibody repertoire correlating with the bactericidal response elicited in each subject. This work represents an example of epitope mapping of the immune response induced by a multicomponent vaccine in different age groups with the identification of protective signatures. It shows the high flexibility of this microarray based methodology in terms of high-throughput information and minimal volume of biological samples needed. 4CMenB is an approved multi-component vaccine against Serogroup B meningococcus. Here the authors develop a protein microarray for three major 4CMenB antigenic components (fHbp, NHBA and NadA) and describe antibody repertoires in sera from vaccinated infants, adolescents and adults correlating with bactericidal response.
Collapse
|
34
|
Takahashi H, Dohmae N, Kim KS, Shimuta K, Ohnishi M, Yokoyama S, Yanagisawa T. Genetic incorporation of non-canonical amino acid photocrosslinkers in Neisseria meningitidis: New method provides insights into the physiological function of the function-unknown NMB1345 protein. PLoS One 2020; 15:e0237883. [PMID: 32866169 PMCID: PMC7458321 DOI: 10.1371/journal.pone.0237883] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 08/04/2020] [Indexed: 02/01/2023] Open
Abstract
Although whole-genome sequencing has provided novel insights into Neisseria meningitidis, many open reading frames have only been annotated as hypothetical proteins with unknown biological functions. Our previous genetic analyses revealed that the hypothetical protein, NMB1345, plays a crucial role in meningococcal infection in human brain microvascular endothelial cells; however, NMB1345 has no homology to any identified protein in databases and its physiological function could not be elucidated using pre-existing methods. Among the many biological technologies to examine transient protein-protein interaction in vivo, one of the developed methods is genetic code expansion with non-canonical amino acids (ncAAs) utilizing a pyrrolysyl-tRNA synthetase/tRNAPyl pair from Methanosarcina species: However, this method has never been applied to assign function-unknown proteins in pathogenic bacteria. In the present study, we developed a new method to genetically incorporate ncAAs-encoded photocrosslinking probes into N. meningitidis by utilizing a pyrrolysyl-tRNA synthetase/tRNAPyl pair and elucidated the biological function(s) of the NMB1345 protein. The results revealed that the NMB1345 protein directly interacts with PilE, a major component of meningococcal pili, and further physicochemical and genetic analyses showed that the interaction between the NMB1345 protein and PilE was important for both functional pilus formation and meningococcal infectious ability in N. meningitidis. The present study using this new methodology for N. meningitidis provides novel insights into meningococcal pathogenesis by assigning the function of a hypothetical protein.
Collapse
Affiliation(s)
- Hideyuki Takahashi
- National Institute of Infectious Diseases, Department of Bacteriology I, Shinjuku-ku, Japan
- * E-mail:
| | - Naoshi Dohmae
- Biomolecular Characterization Unit, RIKEN Center for Sustainable Resource Science, Wako, Japan
| | - Kwang Sik Kim
- Division of Pediatric Infectious Diseases, Department of Pediatrics, School of Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Ken Shimuta
- National Institute of Infectious Diseases, Department of Bacteriology I, Shinjuku-ku, Japan
| | - Makoto Ohnishi
- National Institute of Infectious Diseases, Department of Bacteriology I, Shinjuku-ku, Japan
| | - Shigeyuki Yokoyama
- RIKEN Structural Biology Laboratory, Yokohama, Japan
- RIKEN Cluster for Science, Technology and Innovation Hub, Yokohama, Japan
| | - Tatsuo Yanagisawa
- RIKEN Structural Biology Laboratory, Yokohama, Japan
- RIKEN Cluster for Science, Technology and Innovation Hub, Yokohama, Japan
| |
Collapse
|
35
|
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
|
36
|
Principato S, Pizza M, Rappuoli R. Meningococcal factor H binding protein as immune evasion factor and vaccine antigen. FEBS Lett 2020; 594:2657-2669. [PMID: 32298465 DOI: 10.1002/1873-3468.13793] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 03/25/2020] [Accepted: 03/30/2020] [Indexed: 01/15/2023]
Abstract
Factor H binding protein (fHbp) is a key virulence factor of Neisseria meningitidis and a main component of the two licensed vaccines against serogroup B meningococcus (Bexsero and Trumenba). fHbp is a surface-exposed lipoprotein that enables the bacterium to survive in human blood by binding the human complement regulator factor H (fH). When used as vaccine, the protein induces antibodies with potent bactericidal activity. While the fHbp gene is present in the majority of N. meningitidis serogroup B isolates, the expression level varies up to 15 times between different strains and more than 700 different sequence variants have been described. Antigenically, the protein has been divided into three variants or two subfamilies. The 3D structure of fHbp alone, in combination with fH or in complex with bactericidal antibodies, has been key to understanding the molecular details of the protein. In this article, we will review the biochemical and immunological properties of fHbp, and its key role in meningococcal pathogenesis, complement regulation, and immune evasion.
Collapse
|
37
|
Aston-Deaville S, Carlsson E, Saleem M, Thistlethwaite A, Chan H, Maharjan S, Facchetti A, Feavers IM, Alistair Siebert C, Collins RF, Roseman A, Derrick JP. An assessment of the use of Hepatitis B Virus core protein virus-like particles to display heterologous antigens from Neisseria meningitidis. Vaccine 2020; 38:3201-3209. [PMID: 32178907 PMCID: PMC7113836 DOI: 10.1016/j.vaccine.2020.03.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 02/25/2020] [Accepted: 03/01/2020] [Indexed: 12/15/2022]
Abstract
Neisseria meningitidis is the causative agent of meningococcal meningitis and sepsis and remains a significant public health problem in many countries. Efforts to develop a comprehensive vaccine against serogroup B meningococci have focused on the use of surface-exposed outer membrane proteins. Here we report the use of virus-like particles derived from the core protein of Hepatitis B Virus, HBc, to incorporate antigen domains derived from Factor H binding protein (FHbp) and the adhesin NadA. The extracellular domain of NadA was inserted into the major immunodominant region of HBc, and the C-terminal domain of FHbp at the C-terminus (CFHbp), creating a single polypeptide chain 3.7-fold larger than native HBc. Remarkably, cryoelectron microscopy revealed that the construct formed assemblies that were able to incorporate both antigens with minimal structural changes to native HBc. Electron density was weak for NadA and absent for CFHbp, partly attributable to domain flexibility. Following immunization of mice, three HBc fusions (CFHbp or NadA alone, NadA + CFHbp) were able to induce production of IgG1, IgG2a and IgG2b antibodies reactive against their respective antigens at dilutions in excess of 1:18,000. However, only HBc fusions containing NadA elicited the production of antibodies with serum bactericidal activity. It is hypothesized that this improved immune response is attributable to the adoption of a more native-like folding of crucial conformational epitopes of NadA within the chimeric VLP. This work demonstrates that HBc can incorporate insertions of large antigen domains but that maintenance of their three-dimensional structure is likely to be critical in obtaining a protective response.
Collapse
Affiliation(s)
- Sebastian Aston-Deaville
- 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
| | - Emil Carlsson
- 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
| | - Muhammad Saleem
- 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
| | - Angela Thistlethwaite
- 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
| | - Hannah Chan
- National Institute for Biological Standards and Control, Blanche Lane, South Mimms, Hertfordshire EN6 3QG, UK
| | - Sunil Maharjan
- National Institute for Biological Standards and Control, Blanche Lane, South Mimms, Hertfordshire EN6 3QG, UK
| | - Alessandra Facchetti
- National Institute for Biological Standards and Control, Blanche Lane, South Mimms, Hertfordshire EN6 3QG, UK
| | - Ian M Feavers
- National Institute for Biological Standards and Control, Blanche Lane, South Mimms, Hertfordshire EN6 3QG, UK
| | - C Alistair Siebert
- Electron Bio-Imaging Centre, Diamond Light Source, Harwell Science & Innovation Campus, Didcot, Oxfordshire, UK
| | - Richard F Collins
- 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
| | - Alan Roseman
- 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
| | - 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
|
38
|
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
|
39
|
Beernink PT. Effect of complement Factor H on antibody repertoire and protection elicited by meningococcal capsular group B vaccines containing Factor H binding protein. Hum Vaccin Immunother 2020; 16:703-712. [PMID: 31526219 DOI: 10.1080/21645515.2019.1664241] [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: 10/26/2022] Open
Abstract
Bacteria produce surface ligands for host complement regulators including Factor H (FH), which allows the bacteria to evade immunity. Meningococcal Factor H binding protein (FHbp) is both a virulence factor and a vaccine antigen. Antibodies to FHbp can neutralize its function by inhibiting binding of FH to the bacteria and confer robust complement-mediated protection. However, in the presence of human or primate FH, antibodies to FHbp do not inhibit FH binding and the protective antibody responses are decreased. This immune suppression can be overcome by modification of the FHbp antigen to decrease FH binding, which modulates the antibody repertoire to inhibit FH binding and increase protection. When FHbp is present at sufficient density on the bacterial surface, two or more antibodies can synergize to activate the complement system. Thus, modification of FHbp antigens to decrease FH binding expands the anti-FHbp antibody repertoire and increases the potential for synergistic activity.
Collapse
Affiliation(s)
- Peter T Beernink
- Center for Immunobiology and Vaccine Development, UCSF Benioff Children's Hospital Oakland, Oakland, CA, USA.,Department of Pediatrics, School of Medicine, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
40
|
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
|
41
|
Xu Z, Moyle PM. A Self‐Adjuvanting Vaccine Platform: Optimization of Site‐Specific Sortase A Mediated Conjugation of Toll‐Like Receptor 2 Ligands onto the Carboxyl or Amino terminus of Recombinant Protein Antigens. Chempluschem 2020; 85:227-236. [DOI: 10.1002/cplu.201900687] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 01/13/2020] [Indexed: 12/16/2022]
Affiliation(s)
- Zhenghui Xu
- Pharmacy Australia Centre of Excellence School of Pharmacy The University of Queensland 20 Cornwall St Woolloongabba QLD 4102 Australia
| | - Peter Michael Moyle
- Pharmacy Australia Centre of Excellence School of Pharmacy The University of Queensland 20 Cornwall St Woolloongabba QLD 4102 Australia
| |
Collapse
|
42
|
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
|
43
|
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
|
44
|
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
|
45
|
Boccalini S, Bechini A, Sartor G, Paolini D, Innocenti M, Bonanni P, Panatto D, Lai PL, Zangrillo F, Marchini F, Lecini E, Iovine M, Amicizia D, Landa P. [Health Technology Assessment of meningococcal B vaccine (Trumenba ®) in adolescent in Italy]. JOURNAL OF PREVENTIVE MEDICINE AND HYGIENE 2019; 60:E1-E94. [PMID: 32047867 PMCID: PMC7007189 DOI: 10.15167/2421-4248/jpmh2019.60.3s2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- S Boccalini
- Dipartimento di Scienze della Salute, Università degli Studi di Firenze
| | - A Bechini
- Dipartimento di Scienze della Salute, Università degli Studi di Firenze
| | - G Sartor
- Dipartimento di Scienze della Salute, Università degli Studi di Firenze
| | - D Paolini
- Dipartimento di Scienze della Salute, Università degli Studi di Firenze
| | - M Innocenti
- Dipartimento di Scienze della Salute, Università degli Studi di Firenze
| | - P Bonanni
- Dipartimento di Scienze della Salute, Università degli Studi di Firenze
| | - D Panatto
- Dipartimento di Scienze della Salute, Università degli Studi di Genova
| | - P L Lai
- Dipartimento di Scienze della Salute, Università degli Studi di Genova
| | - F Zangrillo
- Dipartimento di Scienze della Salute, Università degli Studi di Genova
| | - F Marchini
- Dipartimento di Scienze della Salute, Università degli Studi di Genova
| | - E Lecini
- Dipartimento di Scienze della Salute, Università degli Studi di Genova
| | - M Iovine
- Dipartimento di Scienze della Salute, Università degli Studi di Genova
| | - D Amicizia
- Dipartimento di Scienze della Salute, Università degli Studi di Genova
| | - P Landa
- Dipartimento di Economia, Università degli Studi di Genova
| |
Collapse
|
46
|
Kurtovic L, Boyle MJ, Opi DH, Kennedy AT, Tham WH, Reiling L, Chan JA, Beeson JG. Complement in malaria immunity and vaccines. Immunol Rev 2019; 293:38-56. [PMID: 31556468 PMCID: PMC6972673 DOI: 10.1111/imr.12802] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 09/03/2019] [Indexed: 12/20/2022]
Abstract
Developing efficacious vaccines for human malaria caused by Plasmodium falciparum is a major global health priority, although this has proven to be immensely challenging over the decades. One major hindrance is the incomplete understanding of specific immune responses that confer protection against disease and/or infection. While antibodies to play a crucial role in malaria immunity, the functional mechanisms of these antibodies remain unclear as most research has primarily focused on the direct inhibitory or neutralizing activity of antibodies. Recently, there is a growing body of evidence that antibodies can also mediate effector functions through activating the complement system against multiple developmental stages of the parasite life cycle. These antibody‐complement interactions can have detrimental consequences to parasite function and viability, and have been significantly associated with protection against clinical malaria in naturally acquired immunity, and emerging findings suggest these mechanisms could contribute to vaccine‐induced immunity. In order to develop highly efficacious vaccines, strategies are needed that prioritize the induction of antibodies with enhanced functional activity, including the ability to activate complement. Here we review the role of complement in acquired immunity to malaria, and provide insights into how this knowledge could be used to harness complement in malaria vaccine development.
Collapse
Affiliation(s)
- Liriye Kurtovic
- Burnet Institute, Melbourne, Vic., Australia.,Central Clinical School, Monash University, Melbourne, Vic., Australia
| | | | | | - Alexander T Kennedy
- Walter and Eliza Hall Institute, Melbourne, Vic., Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Vic., Australia
| | - Wai-Hong Tham
- Walter and Eliza Hall Institute, Melbourne, Vic., Australia
| | | | - Jo-Anne Chan
- Burnet Institute, Melbourne, Vic., Australia.,Central Clinical School, Monash University, Melbourne, Vic., Australia
| | - James G Beeson
- Burnet Institute, Melbourne, Vic., Australia.,Central Clinical School, Monash University, Melbourne, Vic., Australia.,Department of Microbiology, Monash University, Clayton, Vic., Australia.,Department of Medicine, The University of Melbourne, Parkville, Vic., Australia
| |
Collapse
|
47
|
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
|
48
|
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
|
49
|
Beernink PT, Ispasanie E, Lewis LA, Ram S, Moe GR, Granoff DM. A Meningococcal Native Outer Membrane Vesicle Vaccine With Attenuated Endotoxin and Overexpressed Factor H Binding Protein Elicits Gonococcal Bactericidal Antibodies. J Infect Dis 2019; 219:1130-1137. [PMID: 30346576 PMCID: PMC6420169 DOI: 10.1093/infdis/jiy609] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 10/12/2018] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Meningococcal outer membrane vesicle (OMV) vaccines are prepared with detergents to remove endotoxin, which also remove desirable antigens such as factor H binding protein (FHbp). Native OMV (NOMV) vaccines with genetically attenuated endotoxin do not require detergent treatment and elicit broader serum bactericidal antibody (SBA) responses than OMV or recombinant FHbp (rFHbp) vaccines. METHODS We measured human complement-mediated SBA responses in mice immunized with NOMV with overexpressed FHbp subfamily B (NOMV-FHbp), NOMV with FHbp genetically inactivated (NOMV-KO), and/or a control rFHbp vaccine against meningococcal and gonococcal strains. RESULTS Despite having 36-fold less FHbp per dose, the NOMV-FHbp vaccine elicited a ≥3-fold higher serum IgG anti-FHbp geometric mean titer than control vaccines containing rFHbp (P ≤ .003). Against 2 meningococcal outbreak strains with mismatched PorA and heterologous FHbp subfamily B sequence variants, the NOMV-FHbp vaccine produced ≥30-fold higher SBA titers than control vaccines. Mice immunized with NOMV-FHbp and NOMV-KO vaccines also elicited SBA against a gonococcal strain (P < .0001 vs the adjuvant-only control group). In contrast, 2 licensed meningococcal serogroup B vaccines, including one containing detergent-extracted OMV, did not produce gonococcal SBA in humans. CONCLUSIONS A meningococcal NOMV vaccine elicits SBA against gonococci and with overexpressed FHbp elicits SBA against meningococci.
Collapse
Affiliation(s)
- Peter T Beernink
- Center for Immunobiology and Vaccine Development, University of California–San Francisco (UCSF) Benioff Children’s Hospital, Oakland
- Department of Pediatrics, School of Medicine, UCSF, San Francisco
| | - Emma Ispasanie
- Center for Immunobiology and Vaccine Development, University of California–San Francisco (UCSF) Benioff Children’s Hospital, Oakland
| | - Lisa A Lewis
- Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester
| | - Sanjay Ram
- Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester
| | - Gregory R Moe
- Center for Immunobiology and Vaccine Development, University of California–San Francisco (UCSF) Benioff Children’s Hospital, Oakland
- Department of Pediatrics, School of Medicine, UCSF, San Francisco
| | - Dan M Granoff
- Center for Immunobiology and Vaccine Development, University of California–San Francisco (UCSF) Benioff Children’s Hospital, Oakland
| |
Collapse
|
50
|
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
|