1
|
Caradonna V, Pinto M, Alfini R, Giannelli C, Iturriza M, Micoli F, Rossi O, Mancini F. High-Throughput Luminescence-Based Serum Bactericidal Assay Optimization and Characterization to Assess Human Sera Functionality Against Multiple Shigella flexneri Serotypes. Int J Mol Sci 2024; 25:11123. [PMID: 39456904 PMCID: PMC11508014 DOI: 10.3390/ijms252011123] [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: 09/02/2024] [Revised: 10/02/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
Shigellosis represents a significant global health concern particularly affecting children under 5 years in low- and middle-income countries (LMICs) and is associated with stunting and antimicrobial resistance. There is a critical need for an effective vaccine offering broad protection against the different Shigella serotypes. A correlate of protection has not yet been established but there is a general consensus about the relevant role of anti-O-Antigen-specific IgG and its functionality evaluated by the Serum Bactericidal Assay (SBA). This study aims to characterize a high-throughput luminescence-based SBA (L-SBA) against seven widespread Shigella serotypes. The assay was previously developed and characterized for S. sonnei and S. flexneri 1b, 2a, and 3a and has now been refined and extended to an additional five serotypes (S. flexneri 4a, 5b, 6, X, and Y). The characterization of the assay with human sera confirmed the repeatability, intermediate precision, and linearity of the assays; both homologous and heterologous specificity were verified as well; finally, limit of detection and quantification were established for all assays. Moreover, different sources of baby rabbit complement showed to have no impact on L-SBA output. The results obtained confirm the possibility of extending the L-SBA to multiple Shigella serotypes, thus enabling analysis of the functional response induced by natural exposure to Shigella in epidemiological studies and the ability of candidate vaccines to elicit cross-functional antibodies able to kill a broad panel of prevalent Shigella serotypes in a complement-mediated fashion.
Collapse
Affiliation(s)
- Valentina Caradonna
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy;
- GSK Vaccines Institute for Global Health (GVGH), 53100 Siena, Italy (R.A.); (C.G.)
| | - Marika Pinto
- GSK Vaccines Institute for Global Health (GVGH), 53100 Siena, Italy (R.A.); (C.G.)
| | - Renzo Alfini
- GSK Vaccines Institute for Global Health (GVGH), 53100 Siena, Italy (R.A.); (C.G.)
| | - Carlo Giannelli
- GSK Vaccines Institute for Global Health (GVGH), 53100 Siena, Italy (R.A.); (C.G.)
| | - Miren Iturriza
- GSK Vaccines Institute for Global Health (GVGH), 53100 Siena, Italy (R.A.); (C.G.)
| | - Francesca Micoli
- GSK Vaccines Institute for Global Health (GVGH), 53100 Siena, Italy (R.A.); (C.G.)
| | - Omar Rossi
- GSK Vaccines Institute for Global Health (GVGH), 53100 Siena, Italy (R.A.); (C.G.)
| | - Francesca Mancini
- GSK Vaccines Institute for Global Health (GVGH), 53100 Siena, Italy (R.A.); (C.G.)
| |
Collapse
|
2
|
Escribano MP, Balado M, Santos B, Toranzo AE, Lemos ML, Magariños B. Outer membrane vesicles (OMVs) from Tenacibaculum maritimum as a potential vaccine against fish tenacibaculosis. FISH & SHELLFISH IMMUNOLOGY 2024; 154:109943. [PMID: 39370018 DOI: 10.1016/j.fsi.2024.109943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/10/2024] [Accepted: 10/04/2024] [Indexed: 10/08/2024]
Abstract
Outer membrane vesicles (OMVs) have been gained increasing attention in vaccinology due to their ability to induce strong protective humoral and cell-mediated immunity. The Gram-negative bacterium Tenacibaculum maritimum, the causative agent of marine tenacibaculosis, poses a significant challenge to the global aquaculture industry due to its difficult prophylaxis. In previous studies, we demonstrated that OMV production is a key virulence mechanism in T. maritimum. Building on this, the present study aimed to evaluate the efficacy of a natural, encapsulated multi-antigen vaccine made from adjuvant-free, crude T. maritimum OMVs (Tm-OMVs). A vaccination experiment using SP9.1-OMVs was conducted in juvenile turbot (Scophthalmus maximus L.), followed by a T. maritimum bath challenge. Immune responses in the turbot were assessed by measuring anti-Tm antibody levels and analyzing the expression of eight key immune-related genes (il-1β, il-8, il-22, pcna, c3, cd4-1, ifng2, cd8α). The results showed that immunization with SP9.1-OMVs provided significant protection against T. maritimum infection (RPS = 70 %). Vaccinated fish exhibited a dose-dependent increase in anti-Tm antibody titers in blood plasma, along with rapid induction of both innate (il-1β, il-8, il-22, c3) and adaptive (cd4-1, ifng2, cd8α) immune genes as early as 4 h post-bath challenge. These findings offer new insights into the early immune response of turbot following T. maritimum infection and could serve as a foundation for developing novel OMV-based vaccines.
Collapse
Affiliation(s)
- M Pilar Escribano
- Department of Microbiology and Parasitology, Aquatic One Health Research Center (iARCUS) and Faculty of Biology-CIBUS, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Miguel Balado
- Department of Microbiology and Parasitology, Aquatic One Health Research Center (iARCUS) and Faculty of Biology-CIBUS, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Beatriz Santos
- Department of Microbiology and Parasitology, Aquatic One Health Research Center (iARCUS) and Faculty of Biology-CIBUS, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Alicia E Toranzo
- Department of Microbiology and Parasitology, Aquatic One Health Research Center (iARCUS) and Faculty of Biology-CIBUS, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Manuel L Lemos
- Department of Microbiology and Parasitology, Aquatic One Health Research Center (iARCUS) and Faculty of Biology-CIBUS, University of Santiago de Compostela, Santiago de Compostela, Spain.
| | - Beatriz Magariños
- Department of Microbiology and Parasitology, Aquatic One Health Research Center (iARCUS) and Faculty of Biology-CIBUS, University of Santiago de Compostela, Santiago de Compostela, Spain.
| |
Collapse
|
3
|
Tong Z, Zhang X, Guo X, Wu G, Cao S, Zhang Y, Meng X, Wang T, Wang Y, Song Y, Yang R, Du Z. Delivery of Yersinia pestis antigens via Escherichia coli outer membrane vesicles offered improved protection against plague. mSphere 2024; 9:e0033024. [PMID: 39158304 PMCID: PMC11423571 DOI: 10.1128/msphere.00330-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 07/09/2024] [Indexed: 08/20/2024] Open
Abstract
Outer membrane vesicles (OMVs) from Gram-negative bacteria can be used as a vaccine platform to deliver heterologous antigens. Here, the major protective antigens of Yersinia pestis, F1 and LcrV, were fused either with the leader sequence or the transmembrane domain of the outer membrane protein A (OmpA), resulting in chimeric proteins OmpA-ls-F1V and OmpA46-159-F1V, respectively. We show that OmpA-ls-F1V and OmpA46-159-F1V can be successfully delivered into the lumen and membrane of the OMVs of Escherichia coli, respectively. Mutation of ompA but not tolR in E. coli enhanced the delivery efficiency of OmpA-ls-F1V into OMVs. The OmpA-ls-F1V protein comprises up to 20% of the total protein in OMVs derived from the ompA mutant (OMVdA-ALS-F1V), a proportion significantly higher than the 1% observed for OmpA46-159-F1V in OMVs produced by an ompA mutant that expresses OmpA46-159-F1V, referred to as OMVdA-LATM5-F1V. Intramuscular (i.m.) immunization of mice with OMVdA-ALS-F1V induced significantly higher levels of serum anti-LcrV and anti-F1 IgG, and provided higher efficacy in protection against subcutaneous (s.c.) Y. pestis infection compared to OMVdA-LATM5-F1V and the purified recombinant F1V (rF1V) protein adsorbed to aluminum hydroxide. The three-dose i.m. immunization with OMVdA-ALS-F1V, administered at 14-day intervals, provides complete protection to mice against s.c. infection with 130 LD50 of Y. pestis 201 and conferred 80% against intranasal (i.n.) challenge with 11.4 LD50 of Y. pestis 201. Taken together, our findings indicate that the engineered OMVs containing F1V fused with the leader sequence of OmpA provide significantly higher protection than rF1V against both s.c. and i.n. infection of Y. pestis and more balanced Th1/Th2 responses.IMPORTANCEThe two major protective antigens of Y. pestis, LcrV and F1, have demonstrated the ability to elicit systemic and local mucosal immune responses as subunit vaccines. However, these vaccines have failed to provide adequate protection against pneumonic plague in African green monkeys. Here, Y. pestis F1 and LcrV antigens were successfully incorporated into the lumen and the surface of the outer membrane vesicles (OMVs) of E. coli by fusion either with the leader sequence or the transmembrane domain of OmpA. We compared the humoral immune response elicited by these OMV formulations and their protective efficacy in mice against Y. pestis. Our results demonstrate that the plague OMV vaccine candidates can induce robust protective immunity against both s.c. and i.n. Y. pestis infections, surpassing the effectiveness of rF1V. In addition, immunization with OMVs generated a relatively balanced Th1/Th2 immune response compared to rF1V immunization. These findings underscore the potential of OMVs-based plague vaccines for further development.
Collapse
Affiliation(s)
- Zehui Tong
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Xiangting Zhang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
- Public Health School, Mudanjiang Medical University, Mudanjiang, China
| | - Xiao Guo
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Gengshan Wu
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Shiyang Cao
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Yuan Zhang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Xiangze Meng
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
- Public Health School, Mudanjiang Medical University, Mudanjiang, China
| | - Tong Wang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Yiqian Wang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Yajun Song
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Ruifu Yang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Zongmin Du
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| |
Collapse
|
4
|
Manoharan S, Farman TA, Piliou S, Mastroeni P. Characterisation and Immunogenicity of Neisseria cinerea outer membrane vesicles displaying NadA, NHBA and fHbp from Neisseria meningitidis serogroup B. Front Immunol 2024; 15:1473064. [PMID: 39380985 PMCID: PMC11458423 DOI: 10.3389/fimmu.2024.1473064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 09/04/2024] [Indexed: 10/10/2024] Open
Abstract
More affordable and effective vaccines against bacterial meningitis caused by Neisseria meningitidis serogroup B are still required for global prevention. We have previously shown that modified outer membrane vesicles (mOMVs) from commensal Neisseria cinerea can be used as a platform to induce immune responses against meningococcal antigens. The aim of the present study was to use a combination of two genetically engineered mOMVs to express multiple antigens from N. meningitidis known to be involved in protective immunity to meningococcal meningitis (different variants of factor H binding protein (fHbp), Neisseria Heparin Binding Antigen (NHBA) and Neisseria Adhesin A (NadA)). Antigen expression in the mOMVs was confirmed by Western blotting; detoxification of the lipooligosaccharide (LOS) was confirmed by measuring human Toll-like receptor 4 (hTLR4) activation using in vitro cell assays. Mice immunised with a combination of two mOMVs expressing fHbp, NHBA and NadA produced antibodies to all the antigens. Furthermore, serum bactericidal activity (SBA) was induced by the immunisation, with mOMVs expressing NadA displaying high SBA titres against a nadA+ MenB strain. The work highlights the potential of mOMVs from N. cinerea to induce functional immune responses against multiple antigens involved in the protective immune response to meningococcal disease.
Collapse
MESH Headings
- Antigens, Bacterial/immunology
- Antigens, Bacterial/genetics
- Animals
- Adhesins, Bacterial/immunology
- Adhesins, Bacterial/genetics
- Neisseria meningitidis, Serogroup B/immunology
- Bacterial Proteins/immunology
- Bacterial Proteins/genetics
- Mice
- Meningococcal Vaccines/immunology
- Humans
- Antibodies, Bacterial/immunology
- Antibodies, Bacterial/blood
- Meningitis, Meningococcal/immunology
- Meningitis, Meningococcal/prevention & control
- Meningitis, Meningococcal/microbiology
- Neisseria cinerea/immunology
- Bacterial Outer Membrane/immunology
- Female
- Extracellular Vesicles/immunology
- Bacterial Outer Membrane Proteins/immunology
- Bacterial Outer Membrane Proteins/genetics
- Mice, Inbred BALB C
- Carrier Proteins
Collapse
Affiliation(s)
- Shathviga Manoharan
- Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | | | | | | |
Collapse
|
5
|
Da Costa RM, Rooke JL, Wells TJ, Cunningham AF, Henderson IR. Type 5 secretion system antigens as vaccines against Gram-negative bacterial infections. NPJ Vaccines 2024; 9:159. [PMID: 39218947 PMCID: PMC11366766 DOI: 10.1038/s41541-024-00953-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 08/14/2024] [Indexed: 09/04/2024] Open
Abstract
Infections caused by Gram-negative bacteria are leading causes of mortality worldwide. Due to the rise in antibiotic resistant strains, there is a desperate need for alternative strategies to control infections caused by these organisms. One such approach is the prevention of infection through vaccination. While live attenuated and heat-killed bacterial vaccines are effective, they can lead to adverse reactions. Newer vaccine technologies focus on utilizing polysaccharide or protein subunits for safer and more targeted vaccination approaches. One promising avenue in this regard is the use of proteins released by the Type 5 secretion system (T5SS). This system is the most prevalent secretion system in Gram-negative bacteria. These proteins are compelling vaccine candidates due to their demonstrated protective role in current licensed vaccines. Notably, Pertactin, FHA, and NadA are integral components of licensed vaccines designed to prevent infections caused by Bordetella pertussis or Neisseria meningitidis. In this review, we delve into the significance of incorporating T5SS proteins into licensed vaccines, their contributions to virulence, conserved structural motifs, and the protective immune responses elicited by these proteins.
Collapse
Affiliation(s)
- Rochelle M Da Costa
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Jessica L Rooke
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Timothy J Wells
- Frazer Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Adam F Cunningham
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Ian R Henderson
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
6
|
Muñoz-Echeverri LM, Benavides-López S, Geiger O, Trujillo-Roldán MA, Valdez-Cruz NA. Bacterial extracellular vesicles: biotechnological perspective for enhanced productivity. World J Microbiol Biotechnol 2024; 40:174. [PMID: 38642254 PMCID: PMC11032300 DOI: 10.1007/s11274-024-03963-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 03/19/2024] [Indexed: 04/22/2024]
Abstract
Bacterial extracellular vesicles (BEVs) are non-replicative nanostructures released by Gram-negative and Gram-positive bacteria as a survival mechanism and inter- and intraspecific communication mechanism. Due to BEVs physical, biochemical, and biofunctional characteristics, there is interest in producing and using them in developing new therapeutics, vaccines, or delivery systems. However, BEV release is typically low, limiting their application. Here, we provide a biotechnological perspective to enhance BEV production, highlighting current strategies. The strategies include the production of hypervesiculating strains through gene modification, bacteria culture under stress conditions, and artificial vesicles production. We discussed the effect of these production strategies on BEVs types, morphology, composition, and activity. Furthermore, we summarized general aspects of BEV biogenesis, functional capabilities, and applications, framing their current importance and the need to produce them in abundance. This review will expand the knowledge about the range of strategies associated with BEV bioprocesses to increase their productivity and extend their application possibilities.
Collapse
Affiliation(s)
- Laura M Muñoz-Echeverri
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México AP. 70228, Ciudad de México, C.P. 04510, México
- Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Unidad de Posgrado, Edificio D, 1° Piso, Circuito de Posgrados, Ciudad Universitaria, Coyoacán CDMX, C.P. 04510, México
| | - Santiago Benavides-López
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México AP. 70228, Ciudad de México, C.P. 04510, México
- Posgrado en Ciencias Biomédicas, Universidad Nacional Autónoma de México, Unidad de Posgrado, Edificio B, 1° Piso, Circuito de Posgrados, Ciudad Universitaria, Coyoacán CDMX, C.P. 04510, México
| | - Otto Geiger
- Centro de Ciencias Genómicas, Universidad Nacional Autónoma de México, Av. Universidad s/n, Cuernavaca, Morelos, CP 62210, México
| | - Mauricio A Trujillo-Roldán
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México AP. 70228, Ciudad de México, C.P. 04510, México
- Centro de Nanociencias y Nanotecnología, Universidad Nacional Autónoma de México, Km 107 Carretera, Tijuana-Ensenada, Baja California, 22860, México
| | - Norma A Valdez-Cruz
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México AP. 70228, Ciudad de México, C.P. 04510, México.
- Centro de Nanociencias y Nanotecnología, Universidad Nacional Autónoma de México, Km 107 Carretera, Tijuana-Ensenada, Baja California, 22860, México.
| |
Collapse
|
7
|
Giannelli C, Necchi F, Palmieri E, Oldrini D, Ricchetti B, Papathanasiou MM, Kis Z, Kontoravdi C, Campa C, Micoli F. Quality by Design Framework Applied to GMMA Purification. AAPS J 2024; 26:32. [PMID: 38459151 DOI: 10.1208/s12248-024-00902-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 02/22/2024] [Indexed: 03/10/2024] Open
Abstract
In recent years, Generalized Modules for Membrane Antigens (GMMA) have received increased attention as an innovative vaccine platform against bacterial pathogens, particularly attractive for low- and middle-income countries because of manufacturing simplicity. The assessment of critical quality attributes (CQAs), product-process interactions, identification of appropriate in process analytical methods, and process modeling is part of a robust quality by design (QbD) framework to support further development and control of manufacturing processes. QbD implementation in the context of the GMMA platform will ensure robust manufacturing of batches with desired characteristics, facilitating technical transfer to local manufacturers, regulatory approval, and commercialization of vaccines based on this technology. Here, we summarize the methodology suggested, applied to a first step of GMMA manufacturing process.
Collapse
Affiliation(s)
- Carlo Giannelli
- GSK Vaccines Institute for Global Health (GVGH), Via Fiorentina 1, 53100, Siena, Italy.
| | - Francesca Necchi
- GSK Vaccines Institute for Global Health (GVGH), Via Fiorentina 1, 53100, Siena, Italy
| | - Elena Palmieri
- GSK Vaccines Institute for Global Health (GVGH), Via Fiorentina 1, 53100, Siena, Italy
| | - Davide Oldrini
- GSK Vaccines Institute for Global Health (GVGH), Via Fiorentina 1, 53100, Siena, Italy
| | - Beatrice Ricchetti
- GSK Vaccines Institute for Global Health (GVGH), Via Fiorentina 1, 53100, Siena, Italy
| | - Maria M Papathanasiou
- The Sargent Centre for Process Systems Engineering, Department of Chemical Engineering, Imperial College London, South Kensington Campus, London, SW7 2AZ, UK
| | - Zoltan Kis
- The Sargent Centre for Process Systems Engineering, Department of Chemical Engineering, Imperial College London, South Kensington Campus, London, SW7 2AZ, UK
- Department of Chemical and Biological Engineering, The University of Sheffield, Mappin Street, Sheffield, S1 3JD, UK
| | - Cleo Kontoravdi
- The Sargent Centre for Process Systems Engineering, Department of Chemical Engineering, Imperial College London, South Kensington Campus, London, SW7 2AZ, UK
| | | | - Francesca Micoli
- GSK Vaccines Institute for Global Health (GVGH), Via Fiorentina 1, 53100, Siena, Italy
| |
Collapse
|
8
|
Lei EK, Azmat A, Henry KA, Hussack G. Outer membrane vesicles as a platform for the discovery of antibodies to bacterial pathogens. Appl Microbiol Biotechnol 2024; 108:232. [PMID: 38396192 PMCID: PMC10891261 DOI: 10.1007/s00253-024-13033-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/24/2024] [Accepted: 01/26/2024] [Indexed: 02/25/2024]
Abstract
Bacterial outer membrane vesicles (OMVs) are nanosized spheroidal particles shed by gram-negative bacteria that contain biomolecules derived from the periplasmic space, the bacterial outer membrane, and possibly other compartments. OMVs can be purified from bacterial culture supernatants, and by genetically manipulating the bacterial cells that produce them, they can be engineered to harbor cargoes and/or display molecules of interest on their surfaces including antigens that are immunogenic in mammals. Since OMV bilayer-embedded components presumably maintain their native structures, OMVs may represent highly useful tools for generating antibodies to bacterial outer membrane targets. OMVs have historically been utilized as vaccines or vaccine constituents. Antibodies that target bacterial surfaces are increasingly being explored as antimicrobial agents either in unmodified form or as targeting moieties for bactericidal compounds. Here, we review the properties of OMVs, their use as immunogens, and their ability to elicit antibody responses against bacterial antigens. We highlight antigens from bacterial pathogens that have been successfully targeted using antibodies derived from OMV-based immunization and describe opportunities and limitations for OMVs as a platform for antimicrobial antibody development. KEY POINTS: • Outer membrane vesicles (OMVs) of gram-negative bacteria bear cell-surface molecules • OMV immunization allows rapid antibody (Ab) isolation to bacterial membrane targets • Review and analysis of OMV-based immunogens for antimicrobial Ab development.
Collapse
Affiliation(s)
- Eric K Lei
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Aruba Azmat
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Kevin A Henry
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Greg Hussack
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada.
| |
Collapse
|
9
|
Effah CY, Ding X, Drokow EK, Li X, Tong R, Sun T. Bacteria-derived extracellular vesicles: endogenous roles, therapeutic potentials and their biomimetics for the treatment and prevention of sepsis. Front Immunol 2024; 15:1296061. [PMID: 38420121 PMCID: PMC10899385 DOI: 10.3389/fimmu.2024.1296061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 01/24/2024] [Indexed: 03/02/2024] Open
Abstract
Sepsis is one of the medical conditions with a high mortality rate and lacks specific treatment despite several years of extensive research. Bacterial extracellular vesicles (bEVs) are emerging as a focal target in the pathophysiology and treatment of sepsis. Extracellular vesicles (EVs) derived from pathogenic microorganisms carry pathogenic factors such as carbohydrates, proteins, lipids, nucleic acids, and virulence factors and are regarded as "long-range weapons" to trigger an inflammatory response. In particular, the small size of bEVs can cross the blood-brain and placental barriers that are difficult for pathogens to cross, deliver pathogenic agents to host cells, activate the host immune system, and possibly accelerate the bacterial infection process and subsequent sepsis. Over the years, research into host-derived EVs has increased, leading to breakthroughs in cancer and sepsis treatments. However, related approaches to the role and use of bacterial-derived EVs are still rare in the treatment of sepsis. Herein, this review looked at the dual nature of bEVs in sepsis by highlighting their inherent functions and emphasizing their therapeutic characteristics and potential. Various biomimetics of bEVs for the treatment and prevention of sepsis have also been reviewed. Finally, the latest progress and various obstacles in the clinical application of bEVs have been highlighted.
Collapse
Affiliation(s)
- Clement Yaw Effah
- Department of Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Henan Engineering Research Center for Critical Care Medicine, Henan Key Laboratory of Critical Care Medicine, Zhengzhou, China
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Henan Engineering Research Center for Critical Care Medicine, Henan Key Laboratory of Critical Care Medicine, Zhengzhou, China
- Zhengzhou Key Laboratory of Sepsis, Henan Sepsis Diagnosis and Treatment Center, Henan Key Laboratory of Sepsis in Health Commission, Zhengzhou, China
| | - Xianfei Ding
- Department of Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Henan Engineering Research Center for Critical Care Medicine, Henan Key Laboratory of Critical Care Medicine, Zhengzhou, China
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Henan Engineering Research Center for Critical Care Medicine, Henan Key Laboratory of Critical Care Medicine, Zhengzhou, China
- Zhengzhou Key Laboratory of Sepsis, Henan Sepsis Diagnosis and Treatment Center, Henan Key Laboratory of Sepsis in Health Commission, Zhengzhou, China
| | - Emmanuel Kwateng Drokow
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Department of Epidemiology and Biostatistics, Xiangya School of Public Health, Central South University, Changsha, Hunan, China
| | - Xiang Li
- Department of Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Henan Engineering Research Center for Critical Care Medicine, Henan Key Laboratory of Critical Care Medicine, Zhengzhou, China
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Henan Engineering Research Center for Critical Care Medicine, Henan Key Laboratory of Critical Care Medicine, Zhengzhou, China
- Zhengzhou Key Laboratory of Sepsis, Henan Sepsis Diagnosis and Treatment Center, Henan Key Laboratory of Sepsis in Health Commission, Zhengzhou, China
| | - Ran Tong
- Department of Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Henan Engineering Research Center for Critical Care Medicine, Henan Key Laboratory of Critical Care Medicine, Zhengzhou, China
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Henan Engineering Research Center for Critical Care Medicine, Henan Key Laboratory of Critical Care Medicine, Zhengzhou, China
- Zhengzhou Key Laboratory of Sepsis, Henan Sepsis Diagnosis and Treatment Center, Henan Key Laboratory of Sepsis in Health Commission, Zhengzhou, China
| | - Tongwen Sun
- Department of Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Henan Engineering Research Center for Critical Care Medicine, Henan Key Laboratory of Critical Care Medicine, Zhengzhou, China
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Henan Engineering Research Center for Critical Care Medicine, Henan Key Laboratory of Critical Care Medicine, Zhengzhou, China
- Zhengzhou Key Laboratory of Sepsis, Henan Sepsis Diagnosis and Treatment Center, Henan Key Laboratory of Sepsis in Health Commission, Zhengzhou, China
| |
Collapse
|
10
|
Kapulu MC, Muthumbi E, Otieno E, Rossi O, Ferruzzi P, Necchi F, Acquaviva A, Martin LB, Orindi B, Mwai K, Kibet H, Mwanzu A, Bigogo GM, Verani JR, Mbae C, Nyundo C, Agoti CN, Nakakana UN, Conti V, Bejon P, Kariuki S, Scott JAG, Micoli F, Podda A. Age-dependent acquisition of IgG antibodies to Shigella serotypes-a retrospective analysis of seroprevalence in Kenyan children with implications for infant vaccination. Front Immunol 2024; 15:1340425. [PMID: 38361949 PMCID: PMC10867106 DOI: 10.3389/fimmu.2024.1340425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 01/12/2024] [Indexed: 02/17/2024] Open
Abstract
Background Shigellosis mainly affects children under 5 years of age living in low- and middle-income countries, who are the target population for vaccination. There are, however, limited data available to define the appropriate timing for vaccine administration in this age group. Information on antibody responses following natural infection, proxy for exposure, could help guide vaccination strategies. Methods We undertook a retrospective analysis of antibodies to five of the most prevalent Shigella serotypes among children aged <5 years in Kenya. Serum samples from a cross-sectional serosurvey in three Kenyan sites (Nairobi, Siaya, and Kilifi) were analyzed by standardized ELISA to measure IgG against Shigella sonnei and Shigella flexneri 1b, 2a, 3a, and 6. We identified factors associated with seropositivity to each Shigella serotype, including seropositivity to other Shigella serotypes. Results A total of 474 samples, one for each participant, were analyzed: Nairobi (n = 169), Siaya (n = 185), and Kilifi (n = 120). The median age of the participants was 13.4 months (IQR 7.0-35.6), and the male:female ratio was 1:1. Geometric mean concentrations (GMCs) for each serotype increased with age, mostly in the second year of life. The overall seroprevalence of IgG antibodies increased with age except for S. flexneri 6 which was high across all age subgroups. In the second year of life, there was a statistically significant increase of antibody GMCs against all five serotypes (p = 0.01-0.0001) and a significant increase of seroprevalence for S. flexneri 2a (p = 0.006), S. flexneri 3a (p = 0.006), and S. sonnei (p = 0.05) compared with the second part of the first year of life. Among all possible pairwise comparisons of antibody seropositivity, there was a significant association between S. flexneri 1b and 2a (OR = 6.75, 95% CI 3-14, p < 0.001) and between S. flexneri 1b and 3a (OR = 23.85, 95% CI 11-54, p < 0.001). Conclusion Children living in low- and middle-income settings such as Kenya are exposed to Shigella infection starting from the first year of life and acquire serotype-specific antibodies against multiple serotypes. The data from this study suggest that Shigella vaccination should be targeted to infants, ideally at 6 or at least 9 months of age, to ensure children are protected in the second year of life when exposure significantly increases.
Collapse
Affiliation(s)
- Melissa C. Kapulu
- KEMRI-Wellcome Trust Programme, Kilifi, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Esther Muthumbi
- KEMRI-Wellcome Trust Programme, Kilifi, Kenya
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | | | - Omar Rossi
- GSK Vaccines Institute for Global Health, Siena, Italy
| | | | | | | | | | | | - Kennedy Mwai
- KEMRI-Wellcome Trust Programme, Kilifi, Kenya
- Epidemiology and Biostatistics Division, School of Public Health, University of the Witwatersrand, Johannesburg, South Africa
| | | | | | - Godfrey M. Bigogo
- Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Jennifer R. Verani
- Division of Global Health Protection, US Centers for Disease Control and Prevention, Nairobi, Kenya
| | - Cecilia Mbae
- Centre for Microbiology Research, Kenya Medical Research Institute, Nairobi, Kenya
| | | | | | | | | | - Philip Bejon
- KEMRI-Wellcome Trust Programme, Kilifi, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Samuel Kariuki
- Centre for Microbiology Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - J. Anthony G. Scott
- KEMRI-Wellcome Trust Programme, Kilifi, Kenya
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | | | - Audino Podda
- GSK Vaccines Institute for Global Health, Siena, Italy
| |
Collapse
|
11
|
Tondi S, Siena E, Essaghir A, Bozzetti B, Bechtold V, Scaillet A, Clemente B, Marrocco M, Sammicheli C, Tavarini S, Micoli F, Oldrini D, Pezzicoli A, Di Fede M, Brazzoli M, Ulivieri C, Schiavetti F. Molecular Signature of Monocytes Shaped by the Shigella sonnei 1790-Generalized Modules for Membrane Antigens Vaccine. Int J Mol Sci 2024; 25:1116. [PMID: 38256189 PMCID: PMC10816432 DOI: 10.3390/ijms25021116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 12/13/2023] [Accepted: 12/20/2023] [Indexed: 01/24/2024] Open
Abstract
Shigellosis, an acute gastroenteritis infection caused by Shigella species, remains a public health burden in developing countries. Recently, many outbreaks due to Shigella sonnei multidrug-resistant strains have been reported in high-income countries, and the lack of an effective vaccine represents a major hurdle to counteract this bacterial pathogen. Vaccine candidates against Shigella sonnei are under clinical development, including a Generalized Modules for Membrane Antigens (GMMA)-based vaccine. The mechanisms by which GMMA-based vaccines interact and activate human immune cells remain elusive. Our previous study provided the first evidence that both adaptive and innate immune cells are targeted and functionally shaped by the GMMA-based vaccine. Here, flow cytometry and confocal microscopy analysis allowed us to identify monocytes as the main target population interacting with the S. sonnei 1790-GMMA vaccine on human peripheral blood. In addition, transcriptomic analysis of this cell population revealed a molecular signature induced by 1790-GMMA mostly correlated with the inflammatory response and cytokine-induced processes. This also impacts the expression of genes associated with macrophages' differentiation and T cell regulation, suggesting a dual function for this vaccine platform both as an antigen carrier and as a regulator of immune cell activation and differentiation.
Collapse
Affiliation(s)
- Serena Tondi
- Preclinical Research & Development, GSK, 53100 Siena, Italy; (S.T.)
- Department of Life Sciences, University of Siena, 53100 Siena, Italy
| | - Emilio Siena
- Preclinical Research & Development, GSK, 53100 Siena, Italy; (S.T.)
| | - Ahmed Essaghir
- Preclinical Research & Development, GSK, 53100 Siena, Italy; (S.T.)
| | - Benoît Bozzetti
- Preclinical Research & Development, GSK, 1330 Rixensart, Belgium
| | - Viviane Bechtold
- Preclinical Research & Development, GSK, 1330 Rixensart, Belgium
| | - Aline Scaillet
- Preclinical Research & Development, GSK, 1330 Rixensart, Belgium
| | - Bruna Clemente
- Preclinical Research & Development, GSK, 53100 Siena, Italy; (S.T.)
| | - Mariateresa Marrocco
- Preclinical Research & Development, GSK, 53100 Siena, Italy; (S.T.)
- Department of Life Sciences, University of Siena, 53100 Siena, Italy
| | | | - Simona Tavarini
- Preclinical Research & Development, GSK, 53100 Siena, Italy; (S.T.)
| | - Francesca Micoli
- GSK Vaccines Institute for Global Health S.R.L. (GVGH), 53100 Siena, Italy
| | - Davide Oldrini
- GSK Vaccines Institute for Global Health S.R.L. (GVGH), 53100 Siena, Italy
| | | | - Martina Di Fede
- Preclinical Research & Development, GSK, 53100 Siena, Italy; (S.T.)
| | - Michela Brazzoli
- Preclinical Research & Development, GSK, 53100 Siena, Italy; (S.T.)
| | - Cristina Ulivieri
- Department of Life Sciences, University of Siena, 53100 Siena, Italy
| | | |
Collapse
|
12
|
Abstract
Outer membrane vesicles (OMVs) are spontaneously released by many gram-negative bacteria during their growth and constitute an important virulence factor for bacteria, helping them to survive through harsh environmental conditions. Native OMVs, naturally-released from bacteria, are produced at a level too low for vaccine manufacturing, requiring chemical treatment (detergent-extracted) or genetic manipulation, resulting in generalized modules for membrane antigens (GMMAs). Over the years, the nature and properties of OMVs have made them a viable platform for vaccine development. There are a few licensed OMV vaccines mainly for the prevention of meningitis caused by Neisseria meningitidis serogroup B (MenB) and Haemophilus influenzae type b (Hib). There are several candidates in clinical development against other gram-negative organisms from which the OMVs are derived, but also against heterologous targets in which the OMVs are used as carriers (e.g. coronavirus disease 2019 [COVID-19]). The use of OMVs for targets other than those from which they are derived is a major advancement in OMV technology, improving its versatility by being able to deliver protein or polysaccharide antigens. Other advances include the range of genetic modifications that can be made to improve their safety, reduce reactogenicity, and increase immunogenicity and protective efficacy. However, significant challenges remain, such as identification of general tools for high-content surface expression of heterologous proteins on the OMV surface. Here, we outline the progress of OMV vaccines to date, particularly discussing licensed OMV-based vaccines and candidates in clinical development. Recent trends in preclinical research are described, mainly focused on genetic manipulation and chemical conjugation for the use of OMVs as carriers for heterologous protein and polysaccharide antigens. Remaining challenges with the use of OMVs and directions for future research are also discussed.
Collapse
Affiliation(s)
- Francesca Micoli
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., Siena, Italy.
| | | | - Usman Nakakana
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., Siena, Italy
| |
Collapse
|
13
|
Liu J, Tian R, Sun C, Guo Y, Dong L, Li Y, Song X. Microbial metabolites are involved in tumorigenesis and development by regulating immune responses. Front Immunol 2023; 14:1290414. [PMID: 38169949 PMCID: PMC10758836 DOI: 10.3389/fimmu.2023.1290414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 12/04/2023] [Indexed: 01/05/2024] Open
Abstract
The human microbiota is symbiotic with the host and can create a variety of metabolites. Under normal conditions, microbial metabolites can regulate host immune function and eliminate abnormal cells in a timely manner. However, when metabolite production is abnormal, the host immune system might be unable to identify and get rid of tumor cells at the early stage of carcinogenesis, which results in tumor development. The mechanisms by which intestinal microbial metabolites, including short-chain fatty acids (SCFAs), microbial tryptophan catabolites (MTCs), polyamines (PAs), hydrogen sulfide, and secondary bile acids, are involved in tumorigenesis and development by regulating immune responses are summarized in this review. SCFAs and MTCs can prevent cancer by altering the expression of enzymes and epigenetic modifications in both immune cells and intestinal epithelial cells. MTCs can also stimulate immune cell receptors to inhibit the growth and metastasis of the host cancer. SCFAs, MTCs, bacterial hydrogen sulfide and secondary bile acids can control mucosal immunity to influence the occurrence and growth of tumors. Additionally, SCFAs, MTCs, PAs and bacterial hydrogen sulfide can also affect the anti-tumor immune response in tumor therapy by regulating the function of immune cells. Microbial metabolites have a good application prospect in the clinical diagnosis and treatment of tumors, and our review provides a good basis for related research.
Collapse
Affiliation(s)
- Jiahui Liu
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
| | - Ruxian Tian
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
| | - Caiyu Sun
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
| | - Ying Guo
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
| | - Lei Dong
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
| | - Yumei Li
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
| | - Xicheng Song
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
| |
Collapse
|
14
|
Piliou S, Farman TA, Marini A, Manoharan S, Mastroeni P. Commensal Neisseria cinerea outer membrane vesicles as a platform for the delivery of meningococcal and gonococcal antigens to the immune system. Vaccine 2023; 41:7671-7681. [PMID: 38008665 DOI: 10.1016/j.vaccine.2023.11.034] [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: 09/18/2023] [Revised: 11/17/2023] [Accepted: 11/17/2023] [Indexed: 11/28/2023]
Abstract
An affordable, accessible, and broadly protective vaccine is required to tackle the re-occurring bacterial meningococcal epidemics in Sub-Saharan Africa as well as an effective control of multi-drug resistant strains of gonococcus. Outer membrane vesicles (OMVs) secreted from Gram-negative bacteria represent an attractive platform for antigen delivery to the immune system and therefore for development of multi-component vaccines. In this study, we describe the generation of modified OMVs (mOMVs) from commensal biosafety-level 1 (BSL-1) Neisseria cinerea ATCC® 14685TM, which is phylogenetically close to the pathogenic bacteria Neisseria meningitidis and Neisseria gonorrhoeae. mOMVs were prepared from N. cinerea engineered to express heterologous antigens from N. meningitidis (factor H binding protein (fHbp) and Neisseria Heparin Binding Antigen (NHBA-2)) and from N. gonorrhoeae (NHBA-542). Mice immunised with the mOMVs produced antibodies against fHbp and NHBA. The work indicates that mOMV from N. cinerea can be used as a platform to induce immune responses against antigens involved in the protective immune response against meningococcal and gonococcal diseases.
Collapse
Affiliation(s)
- Stavroula Piliou
- Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, UK
| | - Theo A Farman
- Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, UK
| | - Arianna Marini
- Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, UK
| | - Shathviga Manoharan
- Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, UK.
| | - Pietro Mastroeni
- Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, UK
| |
Collapse
|
15
|
Di Benedetto R, Mancini F, Caradonna V, Aruta MG, Giannelli C, Rossi O, Micoli F. Comparison of Shigella GMMA and glycoconjugate four-component formulations in animals. Front Mol Biosci 2023; 10:1284515. [PMID: 38046812 PMCID: PMC10690372 DOI: 10.3389/fmolb.2023.1284515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 10/30/2023] [Indexed: 12/05/2023] Open
Abstract
Shigellosis is leading bacterial cause of diarrhea with high prevalence in children younger than 5 years in low- and middle-income countries, and increasing number of reports of Shigella cases associated to anti-microbial resistance. No vaccines against Shigella are still licensed, but different candidates based on the O-antigen portion of lipopolysaccharides are in clinic. Generalized Modules for Membrane Antigens (GMMA) have been proposed as an alternative delivery system for the O-antigen, and a 4-component vaccine candidate (altSonflex1-2-3), containing GMMA from S. sonnei and S. flexneri 1b, 2a and 3a is being tested in a phase 1/2 clinical trial, with the aim to elicit broad protection against the most prevalent Shigella serotypes. Here, the 4-component GMMA vaccine candidate has been compared to a more traditional glycoconjugate formulation for the ability to induce functional antibodies in mice and rabbits. In mice, in the absence of Alhydrogel, GMMA induce higher IgG antibodies than glycoconjugates and stronger bactericidal titers against all Shigella serotypes. In the presence of Alhydrogel, GMMA induce O-antigen specific IgG levels similar to traditional glycoconjugates, but with a broader range of IgG subclasses, resulting in stronger bactericidal activity. In rabbits, GMMA elicit higher functional antibodies than glycoconjugates against S. sonnei, and similar responses to S. flexneri 1b, 2a and 3a, independently from the presence of Alhydrogel. Different O-antigen based vaccines against Shigella are now in clinical stage and it will be of particular interest to understand how the preclinical findings in the different animal models translate in humans.
Collapse
Affiliation(s)
- Roberta Di Benedetto
- GSK Vaccines Institute for Global Health (GVGH), Siena, Italy
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | | | | | | | - Carlo Giannelli
- GSK Vaccines Institute for Global Health (GVGH), Siena, Italy
| | - Omar Rossi
- GSK Vaccines Institute for Global Health (GVGH), Siena, Italy
| | | |
Collapse
|
16
|
Croia L, Boscato Sopetto G, Zanella I, Caproni E, Gagliardi A, Tamburini S, König E, Benedet M, Di Lascio G, Corbellari R, Grandi A, Tomasi M, Grandi G. Immunogenicity of Escherichia coli Outer Membrane Vesicles: Elucidation of Humoral Responses against OMV-Associated Antigens. MEMBRANES 2023; 13:882. [PMID: 37999368 PMCID: PMC10673343 DOI: 10.3390/membranes13110882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/10/2023] [Accepted: 11/11/2023] [Indexed: 11/25/2023]
Abstract
Outer membrane vesicles (OMVs) produced by Gram-negative bacteria have emerged as a novel and flexible vaccine platform. OMVs can be decorated with foreign antigens and carry potent immunostimulatory components. Therefore, after their purification from the culture supernatant, they are ready to be formulated for vaccine use. It has been extensively demonstrated that immunization with engineered OMVs can elicit excellent antibody responses against the heterologous antigens. However, the definition of the conditions necessary to reach the optimal antibody titers still needs to be investigated. Here, we defined the protein concentrations required to induce antigen-specific antibodies, and the amount of antigen and OMVs necessary and sufficient to elicit saturating levels of antigen-specific antibodies. Since not all antigens can be expressed in OMVs, we also investigated the effectiveness of vaccines in which OMVs and purified antigens are mixed together without using any procedure for their physical association. Our data show that in most of the cases OMV-antigen mixtures are very effective in eliciting antigen-specific antibodies. This is probably due to the capacity of OMVs to "absorb" antigens, establishing sufficiently stable interactions that allow antigen-OMV co-presentation to the same antigen presenting cell. In those cases when antigen-OMV interaction is not sufficiently stable, the addition of alum to the formulation guarantees the elicitation of high titers of antigen-specific antibodies.
Collapse
Affiliation(s)
- Lorenzo Croia
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Trento, Italy; (L.C.); (G.B.S.); (I.Z.); (E.K.); (R.C.); (M.T.)
| | - Giulia Boscato Sopetto
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Trento, Italy; (L.C.); (G.B.S.); (I.Z.); (E.K.); (R.C.); (M.T.)
| | - Ilaria Zanella
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Trento, Italy; (L.C.); (G.B.S.); (I.Z.); (E.K.); (R.C.); (M.T.)
| | - Elena Caproni
- Toscana Life Sciences Foundation, Via Fiorentina 1, 53100 Siena, Italy; (E.C.); (A.G.); (S.T.); (M.B.); (G.D.L.); (A.G.)
| | - Assunta Gagliardi
- Toscana Life Sciences Foundation, Via Fiorentina 1, 53100 Siena, Italy; (E.C.); (A.G.); (S.T.); (M.B.); (G.D.L.); (A.G.)
| | - Silvia Tamburini
- Toscana Life Sciences Foundation, Via Fiorentina 1, 53100 Siena, Italy; (E.C.); (A.G.); (S.T.); (M.B.); (G.D.L.); (A.G.)
| | - Enrico König
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Trento, Italy; (L.C.); (G.B.S.); (I.Z.); (E.K.); (R.C.); (M.T.)
| | - Mattia Benedet
- Toscana Life Sciences Foundation, Via Fiorentina 1, 53100 Siena, Italy; (E.C.); (A.G.); (S.T.); (M.B.); (G.D.L.); (A.G.)
| | - Gabriele Di Lascio
- Toscana Life Sciences Foundation, Via Fiorentina 1, 53100 Siena, Italy; (E.C.); (A.G.); (S.T.); (M.B.); (G.D.L.); (A.G.)
| | - Riccardo Corbellari
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Trento, Italy; (L.C.); (G.B.S.); (I.Z.); (E.K.); (R.C.); (M.T.)
| | - Alberto Grandi
- Toscana Life Sciences Foundation, Via Fiorentina 1, 53100 Siena, Italy; (E.C.); (A.G.); (S.T.); (M.B.); (G.D.L.); (A.G.)
- BiOMViS Srl, Via Fiorentina 1, 53100 Siena, Italy
| | - Michele Tomasi
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Trento, Italy; (L.C.); (G.B.S.); (I.Z.); (E.K.); (R.C.); (M.T.)
| | - Guido Grandi
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Trento, Italy; (L.C.); (G.B.S.); (I.Z.); (E.K.); (R.C.); (M.T.)
| |
Collapse
|
17
|
Boerth EM, Gong J, Roffler B, Thompson CM, Song B, Malley SF, Hirsch A, MacLennan CA, Zhang F, Malley R, Lu YJ. Induction of Broad Immunity against Invasive Salmonella Disease by a Quadrivalent Combination Salmonella MAPS Vaccine Targeting Salmonella Enterica Serovars Typhimurium, Enteritidis, Typhi, and Paratyphi A. Vaccines (Basel) 2023; 11:1671. [PMID: 38006003 PMCID: PMC10675568 DOI: 10.3390/vaccines11111671] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/26/2023] [Accepted: 10/28/2023] [Indexed: 11/26/2023] Open
Abstract
Bloodstream infections in low- and middle-income countries (LMICs) are most frequently attributed to invasive Salmonella disease caused by four primary serovars of Salmonella enterica: Typhi, Paratyphi A, Typhimurium, and Enteritidis. We showed previously that a bivalent vaccine targeting S. Typhi and S. Paratyphi A using a Multiple Antigen-Presenting System (MAPS) induced functional antibodies against S. Typhi and S. Paratyphi. In the current study, we describe the preclinical development of a first candidate quadrivalent combination Salmonella vaccine with the potential to cover all four leading invasive Salmonella serotypes. We showed that the quadrivalent Salmonella MAPS vaccine, containing Vi from S. Typhi, O-specific Polysaccharide (OSP) from S. Paratyphi A, S. Enteritidis and S. Typhimurium, combined with the Salmonella-specific protein SseB, elicits robust and functional antibody responses to each of the components of the vaccine. Our data indicates that the application of MAPS technology to the development of vaccines targeting invasive forms of Salmonella is practical and merits additional consideration.
Collapse
Affiliation(s)
- Emily M. Boerth
- Division of Infectious Diseases, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Joyce Gong
- Division of Infectious Diseases, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Becky Roffler
- Division of Infectious Diseases, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Claudette M. Thompson
- Division of Infectious Diseases, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Boni Song
- Division of Infectious Diseases, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Sasha F. Malley
- Division of Infectious Diseases, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Angelika Hirsch
- Division of Infectious Diseases, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Calman A. MacLennan
- Enteric & Diarrheal Diseases, Global Health, Bill & Melinda Gates Foundation, 500 5th Ave. N, Seattle, WA 98109, USA
| | - Fan Zhang
- Division of Infectious Diseases, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Richard Malley
- Division of Infectious Diseases, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Ying-Jie Lu
- Division of Infectious Diseases, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
18
|
Tamburini S, Zhang Y, Gagliardi A, Di Lascio G, Caproni E, Benedet M, Tomasi M, Corbellari R, Zanella I, Croia L, Grandi G, Müller M, Grandi A. Bacterial Outer Membrane Vesicles as a Platform for the Development of a Broadly Protective Human Papillomavirus Vaccine Based on the Minor Capsid Protein L2. Vaccines (Basel) 2023; 11:1582. [PMID: 37896984 PMCID: PMC10611245 DOI: 10.3390/vaccines11101582] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 09/27/2023] [Accepted: 10/08/2023] [Indexed: 10/29/2023] Open
Abstract
Human papillomaviruses (HPVs) are a large family of viruses with a capsid composed of the L1 and L2 proteins, which bind to receptors of the basal epithelial cells and promote virus entry. The majority of sexually active people become exposed to HPV and the virus is the most common cause of cervical cancer. Vaccines are available based on the L1 protein, which self-assembles and forms virus-like particles (VLPs) when expressed in yeast and insect cells. Although very effective, these vaccines are HPV type-restricted and their costs limit broad vaccination campaigns. Recently, vaccine candidates based on the conserved L2 epitope from serotypes 16, 18, 31, 33, 35, 6, 51, and 59 were shown to elicit broadly neutralizing anti-HPV antibodies. In this study, we tested whether E. coli outer membrane vesicles (OMVs) could be successfully decorated with L2 polytopes and whether the engineered OMVs could induce neutralizing antibodies. OMVs represent an attractive vaccine platform owing to their intrinsic adjuvanticity and their low production costs. We show that strings of L2 epitopes could be efficiently expressed on the surface of the OMVs and a polypeptide composed of the L2 epitopes from serotypes 18, 33, 35, and 59 provided a broad cross-protective activity against a large panel of HPV serotypes as determined using pseudovirus neutralization assay. Considering the simplicity of the OMV production process, our work provides a highly effective and inexpensive solution to produce universal anti-HPV vaccines.
Collapse
Affiliation(s)
- Silvia Tamburini
- Department of Cellular, Computation and Integrative of Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Trento, Italy; (S.T.); (M.T.); (R.C.); (I.Z.); (L.C.)
| | - Yueru Zhang
- German Cancer Research Center (DKFZ), Im Neuenheimer Feld 242, 69120 Heidelberg, Germany; (Y.Z.); (M.M.)
| | - Assunta Gagliardi
- Toscana Life Sciences Foundation, Via Fiorentina 1, 53100 Siena, Italy; (A.G.); (G.D.L.); (E.C.); (M.B.); (A.G.)
| | - Gabriele Di Lascio
- Toscana Life Sciences Foundation, Via Fiorentina 1, 53100 Siena, Italy; (A.G.); (G.D.L.); (E.C.); (M.B.); (A.G.)
| | - Elena Caproni
- Toscana Life Sciences Foundation, Via Fiorentina 1, 53100 Siena, Italy; (A.G.); (G.D.L.); (E.C.); (M.B.); (A.G.)
| | - Mattia Benedet
- Toscana Life Sciences Foundation, Via Fiorentina 1, 53100 Siena, Italy; (A.G.); (G.D.L.); (E.C.); (M.B.); (A.G.)
| | - Michele Tomasi
- Department of Cellular, Computation and Integrative of Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Trento, Italy; (S.T.); (M.T.); (R.C.); (I.Z.); (L.C.)
| | - Riccardo Corbellari
- Department of Cellular, Computation and Integrative of Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Trento, Italy; (S.T.); (M.T.); (R.C.); (I.Z.); (L.C.)
| | - Ilaria Zanella
- Department of Cellular, Computation and Integrative of Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Trento, Italy; (S.T.); (M.T.); (R.C.); (I.Z.); (L.C.)
| | - Lorenzo Croia
- Department of Cellular, Computation and Integrative of Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Trento, Italy; (S.T.); (M.T.); (R.C.); (I.Z.); (L.C.)
| | - Guido Grandi
- Department of Cellular, Computation and Integrative of Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Trento, Italy; (S.T.); (M.T.); (R.C.); (I.Z.); (L.C.)
| | - Martin Müller
- German Cancer Research Center (DKFZ), Im Neuenheimer Feld 242, 69120 Heidelberg, Germany; (Y.Z.); (M.M.)
| | - Alberto Grandi
- Toscana Life Sciences Foundation, Via Fiorentina 1, 53100 Siena, Italy; (A.G.); (G.D.L.); (E.C.); (M.B.); (A.G.)
- BiOMViS Srl, Via Fiorentina 1, 53100 Siena, Italy
| |
Collapse
|
19
|
Gurunathan S, Kim JH. Bacterial extracellular vesicles: Emerging nanoplatforms for biomedical applications. Microb Pathog 2023; 183:106308. [PMID: 37595812 DOI: 10.1016/j.micpath.2023.106308] [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: 05/12/2023] [Revised: 08/13/2023] [Accepted: 08/14/2023] [Indexed: 08/20/2023]
Abstract
Bacterial extracellular vesicles (BEVs) are nanosized lipid bilayers generated from membranes that are filled with components derived from bacteria. BEVs are important for the physiology, pathogenicity, and interactions between bacteria and their hosts as well. BEVs represent an important mechanism of transport and interaction between cells. Recent advances in biomolecular nanotechnology have enabled the desired properties to be engineered on the surface of BEVs and decoration with desired and diverse biomolecules and nanoparticles, which have potential biomedical applications. BEVs have been the focus of various fields, including nanovaccines, therapeutic agents, and drug delivery vehicles. In this review, we delineate the fundamental aspects of BEVs, including their biogenesis, cargo composition, function, and interactions with host cells. We comprehensively summarize the factors influencing the biogenesis of BEVs. We further highlight the importance of the isolation, purification, and characterization of BEVs because they are essential processes for potential benefits related to host-microbe interactions. In addition, we address recent advancements in BEVs in biomedical applications. Finally, we provide conclusions and future perspectives as well as highlight the remaining challenges of BEVs for different biomedical applications.
Collapse
Affiliation(s)
- Sangiliyandi Gurunathan
- Department of Biotechnology, Rathinam College of Arts and Science, Rathinam Techzone Campus, Eachanari, Coimbatore, 641 021, Tamil Nadu, India.
| | - Jin-Hoi Kim
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul, 05029, Korea.
| |
Collapse
|
20
|
Grandi A, Tomasi M, Ullah I, Bertelli C, Vanzo T, Accordini S, Gagliardi A, Zanella I, Benedet M, Corbellari R, Di Lascio G, Tamburini S, Caproni E, Croia L, Ravà M, Fumagalli V, Di Lucia P, Marotta D, Sala E, Iannacone M, Kumar P, Mothes W, Uchil PD, Cherepanov P, Bolognesi M, Pizzato M, Grandi G. Immunogenicity and Pre-Clinical Efficacy of an OMV-Based SARS-CoV-2 Vaccine. Vaccines (Basel) 2023; 11:1546. [PMID: 37896949 PMCID: PMC10610814 DOI: 10.3390/vaccines11101546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 09/14/2023] [Accepted: 09/27/2023] [Indexed: 10/29/2023] Open
Abstract
The vaccination campaign against SARS-CoV-2 relies on the world-wide availability of effective vaccines, with a potential need of 20 billion vaccine doses to fully vaccinate the world population. To reach this goal, the manufacturing and logistic processes should be affordable to all countries, irrespective of economical and climatic conditions. Outer membrane vesicles (OMVs) are bacterial-derived vesicles that can be engineered to incorporate heterologous antigens. Given the inherent adjuvanticity, such modified OMVs can be used as vaccines to induce potent immune responses against the associated proteins. Here, we show that OMVs engineered to incorporate peptides derived from the receptor binding motif (RBM) of the spike protein from SARS-CoV-2 elicit an effective immune response in vaccinated mice, resulting in the production of neutralizing antibodies (nAbs) with a titre higher than 1:300. The immunity induced by the vaccine is sufficient to protect the animals from intranasal challenge with SARS-CoV-2, preventing both virus replication in the lungs and the pathology associated with virus infection. Furthermore, we show that OMVs can be effectively decorated with the RBM of the Omicron BA.1 variant and that such engineered OMVs induce nAbs against Omicron BA.1 and BA.5, as measured using the pseudovirus neutralization infectivity assay. Importantly, we show that the RBM438-509 ancestral-OMVs elicited antibodies which efficiently neutralize in vitro both the homologous ancestral strain, the Omicron BA.1 and BA.5 variants with a neutralization titre ranging from 1:100 to 1:1500, suggesting its potential use as a vaccine targeting diverse SARS-CoV-2 variants. Altogether, given the convenience associated with the ease of engineering, production and distribution, our results demonstrate that OMV-based SARS-CoV-2 vaccines can be a crucial addition to the vaccines currently available.
Collapse
Affiliation(s)
- Alberto Grandi
- Toscana Life Sciences Foundation, Via Fiorentina 1, 53100 Siena, Italy; (A.G.); (A.G.); (M.B.); (G.D.L.); (E.C.)
- BiOMViS Srl, Via Fiorentina 1, 53100 Siena, Italy
| | - Michele Tomasi
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Trento, Italy; (M.T.); (C.B.); (T.V.); (S.A.); (I.Z.); (R.C.); (S.T.); (L.C.)
| | - Irfan Ullah
- Section of Infectious Diseases, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT 06520, USA; (I.U.); (W.M.); (P.D.U.)
| | - Cinzia Bertelli
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Trento, Italy; (M.T.); (C.B.); (T.V.); (S.A.); (I.Z.); (R.C.); (S.T.); (L.C.)
| | - Teresa Vanzo
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Trento, Italy; (M.T.); (C.B.); (T.V.); (S.A.); (I.Z.); (R.C.); (S.T.); (L.C.)
| | - Silvia Accordini
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Trento, Italy; (M.T.); (C.B.); (T.V.); (S.A.); (I.Z.); (R.C.); (S.T.); (L.C.)
| | - Assunta Gagliardi
- Toscana Life Sciences Foundation, Via Fiorentina 1, 53100 Siena, Italy; (A.G.); (A.G.); (M.B.); (G.D.L.); (E.C.)
| | - Ilaria Zanella
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Trento, Italy; (M.T.); (C.B.); (T.V.); (S.A.); (I.Z.); (R.C.); (S.T.); (L.C.)
| | - Mattia Benedet
- Toscana Life Sciences Foundation, Via Fiorentina 1, 53100 Siena, Italy; (A.G.); (A.G.); (M.B.); (G.D.L.); (E.C.)
| | - Riccardo Corbellari
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Trento, Italy; (M.T.); (C.B.); (T.V.); (S.A.); (I.Z.); (R.C.); (S.T.); (L.C.)
| | - Gabriele Di Lascio
- Toscana Life Sciences Foundation, Via Fiorentina 1, 53100 Siena, Italy; (A.G.); (A.G.); (M.B.); (G.D.L.); (E.C.)
| | - Silvia Tamburini
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Trento, Italy; (M.T.); (C.B.); (T.V.); (S.A.); (I.Z.); (R.C.); (S.T.); (L.C.)
| | - Elena Caproni
- Toscana Life Sciences Foundation, Via Fiorentina 1, 53100 Siena, Italy; (A.G.); (A.G.); (M.B.); (G.D.L.); (E.C.)
| | - Lorenzo Croia
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Trento, Italy; (M.T.); (C.B.); (T.V.); (S.A.); (I.Z.); (R.C.); (S.T.); (L.C.)
| | - Micol Ravà
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (M.R.); (V.F.); (P.D.L.); (D.M.); (E.S.); (M.I.)
| | - Valeria Fumagalli
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (M.R.); (V.F.); (P.D.L.); (D.M.); (E.S.); (M.I.)
- Vita-Salute San Raffaele University, Via Olgettina 58, 00132 Milan, Italy
| | - Pietro Di Lucia
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (M.R.); (V.F.); (P.D.L.); (D.M.); (E.S.); (M.I.)
| | - Davide Marotta
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (M.R.); (V.F.); (P.D.L.); (D.M.); (E.S.); (M.I.)
- Vita-Salute San Raffaele University, Via Olgettina 58, 00132 Milan, Italy
| | - Eleonora Sala
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (M.R.); (V.F.); (P.D.L.); (D.M.); (E.S.); (M.I.)
- Vita-Salute San Raffaele University, Via Olgettina 58, 00132 Milan, Italy
| | - Matteo Iannacone
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (M.R.); (V.F.); (P.D.L.); (D.M.); (E.S.); (M.I.)
- Vita-Salute San Raffaele University, Via Olgettina 58, 00132 Milan, Italy
- Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Priti Kumar
- Department of Microbial Pathogenesis, School of Medicine, Yale University, New Haven, CT 06510, USA;
| | - Walther Mothes
- Section of Infectious Diseases, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT 06520, USA; (I.U.); (W.M.); (P.D.U.)
- Department of Microbial Pathogenesis, School of Medicine, Yale University, New Haven, CT 06510, USA;
| | - Pradeep D. Uchil
- Section of Infectious Diseases, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT 06520, USA; (I.U.); (W.M.); (P.D.U.)
- Department of Microbial Pathogenesis, School of Medicine, Yale University, New Haven, CT 06510, USA;
| | - Peter Cherepanov
- Chromatin Structure and Mobile DNA Laboratory, The Francis Crick Institute, London NW1 1AT, UK;
| | - Martino Bolognesi
- Biosciences Department, University of Milan, Via Celoria 26, 20133 Milan, Italy;
| | - Massimo Pizzato
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Trento, Italy; (M.T.); (C.B.); (T.V.); (S.A.); (I.Z.); (R.C.); (S.T.); (L.C.)
| | - Guido Grandi
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Trento, Italy; (M.T.); (C.B.); (T.V.); (S.A.); (I.Z.); (R.C.); (S.T.); (L.C.)
| |
Collapse
|
21
|
Rossi O, Citiulo F, Giannelli C, Cappelletti E, Gasperini G, Mancini F, Acquaviva A, Raso MM, Sollai L, Alfini R, Aruta MG, Vitali CG, Pizza M, Necchi F, Rappuoli R, Martin LB, Berlanda Scorza F, Colucci AM, Micoli F. A next-generation GMMA-based vaccine candidate to fight shigellosis. NPJ Vaccines 2023; 8:130. [PMID: 37670042 PMCID: PMC10480147 DOI: 10.1038/s41541-023-00725-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 08/04/2023] [Indexed: 09/07/2023] Open
Abstract
Shigellosis is a leading cause of diarrheal disease in low-middle-income countries (LMICs). Effective vaccines will help to reduce the disease burden, exacerbated by increasing antibiotic resistance, in the most susceptible population represented by young children. A challenge for a broadly protective vaccine against shigellosis is to cover the most epidemiologically relevant serotypes among >50 Shigella serotypes circulating worldwide. The GMMA platform has been proposed as an innovative delivery system for Shigella O-antigens, and we have developed a 4-component vaccine against S. sonnei, S. flexneri 1b, 2a and 3a identified among the most prevalent Shigella serotypes in LMICs. Driven by the immunogenicity results obtained in clinic with a first-generation mono-component vaccine, a new S. sonnei GMMA construct was generated and combined with three S. flexneri GMMA in a 4-component Alhydrogel formulation (altSonflex1-2-3). This formulation was highly immunogenic, with no evidence of negative antigenic interference in mice and rabbits. The vaccine induced bactericidal antibodies also against heterologous Shigella strains carrying O-antigens different from those included in the vaccine. The Monocyte Activation Test used to evaluate the potential reactogenicity of the vaccine formulation revealed no differences compared to the S. sonnei mono-component vaccine, shown to be safe in several clinical trials in adults. A GLP toxicology study in rabbits confirmed that the vaccine was well tolerated. The preclinical study results support the clinical evaluation of altSonflex1-2-3 in healthy populations, and a phase 1-2 clinical trial is currently ongoing.
Collapse
Affiliation(s)
- Omar Rossi
- GSK Global Health Vaccines R&D (GVGH), Siena, Italy
| | | | | | | | - Gianmarco Gasperini
- GSK Global Health Vaccines R&D (GVGH), Siena, Italy
- GSK Vaccines Srl, Siena, Italy
| | | | | | | | - Luigi Sollai
- GSK Global Health Vaccines R&D (GVGH), Siena, Italy
| | - Renzo Alfini
- GSK Global Health Vaccines R&D (GVGH), Siena, Italy
| | | | | | - Mariagrazia Pizza
- GSK Global Health Vaccines R&D (GVGH), Siena, Italy
- GSK Vaccines Srl, Siena, Italy
- Imperial College, London, United Kingdom
| | | | - Rino Rappuoli
- GSK Vaccines Srl, Siena, Italy
- Fondazione Biotecnopolo, Siena, Italy
| | - Laura B Martin
- GSK Global Health Vaccines R&D (GVGH), Siena, Italy
- US Pharmacopoeia, Rockville, Maryland, USA
| | | | | | | |
Collapse
|
22
|
Khodak YA. Heterologous Expression of Recombinant Proteins and Their Derivatives Used as Carriers for Conjugate Vaccines. BIOCHEMISTRY. BIOKHIMIIA 2023; 88:1248-1266. [PMID: 37770392 DOI: 10.1134/s0006297923090055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/30/2023] [Accepted: 07/25/2023] [Indexed: 09/30/2023]
Abstract
Carrier proteins that provide an effective and long-term immune response to weak antigens has become a real breakthrough in the disease prevention, making it available to a wider range of patients and making it possible to obtain reliable vaccines against a variety of pathogens. Currently, research is continuing both to identify new peptides, proteins, and their complexes potentially suitable for use as carriers, and to develop new methods for isolation, purification, and conjugation of already known and well-established proteins. The use of recombinant proteins has a number of advantages over isolation from natural sources, such as simpler cultivation of the host organism, the possibility of modifying genetic constructs, use of numerous promoter variants, signal sequences, and other regulatory elements. This review is devoted to the methods of obtaining both traditional and new recombinant proteins and their derivatives already being used or potentially suitable for use as carrier proteins in conjugate vaccines.
Collapse
Affiliation(s)
- Yuliya A Khodak
- Institute of Bioengineering, Federal Research Centre of Biotechnology, Russian Academy of Sciences, Moscow, 117312, Russia.
| |
Collapse
|
23
|
Necchi F, Giannelli C, Acquaviva A, Alfini R, Monaci V, Arato V, Rossi O, Micoli F. From an in vivo to an in vitro relative potency (IVRP) assay to fully characterize a multicomponent O-antigen based vaccine against Shigella. Carbohydr Polym 2023; 314:120920. [PMID: 37173008 DOI: 10.1016/j.carbpol.2023.120920] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 03/16/2023] [Accepted: 04/12/2023] [Indexed: 05/15/2023]
Abstract
Outer membrane vesicles (OMV) represent an innovative platform for the design of polysaccharide based vaccines. Generalized Modules for Membrane Antigens (GMMA), OMV released from engineered Gram-negative bacteria, have been proposed for the delivery of the O-Antigen, key target for protective immunity against several pathogens including Shigella. altSonflex1-2-3 is a GMMA based vaccine, including S. sonnei and S. flexneri 1b, 2a and 3a O-Antigens, with the aim to elicit broad protection against the most prevalent Shigella serotypes, especially affecting children in low-middle income countries. Here we developed an In Vitro Relative Potency assay, based on recognition of O-Antigen by functional monoclonal antibodies selected to bind the key epitopes of the different O-Antigen active ingredients, directly applied to our Alhydrogel-formulated vaccine. Heat-stressed altSonflex1-2-3 formulations were generated and extensively characterized. The impact of detected biochemical changes in in vivo and in vitro potency assays was assessed. The overall results showed how the in vitro assay can replace the use of animals, overcoming the inherently high variability of in vivo potency studies. The entire panel of physico-chemical methods developed will contribute to detect suboptimal batches and will be valuable to perform stability studies. The work on Shigella vaccine candidate can be easily extended to other O-Antigen based vaccines.
Collapse
Affiliation(s)
- Francesca Necchi
- GSK Vaccines Institute for Global Health (GVGH), Via Fiorentina 1, 53100 Siena, Italy.
| | - Carlo Giannelli
- GSK Vaccines Institute for Global Health (GVGH), Via Fiorentina 1, 53100 Siena, Italy.
| | - Alessandra Acquaviva
- GSK Vaccines Institute for Global Health (GVGH), Via Fiorentina 1, 53100 Siena, Italy.
| | - Renzo Alfini
- GSK Vaccines Institute for Global Health (GVGH), Via Fiorentina 1, 53100 Siena, Italy.
| | - Valentina Monaci
- GSK Vaccines Institute for Global Health (GVGH), Via Fiorentina 1, 53100 Siena, Italy.
| | - Vanessa Arato
- GSK Vaccines Institute for Global Health (GVGH), Via Fiorentina 1, 53100 Siena, Italy.
| | - Omar Rossi
- GSK Vaccines Institute for Global Health (GVGH), Via Fiorentina 1, 53100 Siena, Italy.
| | - Francesca Micoli
- GSK Vaccines Institute for Global Health (GVGH), Via Fiorentina 1, 53100 Siena, Italy.
| |
Collapse
|
24
|
Akegbe H, Onyeaka H, Michael Mazi I, Alex Olowolafe O, Dolapo Omotosho A, Olatunji Oladunjoye I, Amuda Tajudeen Y, Seun Ofeh A. The need for Africa to develop capacity for vaccinology as a means of curbing antimicrobial resistance. Vaccine X 2023; 14:100320. [PMID: 37293248 PMCID: PMC10244683 DOI: 10.1016/j.jvacx.2023.100320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 03/08/2023] [Accepted: 05/26/2023] [Indexed: 06/10/2023] Open
Abstract
The high prevalence of infectious diseases in Africa, combined with weak healthcare systems, poor antimicrobial stewardship, and an unchecked drug supply chain, is steadily reversing the trend in the fight against infectious diseases in this part of the world, posing severe threats to antimicrobial resistance (AMR). AMR continuously evolves and threatens to undermine antimicrobial efficacy and undo advances against infectious diseases. This brewing pandemic is now recognized as a significant worldwide health danger, implicated in several cases of morbidity, mortality, and increasing healthcare costs. Vaccine technology has been proven to be the principal remedy to this imminent danger since it prevents microbial infections. However, since Africa cannot produce its vaccines, it relies on external sources and, as a result, it is significantly affected by vaccine nationalism, hoarding, and instabilities in global supply chains. This has further adversely impacted the ability of African governments to regulate rollouts, protect their citizens, and ultimately rejoin the global economy. This dependency is a severe challenge to Africa's health resilience, as it is unsustainable. Given the inevitability of potential global pandemics and the alarming incidences of multi-drug resistance infections reported daily, Africa must develop the capability to produce its vaccines. The review utilized a systematic search of academic databases and grey literature, as well as a manual search of relevant reports and articles. In this review, we outline the public health threats and concerns that AMR poses to Africans, and the hurdles and advances achieved in vaccine development over the years. We also highlight possible strategies, particularly collaborative efforts, that will accelerate vaccine production and ease the strain of infectious diseases and antimicrobial resistance in Africa. Key findings indicate that Africa has significant gaps in its vaccine manufacturing and distribution capacity, with only a few countries having the ability to produce vaccines. Additionally, existing vaccine production facilities are often outdated and require significant investment to meet international standards. The review also highlights successful initiatives in Africa, such as the mRNA vaccine hub and the African Vaccine Manufacturing Initiative, which have demonstrated the potential for building local vaccine manufacturing capacity. The study concludes that Africa needs to prioritize investment in vaccine research and development, regulatory capacity, and infrastructure to build a sustainable vaccine manufacturing ecosystem. Overall, this review emphasizes the urgent need for Africa to develop its vaccine manufacturing capacity to improve vaccine access and strengthen its ability to respond to future pandemics. The findings underscore the importance of collaboration between African governments, international organizations, and the private sector to build a resilient vaccine ecosystem in Africa.
Collapse
Affiliation(s)
- Hope Akegbe
- Department of Microbiology, Faculty of Life Sciences, University of Benin, Benin City, Nigeria
| | - Helen Onyeaka
- School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Ifeanyi Michael Mazi
- Department of Microbiology, Faculty of Life Sciences, University of Benin, Benin City, Nigeria
| | - Opeyemi Alex Olowolafe
- Department of Microbiology, Faculty of Life Sciences, University of Benin, Benin City, Nigeria
| | | | | | - Yusuf Amuda Tajudeen
- Department of Microbiology, Faculty of Life Sciences, University of Ilorin, Ilorin, Nigeria
| | - Augustine Seun Ofeh
- Department of Microbiology, Faculty of Science, Delta State University, Abraka, Nigeria
| |
Collapse
|
25
|
Thapa HB, Ebenberger SP, Schild S. The Two Faces of Bacterial Membrane Vesicles: Pathophysiological Roles and Therapeutic Opportunities. Antibiotics (Basel) 2023; 12:1045. [PMID: 37370364 PMCID: PMC10295235 DOI: 10.3390/antibiotics12061045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/07/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
Bacterial membrane vesicles (MVs) are nanosized lipid particles secreted by lysis or blebbing mechanisms from Gram-negative and -positive bacteria. It is becoming increasingly evident that MVs can promote antimicrobial resistance but also provide versatile opportunities for therapeutic exploitation. As non-living facsimiles of parent bacteria, MVs can carry multiple bioactive molecules such as proteins, lipids, nucleic acids, and metabolites, which enable them to participate in intra- and interspecific communication. Although energetically costly, the release of MVs seems beneficial for bacterial fitness, especially for pathogens. In this review, we briefly discuss the current understanding of diverse MV biogenesis routes affecting MV cargo. We comprehensively highlight the physiological functions of MVs derived from human pathogens covering in vivo adaptation, colonization fitness, and effector delivery. Emphasis is given to recent findings suggesting a vicious cycle of MV biogenesis, pathophysiological function, and antibiotic therapy. We also summarize potential therapeutical applications, such as immunotherapy, vaccination, targeted delivery, and antimicrobial potency, including their experimental validation. This comparative overview identifies common and unique strategies for MV modification used along diverse applications. Thus, the review summarizes timely aspects of MV biology in a so far unprecedented combination ranging from beneficial function for bacterial pathogen survival to future medical applications.
Collapse
Affiliation(s)
- Himadri B. Thapa
- Institute of Molecular Biosciences, University of Graz, Humboldtstrasse 50, 8010 Graz, Austria
| | - Stephan P. Ebenberger
- Institute of Molecular Biosciences, University of Graz, Humboldtstrasse 50, 8010 Graz, Austria
| | - Stefan Schild
- Institute of Molecular Biosciences, University of Graz, Humboldtstrasse 50, 8010 Graz, Austria
- BioTechMed Graz, 8010 Graz, Austria
- Field of Excellence Biohealth, University of Graz, 8010 Graz, Austria
| |
Collapse
|
26
|
Mba IE, Sharndama HC, Anyaegbunam ZKG, Anekpo CC, Amadi BC, Morumda D, Doowuese Y, Ihezuo UJ, Chukwukelu JU, Okeke OP. Vaccine development for bacterial pathogens: Advances, challenges and prospects. Trop Med Int Health 2023; 28:275-299. [PMID: 36861882 DOI: 10.1111/tmi.13865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
The advent and use of antimicrobials have played a key role in treating potentially life-threatening infectious diseases, improving health, and saving the lives of millions of people worldwide. However, the emergence of multidrug resistant (MDR) pathogens has been a significant health challenge that has compromised the ability to prevent and treat a wide range of infectious diseases that were once treatable. Vaccines offer potential as a promising alternative to fight against antimicrobial resistance (AMR) infectious diseases. Vaccine technologies include reverse vaccinology, structural biology methods, nucleic acid (DNA and mRNA) vaccines, generalised modules for membrane antigens, bioconjugates/glycoconjugates, nanomaterials and several other emerging technological advances that are offering a potential breakthrough in the development of efficient vaccines against pathogens. This review covers the opportunities and advancements in vaccine discovery and development targeting bacterial pathogens. We reflect on the impact of the already-developed vaccines targeting bacterial pathogens and the potential of those currently under different stages of preclinical and clinical trials. More importantly, we critically and comprehensively analyse the challenges while highlighting the key indices for future vaccine prospects. Finally, the issues and concerns of AMR for low-income countries (sub-Saharan Africa) and the challenges with vaccine integration, discovery and development in this region are critically evaluated.
Collapse
Affiliation(s)
- Ifeanyi Elibe Mba
- Department of Microbiology, University of Nigeria, Nsukka, Nigeria
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, University of Ibadan, Ibadan, Nigeria
| | | | - Zikora Kizito Glory Anyaegbunam
- Department of Microbiology, University of Nigeria, Nsukka, Nigeria
- Institute for Drug-Herbal Medicine-Excipient Research and Development, University of Nigeria, Nsukka, Nigeria
| | - Chijioke Chinedu Anekpo
- Department of Ear Nose and Throat, College of Medicine, Enugu State University of Science and Technology, Enugu, Nigeria
| | - Ben Chibuzo Amadi
- Pharmaceutical Technology and Industrial Pharmacy, University of Nigeria, Nsukka, Nigeria
| | - Daji Morumda
- Department of Microbiology, Federal University Wukari, Wukari, Taraba, Nigeria
| | - Yandev Doowuese
- Department of Microbiology, Federal University of Health Sciences, Otukpo, Nigeria
| | - Uchechi Justina Ihezuo
- Department of Microbiology, University of Nigeria, Nsukka, Nigeria
- Institute for Drug-Herbal Medicine-Excipient Research and Development, University of Nigeria, Nsukka, Nigeria
| | | | | |
Collapse
|
27
|
Toward a Shigella Vaccine: Opportunities and Challenges to Fight an Antimicrobial-Resistant Pathogen. Int J Mol Sci 2023; 24:ijms24054649. [PMID: 36902092 PMCID: PMC10003550 DOI: 10.3390/ijms24054649] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/23/2023] [Accepted: 02/24/2023] [Indexed: 03/04/2023] Open
Abstract
Shigellosis causes more than 200,000 deaths worldwide and most of this burden falls on Low- and Middle-Income Countries (LMICs), with a particular incidence in children under 5 years of age. In the last decades, Shigella has become even more worrisome because of the onset of antimicrobial-resistant strains (AMR). Indeed, the WHO has listed Shigella as one of the priority pathogens for the development of new interventions. To date, there are no broadly available vaccines against shigellosis, but several candidates are being evaluated in preclinical and clinical studies, bringing to light very important data and information. With the aim to facilitate the understanding of the state-of-the-art of Shigella vaccine development, here we report what is known about Shigella epidemiology and pathogenesis with a focus on virulence factors and potential antigens for vaccine development. We discuss immunity after natural infection and immunization. In addition, we highlight the main characteristics of the different technologies that have been applied for the development of a vaccine with broad protection against Shigella.
Collapse
|
28
|
A Pentavalent Shigella flexneri LPS-Based Vaccine Candidate Is Safe and Immunogenic in Animal Models. Vaccines (Basel) 2023; 11:vaccines11020345. [PMID: 36851223 PMCID: PMC9966156 DOI: 10.3390/vaccines11020345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/19/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
A multivalent vaccine is much needed to achieve protection against predominant Shigella serotypes. Recently, we demonstrated the clinical applicability and immunogenic potential of tri-acylated S. flexneri 2a lipopolysaccharide (Ac3-S-LPS). Using a similar approach, we designed a pentavalent LPS candidate vaccine against S. flexneri 1b, 2a, 3a, 6, and Y (PLVF). In this study, we performed molecular and antigenic characterization of the vaccine candidate and its preclinical evaluation. There were no signs of acute toxicity after subcutaneous administration of PLVF in rabbits at a proposed human dose of 125 μg. No pyrogenic reactions and adverse effects associated with chronic toxicity after repeated administration of PLVF were revealed either. The immunization of mice with PLVF led to ≥16-fold increase in S. flexneri 1b-, 2a-, 3a-, 6-, and Y-specific antibodies. In a serum bactericidal antibody (SBA) assay, we registered 54%, 66%, 35%, 60%, and 60% killing of S. flexneri 1b, 2a, 3a, 6, and Y, respectively. In the guinea pig keratoconjunctivitis model, the efficacy was 50% to 75% against challenge with all five S. flexneri serotypes. These studies demonstrate that PLVF is safe, immunogenic over a wide range of doses, and provides protection against challenge with homologous S. flexneri strains, thus confirming the validity of pentavalent design of the combined vaccine.
Collapse
|
29
|
Mancini F, Alfini R, Caradonna V, Monaci V, Carducci M, Gasperini G, Piccioli D, Biagini M, Giannelli C, Rossi O, Pizza M, Micoli F. Exploring the Role of GMMA Components in the Immunogenicity of a 4-Valent Vaccine against Shigella. Int J Mol Sci 2023; 24:2742. [PMID: 36769063 PMCID: PMC9916818 DOI: 10.3390/ijms24032742] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/24/2023] [Accepted: 01/26/2023] [Indexed: 02/04/2023] Open
Abstract
Shigellosis is the leading cause of diarrheal disease, especially in children of low- and middle-income countries, and is often associated with anti-microbial resistance. Currently, there are no licensed vaccines widely available against Shigella, but several candidates based on the O-antigen (OAg) portion of lipopolysaccharides are in development. We have proposed Generalized Modules for Membrane Antigens (GMMA) as an innovative delivery system for OAg, and a quadrivalent vaccine candidate containing GMMA from S. sonnei and three prevalent S. flexneri serotypes (1b, 2a and 3a) is moving to a phase II clinical trial, with the aim to elicit broad protection against Shigella. GMMA are able to induce anti-OAg-specific functional IgG responses in animal models and healthy adults. We have previously demonstrated that antibodies against protein antigens are also generated upon immunization with S. sonnei GMMA. In this work, we show that a quadrivalent Shigella GMMA-based vaccine is able to promote a humoral response against OAg and proteins of all GMMA types contained in the investigational vaccine. Proteins contained in GMMA provide T cell help as GMMA elicit a stronger anti-OAg IgG response in wild type than in T cell-deficient mice. Additionally, we observed that only the trigger of Toll-like Receptor (TLR) 4 and not of TLR2 contributed to GMMA immunogenicity. In conclusion, when tested in mice, GMMA of a quadrivalent Shigella vaccine candidate combine both adjuvant and carrier activities which allow an increase in the low immunogenic properties of carbohydrate antigens.
Collapse
Affiliation(s)
- Francesca Mancini
- GSK Vaccines Institute for Global Health (GVGH), via Fiorentina 1, 53100 Siena, Italy
| | - Renzo Alfini
- GSK Vaccines Institute for Global Health (GVGH), via Fiorentina 1, 53100 Siena, Italy
| | - Valentina Caradonna
- GSK Vaccines Institute for Global Health (GVGH), via Fiorentina 1, 53100 Siena, Italy
| | - Valentina Monaci
- GSK Vaccines Institute for Global Health (GVGH), via Fiorentina 1, 53100 Siena, Italy
| | - Martina Carducci
- GSK Vaccines Institute for Global Health (GVGH), via Fiorentina 1, 53100 Siena, Italy
| | - Gianmarco Gasperini
- GSK Vaccines Institute for Global Health (GVGH), via Fiorentina 1, 53100 Siena, Italy
| | | | | | - Carlo Giannelli
- GSK Vaccines Institute for Global Health (GVGH), via Fiorentina 1, 53100 Siena, Italy
| | - Omar Rossi
- GSK Vaccines Institute for Global Health (GVGH), via Fiorentina 1, 53100 Siena, Italy
| | - Mariagrazia Pizza
- GSK Vaccines Institute for Global Health (GVGH), via Fiorentina 1, 53100 Siena, Italy
| | - Francesca Micoli
- GSK Vaccines Institute for Global Health (GVGH), via Fiorentina 1, 53100 Siena, Italy
| |
Collapse
|
30
|
Ge Y, Sun F, Zhao B, Kong F, Li Z, Kong X. Bacteria derived extracellular vesicles in the pathogenesis and treatment of gastrointestinal tumours. Front Oncol 2023; 12:1103446. [PMID: 36776356 PMCID: PMC9910087 DOI: 10.3389/fonc.2022.1103446] [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: 11/20/2022] [Accepted: 12/28/2022] [Indexed: 01/27/2023] Open
Abstract
Extracellular vesicles are fundamentally significant in the communication between cells. Outer Membrane Vesicles(OMVs) are a special kind of EVs produced by Gram-negative bacteria, which are minute exosome-like particles budding from the outer membrane, which have been found to play essential roles in diverse bacterial life events, including regulation of microbial interactions, pathogenesis promotion, stress responses and biofilm formation. Recently, and more researches have explored the substantial potentials of EVs as natural functional nanoparticles in the bioengineering applications in infectious diseases, cardiovascular diseases, autoimmune diseases and neurological diseases, such as antibacterial therapy, cancer drugs and immunoadjuvants, with several candidates in clinical trials showing promising efficacy. However, due to the poor understanding of sources, membrane structures and biogenesis mechanisms of EVs, progress in clinical applications still remains timid. In this review, we summarize the latest findings of EVs, especially in gastrointestinal tract tumours, to provide a comprehensive introduction of EVs in tumorigenesis and therapeutics.
Collapse
Affiliation(s)
- Yang Ge
- Changhai Clinical Research Unit, Changhai Hospital, Naval Military Medical University, Shanghai, China,Department of Gastroenterology, Changhai Hospital, Naval Military Medical University, Shanghai, China
| | - Fengyuan Sun
- Changhai Clinical Research Unit, Changhai Hospital, Naval Military Medical University, Shanghai, China
| | - Bo Zhao
- Changhai Clinical Research Unit, Changhai Hospital, Naval Military Medical University, Shanghai, China
| | - Fanyang Kong
- Changhai Clinical Research Unit, Changhai Hospital, Naval Military Medical University, Shanghai, China,Department of Gastroenterology, Changhai Hospital, Naval Military Medical University, Shanghai, China,*Correspondence: Xiangyu Kong, ; Zhaoshen Li, ; Fanyang Kong,
| | - Zhaoshen Li
- Changhai Clinical Research Unit, Changhai Hospital, Naval Military Medical University, Shanghai, China,*Correspondence: Xiangyu Kong, ; Zhaoshen Li, ; Fanyang Kong,
| | - Xiangyu Kong
- Changhai Clinical Research Unit, Changhai Hospital, Naval Military Medical University, Shanghai, China,Department of Gastroenterology, Changhai Hospital, Naval Military Medical University, Shanghai, China,National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical University, Shanghai, China,*Correspondence: Xiangyu Kong, ; Zhaoshen Li, ; Fanyang Kong,
| |
Collapse
|
31
|
Bhaumik U, Halder P, Howlader DR, Banerjee S, Maiti S, Dutta S, Koley H. A tetravalent Shigella Outer Membrane Vesicles based candidate vaccine offered cross-protection against all the serogroups of Shigella in adult mice. Microbes Infect 2023; 25:105100. [PMID: 36696935 DOI: 10.1016/j.micinf.2023.105100] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 12/26/2022] [Accepted: 01/06/2023] [Indexed: 01/23/2023]
Abstract
In today's world and mostly in low and middle income countries, S. flexneri and S. sonnei remains the major causative agent of clinical bacillary dysentery. Based on contemporary epidemiology, a tetravalent Outer Membrane Vesicle (OMVs) based immunogen was formulated using the most commonly circulating Shigella strains, namely, S. flexneri 2a, S. flexneri 3a, S. flexneri 6 and S. sonnei I, in a 1:1:1:1 ratio. Adult BALB/c mice were orally immunized in a prime-boost-boost manner. Tetravalent Shigella OMVs immunogen induced significant and persistent serum and mucosal antibodies against OMVs, Outer Membrane Proteins (OMPs) and lipopolysaccharides (LPS). Tetravalent OMVs also primed cell mediated immune response effectively. Protective efficacy against six heterologous Shigella strains was checked in an intra-peritoneal mouse model. Immunized mice survived lethal infection better than the non-immunized mice cohort with fewer replicating bacteria isolated from their gut. This study establishes the possibilities of tetravalent OMVs immunogen to become a potent vaccine candidate against human shigellosis, overcoming the limitations of sero-specific cross-protection of Shigella species.
Collapse
Affiliation(s)
- Ushasi Bhaumik
- Division of Bacteriology, ICMR- National Institute of Cholera and Enteric Diseases. P-33, C.I.T. Road, Scheme XM, Beliaghata, Kolkata-700 010, India; Center for Vaccine Development and Global Health, School of Medicine, University of Maryland, Baltimore, MD 2120, United States
| | - Prolay Halder
- Division of Bacteriology, ICMR- National Institute of Cholera and Enteric Diseases. P-33, C.I.T. Road, Scheme XM, Beliaghata, Kolkata-700 010, India
| | - Debaki Ranjan Howlader
- Division of Bacteriology, ICMR- National Institute of Cholera and Enteric Diseases. P-33, C.I.T. Road, Scheme XM, Beliaghata, Kolkata-700 010, India; Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65201, United States
| | - Soumalya Banerjee
- Division of Bacteriology, ICMR- National Institute of Cholera and Enteric Diseases. P-33, C.I.T. Road, Scheme XM, Beliaghata, Kolkata-700 010, India
| | - Suhrid Maiti
- Division of Bacteriology, ICMR- National Institute of Cholera and Enteric Diseases. P-33, C.I.T. Road, Scheme XM, Beliaghata, Kolkata-700 010, India; Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65201, United States
| | - Shanta Dutta
- ICMR- National Institute of Cholera and Enteric Diseases. . P-33, C.I.T. Road, Scheme XM, Beliaghata, Kolkata 700 010, India
| | - Hemanta Koley
- Division of Bacteriology, ICMR- National Institute of Cholera and Enteric Diseases. P-33, C.I.T. Road, Scheme XM, Beliaghata, Kolkata-700 010, India. http://www.niced.org.in/
| |
Collapse
|
32
|
Aruta MG, De Simone D, Dale H, Chirwa E, Kadwala I, Mbewe M, Banda H, Gordon M, Pizza M, Berlanda Scorza F, Nyirenda T, Canals R, Rossi O. Development and Characterization of a Luminescence-Based High-Throughput Serum Bactericidal Assay (L-SBA) to Assess Bactericidal Activity of Human Sera against Nontyphoidal Salmonella. Methods Protoc 2022; 5:100. [PMID: 36548142 PMCID: PMC9783057 DOI: 10.3390/mps5060100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/30/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022] Open
Abstract
Salmonella Typhimurium and Salmonella Enteritidis are leading causative agents of invasive nontyphoidal Salmonella (iNTS) disease, which represents one of the major causes of death and morbidity in sub-Saharan Africa, still partially underestimated. Large sero-epidemiological studies are necessary to unravel the burden of disease and guide the introduction of vaccines that are not yet available. Even if no correlate of protection has been determined so far for iNTS, the evaluation of complement-mediated functionality of antibodies generated towards natural infection or elicited upon vaccination may represent a big step towards this achievement. Here we present the setup and the intra-laboratory characterization in terms of repeatability, intermediate precision, linearity, and specificity of a high-throughput luminescence-based serum bactericidal assay (L-SBA). This method could be useful to perform sero-epidemiological studies across iNTS endemic countries and for evaluation of antibodies raised against iNTS vaccine candidates in upcoming clinical trials.
Collapse
Affiliation(s)
- Maria Grazia Aruta
- GSK Vaccines Institute for Global Health Srl, Via Fiorentina 1, 53100 Siena, Italy
| | - Daniele De Simone
- GSK Vaccines Institute for Global Health Srl, Via Fiorentina 1, 53100 Siena, Italy
| | - Helen Dale
- Malawi Liverpool Wellcome Trust Programme, Queen Elizabeth Central Hospital, College of Medicine, Blantyre 30096, Malawi
- Pathology Department, Kamuzu University of Health Sciences, Blantyre 312225, Malawi
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 3BX, UK
| | - Esmelda Chirwa
- Malawi Liverpool Wellcome Trust Programme, Queen Elizabeth Central Hospital, College of Medicine, Blantyre 30096, Malawi
- Pathology Department, Kamuzu University of Health Sciences, Blantyre 312225, Malawi
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 3BX, UK
| | - Innocent Kadwala
- Malawi Liverpool Wellcome Trust Programme, Queen Elizabeth Central Hospital, College of Medicine, Blantyre 30096, Malawi
- Pathology Department, Kamuzu University of Health Sciences, Blantyre 312225, Malawi
| | - Maurice Mbewe
- Malawi Liverpool Wellcome Trust Programme, Queen Elizabeth Central Hospital, College of Medicine, Blantyre 30096, Malawi
- Pathology Department, Kamuzu University of Health Sciences, Blantyre 312225, Malawi
| | - Happy Banda
- Malawi Liverpool Wellcome Trust Programme, Queen Elizabeth Central Hospital, College of Medicine, Blantyre 30096, Malawi
- Pathology Department, Kamuzu University of Health Sciences, Blantyre 312225, Malawi
| | - Melita Gordon
- Malawi Liverpool Wellcome Trust Programme, Queen Elizabeth Central Hospital, College of Medicine, Blantyre 30096, Malawi
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 3BX, UK
| | - Mariagrazia Pizza
- GSK Vaccines Institute for Global Health Srl, Via Fiorentina 1, 53100 Siena, Italy
| | | | - Tonney Nyirenda
- Pathology Department, Kamuzu University of Health Sciences, Blantyre 312225, Malawi
| | - Rocío Canals
- GSK Vaccines Institute for Global Health Srl, Via Fiorentina 1, 53100 Siena, Italy
| | - Omar Rossi
- GSK Vaccines Institute for Global Health Srl, Via Fiorentina 1, 53100 Siena, Italy
| | | |
Collapse
|
33
|
Rossi O, Mastroeni P. Editorial: Pattern recognition receptors at the crosstalk between innate and adaptive immune systems and implications for vaccine development. Front Cell Infect Microbiol 2022; 12:1088029. [PMID: 36530438 PMCID: PMC9755836 DOI: 10.3389/fcimb.2022.1088029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 11/23/2022] [Indexed: 12/03/2022] Open
Affiliation(s)
- Omar Rossi
- GSK Vaccines Institute for Global Health, Siena, Italy,*Correspondence: Omar Rossi,
| | | |
Collapse
|
34
|
Hu K, Palmieri E, Samnuan K, Ricchetti B, Oldrini D, McKay PF, Wu G, Thorne L, Fooks AR, McElhinney LM, Goharriz H, Golding M, Shattock RJ, Micoli F. Generalized Modules for Membrane Antigens (GMMA), an outer membrane vesicle-based vaccine platform, for efficient viral antigen delivery. J Extracell Vesicles 2022; 11:e12247. [PMID: 36377074 PMCID: PMC9663859 DOI: 10.1002/jev2.12247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 04/19/2022] [Accepted: 04/30/2022] [Indexed: 11/17/2022] Open
Abstract
Vaccine platforms enable fast development, testing, and manufacture of more affordable vaccines. Here, we evaluated Generalized Modules for Membrane Antigens (GMMA), outer membrane vesicles (OMVs) generated by genetically modified Gram-negative bacteria, as a vaccine platform for viral pathogens. Influenza A virus hemagglutinin (HA), either physically mixed with GMMA (HA+STmGMMA mix), or covalently linked to GMMA surface (HA-STmGMMA conjugate), significantly increased antigen-specific humoral and cellular responses, with HA-STmGMMA conjugate inducing further enhancement than HA+STmGMMA mix. HA-STmGMMA conjugate protected mice from lethal challenge. The versatility for this platform was confirmed by conjugation of rabies glycoprotein (RABVG) onto GMMA through the same method. RABVG+STmGMMA mix and RABVG-STmGMMA conjugate exhibited similar humoral and cellular response patterns and protection efficacy as the HA formulations, indicating relatively consistent responses for different vaccines based on the GMMA platform. Comparing to soluble protein, GMMA was more efficiently taken up in vivo and exhibited a B-cell preferential uptake in the draining lymph nodes (LNs). Together, GMMA enhances immunity against viral antigens, and the platform works well with different antigens while retaining similar immunomodulatory patterns. The findings of our study imply the great potential of GMMA-based vaccine platform also against viral infectious diseases.
Collapse
Affiliation(s)
- Kai Hu
- Department of Infectious Diseases, Imperial College London, London, UK
| | - Elena Palmieri
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., Siena, Italy
| | - Karnyart Samnuan
- Department of Infectious Diseases, Imperial College London, London, UK
| | | | - Davide Oldrini
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., Siena, Italy
| | - Paul F McKay
- Department of Infectious Diseases, Imperial College London, London, UK
| | - Guanghui Wu
- Animal and Plant Health Agency (APHA), OIE Rabies Reference Laboratory, New Haw, Addlestone, Surrey, UK
| | - Leigh Thorne
- Animal and Plant Health Agency (APHA), OIE Rabies Reference Laboratory, New Haw, Addlestone, Surrey, UK
| | - Anthony R Fooks
- Animal and Plant Health Agency (APHA), OIE Rabies Reference Laboratory, New Haw, Addlestone, Surrey, UK
| | - Lorraine M McElhinney
- Animal and Plant Health Agency (APHA), OIE Rabies Reference Laboratory, New Haw, Addlestone, Surrey, UK
| | - Hooman Goharriz
- Animal and Plant Health Agency (APHA), OIE Rabies Reference Laboratory, New Haw, Addlestone, Surrey, UK
| | - Megan Golding
- Animal and Plant Health Agency (APHA), OIE Rabies Reference Laboratory, New Haw, Addlestone, Surrey, UK
| | - Robin J Shattock
- Department of Infectious Diseases, Imperial College London, London, UK
| | - Francesca Micoli
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., Siena, Italy
| |
Collapse
|
35
|
Piccioli D, Alfini R, Monaci V, Arato V, Carducci M, Aruta MG, Rossi O, Necchi F, Anemona A, Bartolini E, Micoli F. Antigen presentation by Follicular Dendritic cells to cognate B cells is pivotal for Generalised Modules for Membrane Antigens (GMMA) immunogenicity. Vaccine 2022; 40:6305-6314. [PMID: 36137901 DOI: 10.1016/j.vaccine.2022.09.034] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 08/26/2022] [Accepted: 09/09/2022] [Indexed: 01/27/2023]
Abstract
GMMA has been proposed as a potent technology platform for the design of safe, effective and affordable vaccines. As GMMA are vesicles blebbing out of the outer membrane of Gram-negative bacteria, they contain lipopolysaccharides, lipoproteins and peptidoglycans that stimulate immune cells via Toll-like Receptors 4 (TLR4) or TLR2. Being basically nanoparticles, GMMA can be efficiently captured by Follicular Dendritic Cells (FDC) for antigen presentation to cognate B cells. GMMA have shown to be highly immunogenic in preclinical and clinical studies and the engagement of TLR4 and TLR2 or antigen presentation by FDC may have a prominent role in GMMA immunogenicity, which is well worth investigating. By using GMMA derived from Shigella sonnei and Salmonella Typhimurium, we show for the first time that the antigen presentation by FDC to cognate B cells plays a major role in the induction of an effective humoral immune response upon immunization with GMMA by using both models. The engagement of TLR4 is critical to elicit an optimal antibody production, but its effect on antibody functionality is dependent on GMMA type and is dispensable when immunizing with Alum adjuvant, whereas TLR2 does not have any role for GMMA immunogenicity. Our findings represent a substantial advancement of the knowledge on GMMA mode of action and shed a light on novel perspectives for the design of safer and more effective GMMA-based vaccines. ONE SENTENCE SUMMARY: The study demonstrated that the antigen presentation by FDC to cognate B cells plays a major role for GMMA immunogenicity.
Collapse
Affiliation(s)
| | - Renzo Alfini
- GSK Vaccine Institute for Global Health (GVGH), Siena, Italy
| | | | - Vanessa Arato
- GSK Vaccine Institute for Global Health (GVGH), Siena, Italy
| | | | | | - Omar Rossi
- GSK Vaccine Institute for Global Health (GVGH), Siena, Italy
| | | | | | | | | |
Collapse
|
36
|
Kapulu MC, Nakakana U, Sciré AS, Sarakinou E, Conti V, Rossi O, Acquaviva A, Necchi F, Obiero CW, Martin LB, Bejon P, Njuguna P, Micoli F, Podda A. Complement-mediated serum bactericidal activity of antibodies elicited by the Shigella sonnei GMMA vaccine in adults from a shigellosis-endemic country: Exploratory analysis of a Phase 2a randomized study. Front Immunol 2022; 13:971866. [PMID: 36203568 PMCID: PMC9531247 DOI: 10.3389/fimmu.2022.971866] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 08/22/2022] [Indexed: 11/24/2022] Open
Abstract
Shigella is associated with a significant burden of disease worldwide among individuals of all ages and is the major cause of moderate and severe diarrhea in children under five years of age in low- and middle-income countries. Several candidate vaccines against Shigella species are currently under clinical development. The investigational 1790GAHB vaccine against Shigella sonnei is based on GMMA (Generalized Modules for Membrane Antigens) technology. The vaccine was well tolerated and induced high antibody levels in early-phase clinical trials in both Shigella-endemic and non-endemic settings. The present analysis assessed the bactericidal activity of antibodies induced by 1790GAHB in healthy Kenyan adults during a phase 2a, controlled, randomized study (NCT02676895). Participants received two doses of 1790GAHB 4 weeks apart containing either 1.5/25 µg or 6/100 µg O antigen/protein, or active comparator vaccines (Control). Serum bactericidal activity (SBA) against S. sonnei was assessed at pre-vaccination (D1), 28 days post-first dose (D29) and 28 days post-second dose (D57), using a luminescence-based assay. Most participants had SBA titers above the lower limit of quantification of the assay at D1. SBA geometric mean titers increased 3.4-fold in the 1.5/25 µg group and 6.3-fold in the 6/100 µg group by D29 and were maintained at D57. There was no increase in SBA geometric mean titers in the Control group. A strong correlation was observed between SBA titers and anti-S. sonnei lipopolysaccharide serum immunoglobulin G antibody concentrations (Pearson correlation coefficient = 0.918), indicating that SBA can effectively complement enzyme-linked immunosorbent assay data by indicating the functionality of 1790GAHB-induced antibodies.
Collapse
Affiliation(s)
- Melissa C. Kapulu
- Kenya Medical Research Institute (KEMRI)-Wellcome Trust Programme, Kilifi, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Usman Nakakana
- GSK Vaccines Institute for Global Health, Siena, Italy
- *Correspondence: Usman Nakakana,
| | | | | | | | - Omar Rossi
- GSK Vaccines Institute for Global Health, Siena, Italy
| | | | | | | | | | - Philip Bejon
- Kenya Medical Research Institute (KEMRI)-Wellcome Trust Programme, Kilifi, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Patricia Njuguna
- Clinical Research Department, KEMRI-Wellcome Trust Programme, Kilifi, Kenya
| | | | - Audino Podda
- GSK Vaccines Institute for Global Health, Siena, Italy
| |
Collapse
|
37
|
Mancini F, Micoli F, Rossi O. Setup and Characterization of a High-Throughput Luminescence-Based Serum Bactericidal Assay (L-SBA) to Determine Functionality of Human Sera against Shigella flexneri. BIOTECH 2022; 11:biotech11030029. [PMID: 35997337 PMCID: PMC9396978 DOI: 10.3390/biotech11030029] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/19/2022] [Accepted: 07/25/2022] [Indexed: 12/02/2022] Open
Abstract
Shigellosis represents a major public health problem worldwide. The morbidity of the disease, especially in children in developing countries, together with the increase of antimicrobial resistance make a vaccine against Shigella an urgent medical need. Several vaccines under development are targeting Shigella lipopolysaccharide (LPS), whose extreme diversity renders necessary the development of multivalent vaccines. Immunity against Shigella LPS can elicit antibodies capable of killing bacteria in a serotype-specific manner. Therefore, although a correlation of protection against shigellosis has not been established, demonstration of vaccine-elicited antibody bactericidal activity may provide one means of vaccine protection against Shigella. To facilitate Shigella vaccine development, we have set up a high-throughput serum bactericidal assay based on luminescence readout (L-SBA), which has been already used to determine the functionality of antibodies against S. sonnei in multiple clinical trials. Here we present the setup and intra-laboratory characterization of L-SBA against three epidemiologically relevant Shigella flexneri serotypes using human sera. We assessed the linearity, repeatability and reproducibility of the method, demonstrating high assay specificity to detect the activity of antibodies against each homologous strain without any heterologous aspecificity against species-related and non-species-related strains; this assay is ready to be used to determine bactericidal activity of clinical sera raised by multivalent vaccines and in sero-epidemiological studies.
Collapse
|
38
|
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
|
39
|
Long Q, Zheng P, Zheng X, Li W, Hua L, Yang Z, Huang W, Ma Y. Engineered bacterial membrane vesicles are promising carriers for vaccine design and tumor immunotherapy. Adv Drug Deliv Rev 2022; 186:114321. [PMID: 35533789 DOI: 10.1016/j.addr.2022.114321] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 04/18/2022] [Accepted: 04/30/2022] [Indexed: 02/06/2023]
Abstract
Bacterial membrane vesicles (BMVs) have emerged as novel and promising platforms for the development of vaccines and immunotherapeutic strategies against infectious and noninfectious diseases. The rich microbe-associated molecular patterns (MAMPs) and nanoscale membrane vesicle structure of BMVs make them highly immunogenic. In addition, BMVs can be endowed with more functions via genetic and chemical modifications. This article reviews the immunological characteristics and effects of BMVs, techniques for BMV production and modification, and the applications of BMVs as vaccines or vaccine carriers. In summary, given their versatile characteristics and immunomodulatory properties, BMVs can be used for clinical vaccine or immunotherapy applications.
Collapse
|
40
|
Gasperini G, Raso MM, Schiavo F, Aruta MG, Ravenscroft N, Bellich B, Cescutti P, Necchi F, Rappuoli R, Micoli F. Rapid generation of Shigella flexneri GMMA displaying natural or new and cross-reactive O-Antigens. NPJ Vaccines 2022; 7:69. [PMID: 35773292 PMCID: PMC9243986 DOI: 10.1038/s41541-022-00497-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 05/16/2022] [Indexed: 11/09/2022] Open
Abstract
Generalized modules for membrane antigens (GMMA) are exosomes released from engineered Gram-negative bacteria and represent an attractive vaccine platform for the delivery of the O-Antigen (OAg), recognized as the key target for protective immunity against several pathogens such as Shigella. Shigella is a major cause of disease in Low- and Middle-Income countries and the development of a vaccine needs to deal with its large serotypic diversity. All S. flexneri serotypes, except serotype 6, share a conserved OAg backbone, corresponding to serotype Y. Here, a GMMA-producing S. flexneri scaffold strain displaying the OAg backbone was engineered with different OAg-modifying enzymes, either individually or in combinations. This strategy rapidly yielded GMMA displaying 12 natural serotypes and 16 novel serotypes expressing multiple epitopes combinations that do not occur in nature. Importantly, a candidate GMMA displaying a hybrid OAg elicited broadly cross-bactericidal antibodies against a large panel of S. flexneri serotypes.
Collapse
Affiliation(s)
| | - Maria Michelina Raso
- GSK Vaccines Institute for Global Health (GVGH), Siena, Italy.,Università di Trieste, Trieste, Italy
| | - Fabiola Schiavo
- GSK Vaccines Institute for Global Health (GVGH), Siena, Italy
| | | | | | | | | | | | | | | |
Collapse
|
41
|
The tremendous biomedical potential of bacterial extracellular vesicles. Trends Biotechnol 2022; 40:1173-1194. [DOI: 10.1016/j.tibtech.2022.03.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 03/09/2022] [Accepted: 03/15/2022] [Indexed: 12/20/2022]
|
42
|
Lahiri D, Nag M, Dey A, Sarkar T, Pati S, Ray RR. Nanoparticles Based Antibacterial Vaccines: Novel Strategy to Combat Antimicrobial Resistance. Process Biochem 2022. [DOI: 10.1016/j.procbio.2022.05.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
43
|
Micoli F, Nakakana UN, Berlanda Scorza F. Towards a Four-Component GMMA-Based Vaccine against Shigella. Vaccines (Basel) 2022; 10:328. [PMID: 35214786 PMCID: PMC8880054 DOI: 10.3390/vaccines10020328] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/05/2022] [Accepted: 02/16/2022] [Indexed: 02/04/2023] Open
Abstract
Shigellosis remains a major public health problem around the world; it is one of the leading causes of diarrhoeal disease in low- and middle-income countries, particularly in young children. The increasing reports of Shigella cases associated with anti-microbial resistance are an additional element of concern. Currently, there are no licensed vaccines widely available against Shigella, but several vaccine candidates are in development. It has been demonstrated that the incidence of disease decreases following a prior Shigella infection and that serum and mucosal antibody responses are predominantly directed against the serotype-specific Shigella O-antigen portion of lipopolysaccharide membrane molecules. Many Shigella vaccine candidates are indeed O-antigen-based. Here we present the journey towards the development of a potential low-cost four-component Shigella vaccine, eliciting broad protection against the most prevalent Shigella serotypes, that makes use of the GMMA (Generalized Modules for Membrane Antigens) technology, a novel platform based on bacterial outer membranes for delivery of the O-antigen to the immune system.
Collapse
Affiliation(s)
- Francesca Micoli
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., 53100 Siena, Italy; (U.N.N.); (F.B.S.)
| | | | | |
Collapse
|
44
|
Murugaiyan J, Kumar PA, Rao GS, Iskandar K, Hawser S, Hays JP, Mohsen Y, Adukkadukkam S, Awuah WA, Jose RAM, Sylvia N, Nansubuga EP, Tilocca B, Roncada P, Roson-Calero N, Moreno-Morales J, Amin R, Kumar BK, Kumar A, Toufik AR, Zaw TN, Akinwotu OO, Satyaseela MP, van Dongen MBM. Progress in Alternative Strategies to Combat Antimicrobial Resistance: Focus on Antibiotics. Antibiotics (Basel) 2022; 11:200. [PMID: 35203804 PMCID: PMC8868457 DOI: 10.3390/antibiotics11020200] [Citation(s) in RCA: 115] [Impact Index Per Article: 57.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/01/2022] [Accepted: 02/02/2022] [Indexed: 11/24/2022] Open
Abstract
Antibiotic resistance, and, in a broader perspective, antimicrobial resistance (AMR), continues to evolve and spread beyond all boundaries. As a result, infectious diseases have become more challenging or even impossible to treat, leading to an increase in morbidity and mortality. Despite the failure of conventional, traditional antimicrobial therapy, in the past two decades, no novel class of antibiotics has been introduced. Consequently, several novel alternative strategies to combat these (multi-) drug-resistant infectious microorganisms have been identified. The purpose of this review is to gather and consider the strategies that are being applied or proposed as potential alternatives to traditional antibiotics. These strategies include combination therapy, techniques that target the enzymes or proteins responsible for antimicrobial resistance, resistant bacteria, drug delivery systems, physicochemical methods, and unconventional techniques, including the CRISPR-Cas system. These alternative strategies may have the potential to change the treatment of multi-drug-resistant pathogens in human clinical settings.
Collapse
Affiliation(s)
- Jayaseelan Murugaiyan
- Department of Biological Sciences, SRM University-AP, Guntur District, Amaravati 522240, India;
| | - P. Anand Kumar
- Department of Veterinary Microbiology, NTR College of Veterinary Science, Sri Venkateswara Veterinary University, Gannavaram 521102, India;
| | - G. Srinivasa Rao
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science, Sri Venkateswara Veterinary University, Tirupati 517502, India;
| | - Katia Iskandar
- Department of Mathématiques Informatique et Télécommunications, Université Toulouse III, Paul Sabatier, INSERM, UMR 1295, 31000 Toulouse, France;
- INSPECT-LB: Institut National de Santé Publique, d’Épidémiologie Clinique et de Toxicologie-Liban, Beirut 6573, Lebanon
- Faculty of Pharmacy, Lebanese University, Beirut 6573, Lebanon
| | | | - John P. Hays
- Department of Medical Microbiology, Infectious Diseases, Erasmus University Medical Centre (Erasmus MC), 3015 GD Rotterdam, The Netherlands;
| | - Yara Mohsen
- Department of Epidemiology, High Institute of Public Health, Alexandria University, Alexandria 21544, Egypt;
- Infectious Disease Clinical Pharmacist, Antimicrobial Stewardship Department, International Medical Center Hospital, Cairo 11511, Egypt
| | - Saranya Adukkadukkam
- Department of Biological Sciences, SRM University-AP, Guntur District, Amaravati 522240, India;
| | - Wireko Andrew Awuah
- Faculty of Medicine, Sumy State University, 40007 Sumy, Ukraine; (W.A.A.); (A.-R.T.)
| | - Ruiz Alvarez Maria Jose
- Research Coordination and Support Service, National Institute of Health (ISS) Viale Regina -Elena, 299, 00161 Rome, Italy;
| | - Nanono Sylvia
- Infectious Diseases Institute (IDI), College of Health Sciences, Makerere University, Kampala 7072, Uganda;
| | | | - Bruno Tilocca
- Department of Health Science, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (B.T.); (P.R.)
| | - Paola Roncada
- Department of Health Science, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (B.T.); (P.R.)
| | - Natalia Roson-Calero
- ISGlobal, Hospital Clínic-Universitat de Barcelona, 08036 Barcelona, Spain; (N.R.-C.); (J.M.-M.)
| | - Javier Moreno-Morales
- ISGlobal, Hospital Clínic-Universitat de Barcelona, 08036 Barcelona, Spain; (N.R.-C.); (J.M.-M.)
| | - Rohul Amin
- James P Grant School of Public Health, BRAC University, Dhaka 1212, Bangladesh;
| | - Ballamoole Krishna Kumar
- Nitte (Deemed to be University), Division of Infectious Diseases, Nitte University Centre for Science Education and Research, Deralakatte, Mangalore 575018, India;
| | - Abishek Kumar
- Department of Microbiology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal 576104, India;
| | - Abdul-Rahman Toufik
- Faculty of Medicine, Sumy State University, 40007 Sumy, Ukraine; (W.A.A.); (A.-R.T.)
| | - Thaint Nadi Zaw
- Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK;
| | - Oluwatosin O. Akinwotu
- Department of Microbiology and Biotechnology Centre, Maharaja Sayajirao University of Baroda, Vadodara 390002, India;
- Environmental and Biotechnology Unit, Department of Microbiology, University of Ibadan, 200132 Ibadan, Nigeria
| | | | | |
Collapse
|
45
|
Tondi S, Clemente B, Esposito C, Sammicheli C, Tavarini S, Martin LB, Rossi O, Micoli F, Bartolini E, Brazzoli M, Ulivieri C, Blohmke CJ, Schiavetti F. Dissecting in Vitro the Activation of Human Immune Response Induced by Shigella sonnei GMMA. Front Cell Infect Microbiol 2022; 12:767153. [PMID: 35186786 PMCID: PMC8851470 DOI: 10.3389/fcimb.2022.767153] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 01/05/2022] [Indexed: 11/28/2022] Open
Abstract
Generalized Modules for Membrane Antigens (GMMA) are outer membrane exosomes purified from Gram-negative bacteria genetically mutated to increase blebbing and reduce risk of reactogenicity. This is commonly achieved through modification of the lipid A portion of lipopolysaccharide. GMMA faithfully resemble the bacterial outer membrane surface, and therefore represent a powerful and flexible platform for vaccine development. Although GMMA-based vaccines have been demonstrated to induce a strong and functional antibody response in animals and humans maintaining an acceptable reactogenicity profile, the overall impact on immune cells and their mode of action are still poorly understood. To characterize the GMMA-induced immune response, we stimulated human peripheral blood mononuclear cells (hPBMCs) with GMMA from Shigella sonnei. We studied GMMA both with wild-type hexa-acylated lipid A and with the corresponding less reactogenic penta-acylated form. Using multicolor flow cytometry, we assessed the activation of immune cell subsets and we profiled intracellular cytokine production after GMMA stimulation. Moreover, we measured the secretion of thirty cytokines/chemokines in the cell culture supernatants. Our data indicated activation of monocytes, dendritic, NK, B, and γδ T cells. Comparison of the cytokine responses showed that, although the two GMMA have qualitatively similar profiles, GMMA with modified penta-acylated lipid A induced a lower production of pro-inflammatory cytokines/chemokines compared to GMMA with wild-type lipid A. Intracellular cytokine staining indicated monocytes and dendritic cells as the main source of the cytokines produced. Overall, these data provide new insights into the activation of key immune cells potentially targeted by GMMA-based vaccines.
Collapse
Affiliation(s)
- Serena Tondi
- GlaxoSmithKline (GSK), Preclinical Evidence Generation (PEG), Siena, Italy
- Department of Life Sciences, University of Siena, Siena, Italy
| | - Bruna Clemente
- GlaxoSmithKline (GSK), Preclinical Evidence Generation (PEG), Siena, Italy
| | - Carmen Esposito
- GlaxoSmithKline (GSK), Preclinical Evidence Generation (PEG), Siena, Italy
| | - Chiara Sammicheli
- GlaxoSmithKline (GSK), Preclinical Evidence Generation (PEG), Siena, Italy
| | - Simona Tavarini
- GlaxoSmithKline (GSK), Preclinical Evidence Generation (PEG), Siena, Italy
| | - Laura B. Martin
- GlaxoSmithKline (GSK) Vaccines Institute for Global Health S.R.L. (GVGH), Siena, Italy
| | - Omar Rossi
- GlaxoSmithKline (GSK) Vaccines Institute for Global Health S.R.L. (GVGH), Siena, Italy
| | - Francesca Micoli
- GlaxoSmithKline (GSK) Vaccines Institute for Global Health S.R.L. (GVGH), Siena, Italy
| | - Erika Bartolini
- GlaxoSmithKline (GSK), Preclinical Evidence Generation (PEG), Siena, Italy
| | - Michela Brazzoli
- GlaxoSmithKline (GSK), Preclinical Evidence Generation (PEG), Siena, Italy
| | | | - Christoph J. Blohmke
- GlaxoSmithKline (GSK), Artificial Intelligence & Machine Learning (AIML), London, United Kingdom
| | | |
Collapse
|
46
|
Gong W, Pan C, Cheng P, Wang J, Zhao G, Wu X. Peptide-Based Vaccines for Tuberculosis. Front Immunol 2022; 13:830497. [PMID: 35173740 PMCID: PMC8841753 DOI: 10.3389/fimmu.2022.830497] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 01/10/2022] [Indexed: 12/12/2022] Open
Abstract
Tuberculosis (TB) is an infectious disease caused by Mycobacterium tuberculosis. As a result of the coronavirus disease 2019 (COVID-19) pandemic, the global TB mortality rate in 2020 is rising, making TB prevention and control more challenging. Vaccination has been considered the best approach to reduce the TB burden. Unfortunately, BCG, the only TB vaccine currently approved for use, offers some protection against childhood TB but is less effective in adults. Therefore, it is urgent to develop new TB vaccines that are more effective than BCG. Accumulating data indicated that peptides or epitopes play essential roles in bridging innate and adaptive immunity and triggering adaptive immunity. Furthermore, innovations in bioinformatics, immunoinformatics, synthetic technologies, new materials, and transgenic animal models have put wings on the research of peptide-based vaccines for TB. Hence, this review seeks to give an overview of current tools that can be used to design a peptide-based vaccine, the research status of peptide-based vaccines for TB, protein-based bacterial vaccine delivery systems, and animal models for the peptide-based vaccines. These explorations will provide approaches and strategies for developing safer and more effective peptide-based vaccines and contribute to achieving the WHO's End TB Strategy.
Collapse
Affiliation(s)
- Wenping Gong
- Tuberculosis Prevention and Control Key Laboratory/Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The 8th Medical Center of PLA General Hospital, Beijing, China
| | - Chao Pan
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, China
| | - Peng Cheng
- Tuberculosis Prevention and Control Key Laboratory/Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The 8th Medical Center of PLA General Hospital, Beijing, China
- Hebei North University, Zhangjiakou City, China
| | - Jie Wang
- Tuberculosis Prevention and Control Key Laboratory/Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The 8th Medical Center of PLA General Hospital, Beijing, China
| | - Guangyu Zhao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Xueqiong Wu
- Tuberculosis Prevention and Control Key Laboratory/Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The 8th Medical Center of PLA General Hospital, Beijing, China
| |
Collapse
|
47
|
Stefanetti G, Borriello F, Richichi B, Zanoni I, Lay L. Immunobiology of Carbohydrates: Implications for Novel Vaccine and Adjuvant Design Against Infectious Diseases. Front Cell Infect Microbiol 2022; 11:808005. [PMID: 35118012 PMCID: PMC8803737 DOI: 10.3389/fcimb.2021.808005] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 12/22/2021] [Indexed: 12/14/2022] Open
Abstract
Carbohydrates are ubiquitous molecules expressed on the surface of nearly all living cells, and their interaction with carbohydrate-binding proteins is critical to many immunobiological processes. Carbohydrates are utilized as antigens in many licensed vaccines against bacterial pathogens. More recently, they have also been considered as adjuvants. Interestingly, unlike other types of vaccines, adjuvants have improved immune response to carbohydrate-based vaccine in humans only in a few cases. Furthermore, despite the discovery of many new adjuvants in the last years, aluminum salts, when needed, remain the only authorized adjuvant for carbohydrate-based vaccines. In this review, we highlight historical and recent advances on the use of glycans either as vaccine antigens or adjuvants, and we review the use of currently available adjuvants to improve the efficacy of carbohydrate-based vaccines. A better understanding of the mechanism of carbohydrate interaction with innate and adaptive immune cells will benefit the design of a new generation of glycan-based vaccines and of immunomodulators to fight both longstanding and emerging diseases.
Collapse
Affiliation(s)
- Giuseppe Stefanetti
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, United States
| | - Francesco Borriello
- Division of Immunology, Harvard Medical School and Boston Children’s Hospital, Boston, MA, United States
| | - Barbara Richichi
- Department of Chemistry “Ugo Schiff”, University of Florence, Florence, Italy
| | - Ivan Zanoni
- Division of Immunology, Division of Gastroenterology, Harvard Medical School and Boston Children’s Hospital, Boston, MA, United States
| | - Luigi Lay
- Department of Chemistry, University of Milan, Milan, Italy
| |
Collapse
|
48
|
Shigella Outer Membrane Vesicles as Promising Targets for Vaccination. Int J Mol Sci 2022; 23:ijms23020994. [PMID: 35055181 PMCID: PMC8781765 DOI: 10.3390/ijms23020994] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/11/2022] [Accepted: 01/14/2022] [Indexed: 12/17/2022] Open
Abstract
The clinical symptoms of shigellosis, a gastrointestinal infection caused by Shigella spp. range from watery diarrhea to fulminant dysentery. Endemic infections, particularly among children in developing countries, represent the majority of clinical cases. The situation is aggravated due to the high mortality rate of shigellosis, the rapid dissemination of multi-resistant Shigella strains and the induction of only serotype-specific immunity. Thus, infection prevention due to vaccination, encompassing as many of the circulating serotypes as possible, has become a topic of interest. However, vaccines have turned out to be ineffective so far. Outer membrane vesicles (OMVs) are promising novel targets for vaccination. OMVs are constitutively secreted by Gram-negative bacteria including Shigella during growth. They are composed of soluble luminal portions and an insoluble membrane and can contain toxins, bioactive periplasmic and cytoplasmic (lipo-) proteins, (phospho-) lipids, nucleic acids and/or lipopolysaccharides. Thus, OMVs play an important role in bacterial cell–cell communication, growth, survival and pathogenesis. Furthermore, they modulate the secretion and transport of biomolecules, the stress response, antibiotic resistance and immune responses of the host. Thus, OMVs serve as novel secretion machinery. Here, we discuss the current literature and highlight the properties of OMVs as potent vaccine candidates because of their immunomodulatory, antigenic and adjuvant properties.
Collapse
|
49
|
Cable J, Rappuoli R, Klemm EJ, Kang G, Mutreja A, Wright GJ, Pizza M, Castro SA, Hoffmann JP, Alter G, Carfi A, Pollard AJ, Krammer F, Gupta RK, Wagner CE, Machado V, Modjarrad K, Corey L, B Gilbert P, Dougan G, Lurie N, Bjorkman PJ, Chiu C, Nemes E, Gordon SB, Steer AC, Rudel T, Blish CA, Sandberg JT, Brennan K, Klugman KP, Stuart LM, Madhi SA, Karp CL. Innovative vaccine approaches-a Keystone Symposia report. Ann N Y Acad Sci 2022; 1511:59-86. [PMID: 35029310 DOI: 10.1111/nyas.14739] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 12/03/2021] [Indexed: 12/16/2022]
Abstract
The rapid development of COVID-19 vaccines was the result of decades of research to establish flexible vaccine platforms and understand pathogens with pandemic potential, as well as several novel changes to the vaccine discovery and development processes that partnered industry and governments. And while vaccines offer the potential to drastically improve global health, low-and-middle-income countries around the world often experience reduced access to vaccines and reduced vaccine efficacy. Addressing these issues will require novel vaccine approaches and platforms, deeper insight how vaccines mediate protection, and innovative trial designs and models. On June 28-30, 2021, experts in vaccine research, development, manufacturing, and deployment met virtually for the Keystone eSymposium "Innovative Vaccine Approaches" to discuss advances in vaccine research and development.
Collapse
Affiliation(s)
| | | | | | - Gagandeep Kang
- Division of Gastrointestinal Sciences, Christian Medical College, Vellore, India
| | - Ankur Mutreja
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID) and Department of Medicine, University of Cambridge, Cambridge, UK
| | - Gavin J Wright
- Cell Surface Signalling Laboratory, Wellcome Sanger Institute, Hinxton, UK.,Department of Biology, Hull York Medical School, and York Biomedical Research Institute, University of York, York, UK
| | | | - Sowmya Ajay Castro
- Division of Molecular Microbiology, School of Life Sciences, University of Dundee, Dundee, UK
| | - Joseph P Hoffmann
- Departments of Pediatrics and Medicine, Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, Louisiana
| | - Galit Alter
- Ragon Institute of MGH, MIT and Harvard, Harvard Medical School, Cambridge, Massachusetts.,Division of Infectious Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | | | - Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the National Institute for Health Research (NIHR) Oxford Biomedical Research Centre, Oxford, UK
| | - Florian Krammer
- The Tisch Cancer Institute and Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Ravindra K Gupta
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID) and Department of Medicine, University of Cambridge, Cambridge, UK.,Africa Health Research Institute, Durban, South Africa
| | - Caroline E Wagner
- Department of Bioengineering, McGill University, Montreal, Quebec, Canada
| | - Viviane Machado
- Measles and Respiratory Viruses Laboratory, WHO/NIC, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro, Brazil
| | - Kayvon Modjarrad
- Emerging Infectious Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland
| | - Lawrence Corey
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington.,Department of Medicine, University of Washington School of Medicine, Seattle, Washington.,Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Peter B Gilbert
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Gordon Dougan
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID) and Department of Medicine, University of Cambridge, Cambridge, UK
| | - Nicole Lurie
- Coalition for Epidemic Preparedness Innovations, Oslo, Norway.,Harvard Medical School, Boston, Massachusetts
| | - Pamela J Bjorkman
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California
| | - Christopher Chiu
- Department of Infectious Disease, Imperial College London, London, UK
| | - Elisa Nemes
- Division of Immunology, Department of Pathology, South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | | | - Andrew C Steer
- Infection and Immunity, Murdoch Children's Research Institute, Parkville, Victoria, Australia.,Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia.,Department of General Medicine, The Royal Children's Hospital, Melbourne, Victoria, Australia
| | - Thomas Rudel
- Microbiology Biocenter, University of Würzburg, Würzburg, Germany
| | - Catherine A Blish
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford Immunology Program, Stanford University School of Medicine, Stanford, California.,Chan Zuckerberg Biohub, San Francisco, California
| | - John Tyler Sandberg
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Kiva Brennan
- National Children's Research Centre, Crumlin and School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Keith P Klugman
- Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Lynda M Stuart
- Immunology Program, Benaroya Research Institute at Virginia Mason, Seattle, Washington.,Bill & Melinda Gates Foundation, Seattle, Washington
| | - Shabir A Madhi
- South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, University of the Witwatersrand, Johannesburg, South Africa
| | | |
Collapse
|
50
|
Maintenance of the Shigella sonnei virulence plasmid is dependent on its repertoire and amino acid sequence of toxin:antitoxin systems. J Bacteriol 2022; 204:e0051921. [PMID: 34978459 PMCID: PMC8923223 DOI: 10.1128/jb.00519-21] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Shigella sonnei is a major cause of bacillary dysentery and an increasing concern due to the spread of multidrug resistance. S. sonnei harbors pINV, an ∼210 kb plasmid that encodes a type III secretion system (T3SS), which is essential for virulence. During growth in the laboratory, avirulence arises spontaneously in S. sonnei at high frequency, hampering studies on and vaccine development against this important pathogen. Here, we investigated the molecular basis for the emergence of avirulence in S. sonnei and showed that avirulence mainly results from pINV loss, which is consistent with previous findings. Ancestral deletions have led to the loss from S. sonnei pINV of two toxin-antitoxin (TA) systems involved in plasmid maintenance, CcdAB and GmvAT, which are found on pINV in Shigella flexneri. We showed that the introduction of these TA systems into S. sonnei pINV reduced but did not eliminate pINV loss, while the single amino acid polymorphisms found in the S. sonnei VapBC TA system compared with S. flexneri VapBC also contributed to pINV loss. Avirulence also resulted from deletions of T3SS-associated genes in pINV through recombination between insertion sequences (ISs) on the plasmid. These events differed from those observed in S. flexneri due to the different distribution and repertoire of ISs. Our findings demonstrated that TA systems and ISs influenced plasmid dynamics and loss in S. sonnei and could be exploited for the design and evaluation of vaccines. IMPORTANCEShigella sonnei is the major cause of shigellosis in high-income and industrializing countries and is an emerging, multidrug-resistant pathogen. A significant challenge when studying this bacterium is that it spontaneously becomes avirulent during growth in the laboratory through loss of its virulence plasmid (pINV). Here, we deciphered the mechanisms leading to avirulence in S. sonnei and how the limited repertoire and amino acid sequences of plasmid-encoded toxin-antitoxin (TA) systems make the maintenance of pINV in this bacterium less efficient compared with Shigella flexneri. Our findings highlighted how subtle differences in plasmids in closely related species have marked effects and could be exploited to reduce plasmid loss in S. sonnei. This should facilitate research on this bacterium and vaccine development.
Collapse
|