1
|
Ou Q, Lu L, Zhai L, Sang S, Guan Y, Xiong Y, Liu C, Wang H, Hu Q, Wang Y. Exploration of a GMMA-Based Bivalent Vaccine Against Klebsiella pneumoniae. Vaccines (Basel) 2025; 13:226. [PMID: 40266066 PMCID: PMC11946371 DOI: 10.3390/vaccines13030226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 02/12/2025] [Accepted: 02/20/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND An emerging trend of mutual convergence between drug-resistant and highly virulent strains of K. pneumoniae has been identified, highlighting the urgent need for the development of novel vaccines. METHODS To delete the target genes and eliminate the plasmids carrying antibiotic resistance genes, CRISPR-Cas9 technology was employed to perform genome editing on a clinically isolated O2 serotype of K. pneumoniae. Subsequently, this strain was utilized as a host to express genes associated with the synthesis of O1 serotype LPSs to construct the recombinant strain capable of simultaneously expressing LPSs of both O1 and O2 serotypes. This recombinant strain was then used as the production strain for the preparation of vaccines based on GMMAs (Generalized Modules for Membrane Antigens), and its biological characteristics were characterized. Finally, the safety and immunogenicity of the vaccine were evaluated using mice as the model animals. RESULT a GMMA vaccine characterized by a high yield and low toxicity was gained. Importantly, the lipopolysaccharides (LPSs) of both O1 and O2 serotypes of K. pneumoniae were successfully expressed on the surface of the outer membrane vesicles. Following immunization with the GMMA vaccine, mice were capable of producing antibodies against the GMMA and demonstrated resistance to the invasion of both serotypes of clinically isolated K. pneumoniae. CONCLUSIONS The GMMA vaccine showed significant promise as a bivalent vaccine against K. pneumoniae.
Collapse
Affiliation(s)
- Qikun Ou
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Guangxi Medical University, 22 Shuangyong Road, Nanning 530021, China;
| | - Lu Lu
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, 20 Dongda Street, Beijing 100071, China; (L.L.); (L.Z.); (S.S.); (Y.G.); (C.L.)
| | - Lina Zhai
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, 20 Dongda Street, Beijing 100071, China; (L.L.); (L.Z.); (S.S.); (Y.G.); (C.L.)
- Beijing International Science and Technology Cooperation Base for Antiviral Drugs, Beijing Key Laboratory of Environmental and Viral Oncology, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Shuli Sang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, 20 Dongda Street, Beijing 100071, China; (L.L.); (L.Z.); (S.S.); (Y.G.); (C.L.)
| | - Yiyan Guan
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, 20 Dongda Street, Beijing 100071, China; (L.L.); (L.Z.); (S.S.); (Y.G.); (C.L.)
| | - Yuling Xiong
- Department of Clinical Laboratory, The Fourth Medical Centre, Chinese PLA General Hospital, No. 51 Fucheng Road, Beijing 100037, China;
| | - Chunjie Liu
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, 20 Dongda Street, Beijing 100071, China; (L.L.); (L.Z.); (S.S.); (Y.G.); (C.L.)
| | - Haibin Wang
- Department of Clinical Laboratory, The Fourth Medical Centre, Chinese PLA General Hospital, No. 51 Fucheng Road, Beijing 100037, China;
| | - Qiping Hu
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Guangxi Medical University, 22 Shuangyong Road, Nanning 530021, China;
| | - Yanchun Wang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, 20 Dongda Street, Beijing 100071, China; (L.L.); (L.Z.); (S.S.); (Y.G.); (C.L.)
| |
Collapse
|
2
|
Spiga O, Visibelli A, Pettini F, Roncaglia B, Santucci A. SHASI-ML: a machine learning-based approach for immunogenicity prediction in Salmonella vaccine development. Front Cell Infect Microbiol 2025; 15:1536156. [PMID: 40007603 PMCID: PMC11850321 DOI: 10.3389/fcimb.2025.1536156] [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: 11/28/2024] [Accepted: 01/22/2025] [Indexed: 02/27/2025] Open
Abstract
Introduction Accurate prediction of immunogenic proteins is crucial for vaccine development and understanding host-pathogen interactions in bacterial diseases, particularly for Salmonella infections which remain a significant global health challenge. Methods We developed SHASI-ML, a machine learning-based framework for predicting immunogenic proteins in Salmonella species. The model was trained and validated using a curated dataset of experimentally verified immunogenic and non-immunogenic proteins. Three distinct feature groups were extracted from protein sequences: global properties, sequence-derived features, and structural information. The Extreme Gradient Boosting (XGBoost) algorithm was employed for model development and optimization. Results SHASI-ML demonstrated robust performance in identifying bacterial immunogens, achieving 89.3% precision and 91.2% specificity. When applied to the Salmonella enterica serovar Typhimurium proteome, the model identified 292 novel immunogenic protein candidates. Global properties emerged as the most influential feature group in prediction accuracy, followed by structural and sequence information. The model showed superior recall and F1-scores compared to existing computational approaches. Discussion These findings establish SHASI-ML as an efficient computational tool for prioritizing immunogenic candidates in Salmonella vaccine development. By streamlining the identification of vaccine candidates early in the development process, this approach significantly reduces experimental burden and associated costs. The methodology can be applied to guide and optimize both research and industrial-scale production of Salmonella vaccines, potentially accelerating the development of more effective immunization strategies.
Collapse
Affiliation(s)
- Ottavia Spiga
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
- Competence Center Advanced Robotics and enabling digital TEchnologies & Systems 4.0 (ARTES 4.0), Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
- SienabioACTIVE-SbA, Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Anna Visibelli
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Francesco Pettini
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Bianca Roncaglia
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Annalisa Santucci
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
- Competence Center Advanced Robotics and enabling digital TEchnologies & Systems 4.0 (ARTES 4.0), Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
- SienabioACTIVE-SbA, Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| |
Collapse
|
3
|
Cuomo P, Medaglia C, Casillo A, Gentile A, Fruggiero C, Corsaro MM, Capparelli R. Phage-resistance alters Lipid A reactogenicity: a new strategy for LPS-based conjugate vaccines against Salmonella Rissen. Front Immunol 2024; 15:1450600. [PMID: 39723217 PMCID: PMC11668645 DOI: 10.3389/fimmu.2024.1450600] [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: 06/17/2024] [Accepted: 11/22/2024] [Indexed: 12/28/2024] Open
Abstract
Salmonella enterica serovar Rissen (S. Rissen) is an emerging causative agent of foodborne diseases. The current emergence of antibiotic resistance makes necessary alternative therapeutic strategies. In this study, we investigated the potential of a phage-resistant strain of S. Rissen (RR) as a tool for developing an effective lipopolysaccharide (LPS)-based vaccine. The LPS O-antigen is known to play critical roles in protective immunity against Salmonella. However, the high toxicity of the LPS lipid A moiety limits its use in vaccines. Here, we demonstrated that the acquisition of bacteriophage resistance by S. Rissen leads to structural modifications in the LPS structure. Using NMR and mass spectrometry, we characterized the LPS from phage-resistant strains as a smooth variant bearing under-acylated Lipid A portions (penta- and tetra-acylated forms). We then combined RT-qPCR and NMR-based metabolomics to explore the effects of phage resistance and LPS modification on bacterial fitness and virulence. Finally, we conducted in vivo studies to determine whether lysogeny-induced remodeling of LPS affects the host immune response. Results revealed that the under-acylated variant of LPS from RR attenuates the inflammatory response in BALB/c mice, while eliciting a specific antibody response that protects against S. Rissen (RW) infection. In conclusion, our findings suggest that phage resistance, through lipid A modification, may offer a novel strategy for reducing LPS toxicity, highlighting its potential as a promising biological approach for developing LPS-based vaccines against Salmonella infections.
Collapse
Affiliation(s)
- Paola Cuomo
- Department of Agricultural Sciences, University of Naples Federico II, Portici, Italy
- Task Force on Microbiome Studies, University of Naples Federico II, Naples, Italy
| | - Chiara Medaglia
- Functional Genomics Research Center, Fondazione Human Technopole, Milan, Italy
| | - Angela Casillo
- Department of Chemical Sciences, University of Naples Federico II, Naples, Italy
| | - Antonio Gentile
- Department of Agricultural Sciences, University of Naples Federico II, Portici, Italy
- Task Force on Microbiome Studies, University of Naples Federico II, Naples, Italy
| | - Carmine Fruggiero
- Department of Agricultural Sciences, University of Naples Federico II, Portici, Italy
| | | | - Rosanna Capparelli
- Department of Agricultural Sciences, University of Naples Federico II, Portici, Italy
- Task Force on Microbiome Studies, University of Naples Federico II, Naples, Italy
| |
Collapse
|
4
|
Velimirov B, Velimirov BA. Immune Responses Elicited by Outer Membrane Vesicles of Gram-Negative Bacteria: Important Players in Vaccine Development. Life (Basel) 2024; 14:1584. [PMID: 39768292 PMCID: PMC11678573 DOI: 10.3390/life14121584] [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: 10/11/2024] [Revised: 11/12/2024] [Accepted: 11/20/2024] [Indexed: 01/11/2025] Open
Abstract
The attractiveness of OMVs derived from Gram-negative bacteria lies in the fact that they have two biomembranes sandwiching a peptidoglycan layer. It is well known that the envelope of OMVs consists of the outer bacterial membrane [OM] and not of the inner one [IM] of the source bacterium. This implies that all outer membranous molecules found in the OM act as antigens. However, under specific conditions, some of the inner membrane proteins can be exported into the outer membrane layer and perform as antigens. A key information was that the used purification procedures for OMVs, the induction methods to increase the production of OMVs as well as the specific mutant strains obtained via genetic engineering affect the composition of potential antigens on the surface and in the lumen of the OMVs. The available literature allowed us to list the major antigens that could be defined on OMVs. The functions of the antigens within the source bacterium are discussed for a better understanding of the various available hypotheses on the biogenesis of vesicle formation. Also, the impacts of OMV antigens on the immune system using animal models are assessed. Furthermore, information on the pathways of OMVs entering the host cell is presented. An example of a bacterial infection that causes epidemic diseases, namely via Neisseria meningitidis, is used to demonstrate that OMVs derived from this pathogen elicit protective immune responses when administered as a vaccine. Furthermore, information on OMV vaccines under development is presented. The assembled knowledge allowed us to formulate a number of reasons why OMVs are attractive as vaccine platforms, as their undesirable side effects remain small, and to provide an outlook on the potential use of OMVs as a vaccine platform.
Collapse
Affiliation(s)
- Branko Velimirov
- Division of Microbiology and Molecular Biology, Medical Faculty, Private Sigmund Freud University, Freudplatz 3, 1020 Wien, Austria;
| | | |
Collapse
|
5
|
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
|
6
|
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
|
7
|
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
|
8
|
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
|
9
|
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: 6] [Impact Index Per Article: 3.0] [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
|
10
|
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: 10] [Impact Index Per Article: 5.0] [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
|
11
|
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
|
12
|
Aruta MG, Lari E, De Simone D, Semplici B, Semplici C, Dale H, Chirwa E, Kadwala I, Mbewe M, Banda H, Iturriza-Gomara M, Gordon M, Nyirenda T, Piu P, Pizza M, Berlanda Scorza F, Grappi S, Canals R, Rossi O. Characterization of Enzyme-Linked Immunosorbent Assay (ELISA) for Quantification of Antibodies against Salmonella Typhimurium and Salmonella Enteritidis O-Antigens in Human Sera. BIOTECH 2023; 12:54. [PMID: 37606441 PMCID: PMC10443281 DOI: 10.3390/biotech12030054] [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: 07/18/2023] [Revised: 08/04/2023] [Accepted: 08/09/2023] [Indexed: 08/23/2023] Open
Abstract
Nontyphoidal Salmonella (NTS) is a leading cause of morbidity and mortality caused by enteric pathogens worldwide in both children and adults, and vaccines are not yet available. The measurement of antigen-specific antibodies in the sera of vaccinated or convalescent individuals is crucial to understand the incidence of disease and the immunogenicity of vaccine candidates. A solid and standardized assay used to determine the level of specific anti-antigens IgG is therefore of paramount importance. In this work, we presented the characterization of a customized enzyme-linked immunosorbent assay (ELISA) with continuous readouts and a standardized definition of EU/mL. We assessed various performance parameters: standard curve accuracy, dilutional linearity, intermediate precision, specificity, limits of blanks, and quantification. The simplicity of the assay, its high sensitivity and specificity coupled with its low cost and the use of basic consumables and instruments without the need of high automation makes it suitable for transfer and application to different laboratories, including resource-limiting settings where the disease is endemic. This ELISA is, therefore, fit for purpose to be used for quantification of antibodies against Salmonella Typhimurium and Salmonella Enteritidis O-antigens in human samples, both for vaccine clinical trials and large sero-epidemiological studies.
Collapse
Affiliation(s)
- Maria Grazia Aruta
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., 53100 Siena, Italy; (M.G.A.); (D.D.S.); (M.I.-G.); (M.P.); (F.B.S.)
| | - Elisa Lari
- VisMederi S.r.l., 53100 Siena, Italy; (E.L.); (B.S.); (C.S.); (P.P.); (S.G.)
| | - Daniele De Simone
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., 53100 Siena, Italy; (M.G.A.); (D.D.S.); (M.I.-G.); (M.P.); (F.B.S.)
| | - Bianca Semplici
- VisMederi S.r.l., 53100 Siena, Italy; (E.L.); (B.S.); (C.S.); (P.P.); (S.G.)
| | - Claudia Semplici
- VisMederi S.r.l., 53100 Siena, Italy; (E.L.); (B.S.); (C.S.); (P.P.); (S.G.)
| | - Helen Dale
- Malawi Liverpool Wellcome Trust Programme, Blantyre 30096, Malawi; (H.D.); (E.C.); (I.K.); (M.M.); (H.B.); (M.G.)
- 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, Blantyre 30096, Malawi; (H.D.); (E.C.); (I.K.); (M.M.); (H.B.); (M.G.)
- 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, Blantyre 30096, Malawi; (H.D.); (E.C.); (I.K.); (M.M.); (H.B.); (M.G.)
- Pathology Department, Kamuzu University of Health Sciences, Blantyre 312225, Malawi;
| | - Maurice Mbewe
- Malawi Liverpool Wellcome Trust Programme, Blantyre 30096, Malawi; (H.D.); (E.C.); (I.K.); (M.M.); (H.B.); (M.G.)
- Pathology Department, Kamuzu University of Health Sciences, Blantyre 312225, Malawi;
| | - Happy Banda
- Malawi Liverpool Wellcome Trust Programme, Blantyre 30096, Malawi; (H.D.); (E.C.); (I.K.); (M.M.); (H.B.); (M.G.)
- Pathology Department, Kamuzu University of Health Sciences, Blantyre 312225, Malawi;
| | - Miren Iturriza-Gomara
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., 53100 Siena, Italy; (M.G.A.); (D.D.S.); (M.I.-G.); (M.P.); (F.B.S.)
| | - Melita Gordon
- Malawi Liverpool Wellcome Trust Programme, Blantyre 30096, Malawi; (H.D.); (E.C.); (I.K.); (M.M.); (H.B.); (M.G.)
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 3BX, UK
| | - Tonney Nyirenda
- Pathology Department, Kamuzu University of Health Sciences, Blantyre 312225, Malawi;
| | - Pietro Piu
- VisMederi S.r.l., 53100 Siena, Italy; (E.L.); (B.S.); (C.S.); (P.P.); (S.G.)
| | - Mariagrazia Pizza
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., 53100 Siena, Italy; (M.G.A.); (D.D.S.); (M.I.-G.); (M.P.); (F.B.S.)
- Imperial College South Kensington Campus, London SW7 2AZ, UK
| | - Francesco Berlanda Scorza
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., 53100 Siena, Italy; (M.G.A.); (D.D.S.); (M.I.-G.); (M.P.); (F.B.S.)
| | - Silvia Grappi
- VisMederi S.r.l., 53100 Siena, Italy; (E.L.); (B.S.); (C.S.); (P.P.); (S.G.)
| | - Rocío Canals
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., 53100 Siena, Italy; (M.G.A.); (D.D.S.); (M.I.-G.); (M.P.); (F.B.S.)
| | - Omar Rossi
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., 53100 Siena, Italy; (M.G.A.); (D.D.S.); (M.I.-G.); (M.P.); (F.B.S.)
| | | |
Collapse
|
13
|
Tognetti F, Biagini M, Denis M, Berti F, Maione D, Stranges D. Evolution of Vaccines Formulation to Tackle the Challenge of Anti-Microbial Resistant Pathogens. Int J Mol Sci 2023; 24:12054. [PMID: 37569427 PMCID: PMC10418901 DOI: 10.3390/ijms241512054] [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: 06/21/2023] [Revised: 07/14/2023] [Accepted: 07/21/2023] [Indexed: 08/13/2023] Open
Abstract
The increasing diffusion of antimicrobial resistance (AMR) across more and more bacterial species emphasizes the urgency of identifying innovative treatment strategies to counter its diffusion. Pathogen infection prevention is among the most effective strategies to prevent the spread of both disease and AMR. Since their discovery, vaccines have been the strongest prophylactic weapon against infectious diseases, with a multitude of different antigen types and formulative strategies developed over more than a century to protect populations from different pathogens. In this review, we review the main characteristics of vaccine formulations in use and under development against AMR pathogens, focusing on the importance of administering multiple antigens where possible, and the challenges associated with their development and production. The most relevant antigen classes and adjuvant systems are described, highlighting their mechanisms of action and presenting examples of their use in clinical trials against AMR. We also present an overview of the analytical and formulative strategies for multivalent vaccines, in which we discuss the complexities associated with mixing multiple components in a single formulation. This review emphasizes the importance of combining existing knowledge with advanced technologies within a Quality by Design development framework to efficiently develop vaccines against AMR pathogens.
Collapse
Affiliation(s)
- Francesco Tognetti
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padua, Italy
| | | | | | | | | | | |
Collapse
|
14
|
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] [MESH Headings] [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
|
15
|
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
|
16
|
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
|
17
|
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
|
18
|
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
|
19
|
Jiang X, Chu C, Wang Z, Gu J, Hong Y, Li Q, Jiao X. Preclinical evaluation of OMVs as potential vaccine candidates against Salmonella enterica serovar Enteritidis infection. Front Cell Infect Microbiol 2022; 12:1037607. [PMID: 36389161 PMCID: PMC9646977 DOI: 10.3389/fcimb.2022.1037607] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 10/17/2022] [Indexed: 12/15/2024] Open
Abstract
Salmonella enterica serovar Enteritidis is the most prevalent serotype that causes human infections worldwide. Consumption of S. Enteritidis-contaminated animal foods is a major source of human infections; however, eradicating bacteria from animals remains difficult. Therefore, it is necessary to develop new measures to prevent and control salmonellosis. Here, we used the outer-membrane vesicles (OMVs) of S. Enteritidis and assessed their protective efficacy and immune response in mice. Deletion of tolR in S. Enteritidis increased the production and size of OMVs compared to those in the wild type (WT) and ΔrfaQ strains. Intramuscular immunization with OMVs conferred greater protection than intraperitoneal and intranasal immunization. Moreover, OMVs extracted from both WT and ΔtolR strains provided an 83.3% protective rate in mice challenged with S. Enteritidis, which was higher than that provided by OMVs extracted from the ΔrfaQ strain. However, compared with OMVs from the ΔtolR strain, OMVs from WT and ΔrfaQ strains rapidly eradicated S. Enteritidis colonizing the liver, spleen, ileum, and cecum of BALB/c mice after immunization. Immunization with OMVs from each of the three strains induced humoral immune responses and showed no side effects on the growth of mice. Our study revealed that OMVs from various S. Enteritidis strains could be developed for use as subunit vaccine candidates against nontyphoidal Salmonella infections in mammals.
Collapse
Affiliation(s)
- Xi Jiang
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agri-Food Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, China
- Jiangsu Key Lab of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, Yangzhou, China
| | - Chao Chu
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agri-Food Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, China
- Jiangsu Key Lab of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, Yangzhou, China
| | - Zhenyu Wang
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agri-Food Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, China
- Jiangsu Key Lab of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, Yangzhou, China
| | - Jiaojie Gu
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agri-Food Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, China
- Jiangsu Key Lab of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, Yangzhou, China
| | - Yaming Hong
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agri-Food Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, China
- Jiangsu Key Lab of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, Yangzhou, China
| | - Qiuchun Li
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agri-Food Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, China
- Jiangsu Key Lab of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, Yangzhou, China
| | - Xinan Jiao
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agri-Food Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, China
- Jiangsu Key Lab of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, Yangzhou, China
| |
Collapse
|
20
|
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: 3.0] [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
|
21
|
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: 50] [Impact Index Per Article: 16.7] [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
|
22
|
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: 3] [Impact Index Per Article: 1.0] [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
|
23
|
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: 33] [Impact Index Per Article: 11.0] [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
|
24
|
Parveen S, Subramanian K. Emerging Roles of Extracellular Vesicles in Pneumococcal Infections: Immunomodulators to Potential Novel Vaccine Candidates. Front Cell Infect Microbiol 2022; 12:836070. [PMID: 35237534 PMCID: PMC8882830 DOI: 10.3389/fcimb.2022.836070] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 01/24/2022] [Indexed: 11/13/2022] Open
Abstract
The Gram-positive bacterial pathogen, Streptococcus pneumoniae is a major global health threat that kills over one million people worldwide. The pneumococcus commonly colonizes the nasopharynx asymptomatically as a commensal, but is also capable of causing a wide range of life-threatening diseases such as pneumonia, meningitis and septicemia upon migration into the lower respiratory tract and spread to internal organs. Emergence of antibiotic resistant strains and non-vaccine serotypes has led to the classification of pneumococcal bacteria as a priority pathogen by the World Health Organization that needs urgent research into bacterial pathogenesis and development of novel vaccine strategies. Extracellular vesicles are spherical membrane bound structures that are released by both pathogen and host cells, and influence bacterial pathogenesis as well as the immune response. Recent studies have found that while bacterial vesicles shuttle virulence factors and toxins into host cells and regulate inflammatory responses, vesicles released from the infected host cells contain both bacterial and host proteins that are antigenic and immunomodulatory. Bacterial membrane vesicles have great potential to be developed as cell-free vaccine candidates in the future due to their immunogenicity and biostability. Host-derived vesicles isolated from patient biofluids such as blood and bronchoalveolar lavage could be used to identify potential diagnostic biomarkers as well as engineered to deliver desired payloads to specific target cells for immunotherapy. In this review, we summarize the recent developments on the role of bacterial and host vesicles in pneumococcal infections and future prospects in developing novel therapeutics and diagnostics for control of invasive pneumococcal diseases.
Collapse
|
25
|
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: 1.0] [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
|
26
|
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: 11.3] [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
|
27
|
Piccioli D, Bartolini E, Micoli F. GMMA as a 'plug and play' technology to tackle infectious disease to improve global health: context and perspectives for the future. Expert Rev Vaccines 2021; 21:163-172. [PMID: 34913415 DOI: 10.1080/14760584.2022.2009803] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Generalized-Modules-for-Membrane-Antigens (GMMA) is a technology platform developed to design outer membrane vesicle (OMV)-based vaccines. GMMA are basically OMVs derived from a bacterial strain specifically engineered to obtain a fit-for-purpose and affordable vaccine by potentiating, or deleting, expression of specific genes. OMVs can be used as a carrier for antigens by inducing their expression on them, with the aim to improve antigen immunogenicity and design multivalent combination vaccines. AREAS COVERED We expanded this finding to show that the chemical conjugation of different proteic and/or polysaccharidic antigens, to GMMA, is a methodology complementary to the genetic manipulation to obtain highly effective combination vaccines. Here we discuss our findings with a specific focus on the impact that GMMA technology can have on global health, as this technology platform is particularly suited to support the development of affordable vaccines for low-income countries. EXPERT OPINION We believe that it is critical to elucidate the mode of action of GMMA immunogenicity and have provided a summarized description of the immunological questions to be addressed in the near future. The improved knowledge of GMMA might lead to designing more effective and safer GMMA-based vaccines to tackle the most serious vaccine-preventable diseases.
Collapse
Affiliation(s)
| | | | - Francesca Micoli
- GSK Vaccine Institute for Global Health (GVGH), Preclinical Function, Siena, Italy
| |
Collapse
|
28
|
Micoli F, Alfini R, Giannelli C. Methods for Assessment of OMV/GMMA Quality and Stability. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2021; 2414:227-279. [PMID: 34784041 DOI: 10.1007/978-1-0716-1900-1_14] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Outer membrane vesicles (OMV) represent a promising platform for the development of vaccines against bacterial pathogens. More recently, bacteria have been genetically modified to increase OMV yield and modulate the design of resulting particles, also named generalized modules for membrane antigens (GMMA). OMV/GMMA resemble the bacterial surface of the pathogen, where key antigens to elicit a protective immune response are and contain pathogen-associated molecular patterns (e.g., lipopolysaccharides, lipoproteins) conferring self-adjuvanticity. On the other hand, OMV/GMMA are quite complex molecules and a comprehensive panel of analytical methods is needed to ensure quality, consistency of manufacture and to follow their stability over time. Here, we describe several procedures that can be used for OMV/GMMA characterization as particles and for analysis of key antigens displayed on their surface.
Collapse
Affiliation(s)
| | - Renzo Alfini
- GSK Vaccines Institute for Global Health, Siena, Italy
| | | |
Collapse
|
29
|
Micoli F, Alfini R, Di Benedetto R, Necchi F, Schiavo F, Mancini F, Carducci M, Oldrini D, Pitirollo O, Gasperini G, Balocchi C, Bechi N, Brunelli B, Piccioli D, Adamo R. Generalized Modules for Membrane Antigens as Carrier for Polysaccharides: Impact of Sugar Length, Density, and Attachment Site on the Immune Response Elicited in Animal Models. Front Immunol 2021; 12:719315. [PMID: 34594333 PMCID: PMC8477636 DOI: 10.3389/fimmu.2021.719315] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 08/16/2021] [Indexed: 11/13/2022] Open
Abstract
Nanoparticle systems are being explored for the display of carbohydrate antigens, characterized by multimeric presentation of glycan epitopes and special chemico-physical properties of nano-sized particles. Among them, outer membrane vesicles (OMVs) are receiving great attention, combining antigen presentation with the immunopotentiator effect of the Toll-like receptor agonists naturally present on these systems. In this context, we are testing Generalized Modules for Membrane Antigens (GMMA), OMVs naturally released from Gram-negative bacteria mutated to increase blebbing, as carrier for polysaccharides. Here, we investigated the impact of saccharide length, density, and attachment site on the immune response elicited by GMMA in animal models, using a variety of structurally diverse polysaccharides from different pathogens (i.e., Neisseria meningitidis serogroup A and C, Haemophilus influenzae type b, and streptococcus Group A Carbohydrate and Salmonella Typhi Vi). Anti-polysaccharide immune response was not affected by the number of saccharides per GMMA particle. However, lower saccharide loading can better preserve the immunogenicity of GMMA as antigen. In contrast, saccharide length needs to be optimized for each specific antigen. Interestingly, GMMA conjugates induced strong functional immune response even when the polysaccharides were linked to sugars on GMMA. We also verified that GMMA conjugates elicit a T-dependent humoral immune response to polysaccharides that is strictly dependent on the nature of the polysaccharide. The results obtained are important to design novel glycoconjugate vaccines using GMMA as carrier and support the development of multicomponent glycoconjugate vaccines where GMMA can play the dual role of carrier and antigen. In addition, this work provides significant insights into the mechanism of action of glycoconjugates.
Collapse
Affiliation(s)
| | - Renzo Alfini
- GSK Vaccines Institute for Global Health (GVGH), Siena, Italy
| | | | | | - Fabiola Schiavo
- GSK Vaccines Institute for Global Health (GVGH), Siena, Italy
| | | | | | - Davide Oldrini
- GSK Vaccines Institute for Global Health (GVGH), Siena, Italy
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Pan C, Yue H, Zhu L, Ma GH, Wang HL. Prophylactic vaccine delivery systems against epidemic infectious diseases. Adv Drug Deliv Rev 2021; 176:113867. [PMID: 34280513 PMCID: PMC8285224 DOI: 10.1016/j.addr.2021.113867] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/20/2021] [Accepted: 07/11/2021] [Indexed: 01/04/2023]
Abstract
Prophylactic vaccines have evolved from traditional whole-cell vaccines to safer subunit vaccines. However, subunit vaccines still face problems, such as poor immunogenicity and low efficiency, while traditional adjuvants are usually unable to meet specific response needs. Advanced delivery vectors are important to overcome these barriers; they have favorable safety and effectiveness, tunable properties, precise location, and immunomodulatory capabilities. Nevertheless, there has been no systematic summary of the delivery systems to cover a wide range of infectious pathogens. We herein summarized and compared the delivery systems for major or epidemic infectious diseases caused by bacteria, viruses, fungi, and parasites. We also included the newly licensed vaccines (e.g., COVID-19 vaccines) and those close to licensure. Furthermore, we highlighted advanced delivery systems with high efficiency, cross-protection, or long-term protection against epidemic pathogens, and we put forward prospects and thoughts on the development of future prophylactic vaccines.
Collapse
Affiliation(s)
- Chao Pan
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing 100071, PR China
| | - Hua Yue
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Li Zhu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing 100071, PR China
| | - Guang-Hui Ma
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China.
| | - Heng-Liang Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing 100071, PR China.
| |
Collapse
|
31
|
Mancini F, Micoli F, Necchi F, Pizza M, Berlanda Scorza F, Rossi O. GMMA-Based Vaccines: The Known and The Unknown. Front Immunol 2021; 12:715393. [PMID: 34413858 PMCID: PMC8368434 DOI: 10.3389/fimmu.2021.715393] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 07/19/2021] [Indexed: 11/20/2022] Open
Abstract
Generalized Modules for Membrane Antigens (GMMA) are outer membrane vesicles derived from Gram-negative bacteria engineered to provide an over-vesiculating phenotype, which represent an attractive platform for the design of affordable vaccines. GMMA can be further genetically manipulated to modulate the risk of systemic reactogenicity and to act as delivery system for heterologous polysaccharide or protein antigens. GMMA are able to induce strong immunogenicity and protection in animal challenge models, and to be well-tolerated and immunogenic in clinical studies. The high immunogenicity could be ascribed to their particulate size, to their ability to present to the immune system multiple antigens in a natural conformation which mimics the bacterial environment, as well as to their intrinsic self-adjuvanticity. However, GMMA mechanism of action and the role in adjuvanticity are still unclear and need further investigation. In this review, we discuss progresses in the development of the GMMA vaccine platform, highlighting successful applications and identifying knowledge gaps and potential challenges.
Collapse
Affiliation(s)
- Francesca Mancini
- GlaxoSmithKline (GSK) Vaccines Institute for Global Health (GVGH), Siena, Italy
| | - Francesca Micoli
- GlaxoSmithKline (GSK) Vaccines Institute for Global Health (GVGH), Siena, Italy
| | - Francesca Necchi
- GlaxoSmithKline (GSK) Vaccines Institute for Global Health (GVGH), Siena, Italy
| | - Mariagrazia Pizza
- GlaxoSmithKline (GSK) Vaccines Institute for Global Health (GVGH), Siena, Italy
| | | | - Omar Rossi
- GlaxoSmithKline (GSK) Vaccines Institute for Global Health (GVGH), Siena, Italy
| |
Collapse
|
32
|
Harvey DJ. ANALYSIS OF CARBOHYDRATES AND GLYCOCONJUGATES BY MATRIX-ASSISTED LASER DESORPTION/IONIZATION MASS SPECTROMETRY: AN UPDATE FOR 2015-2016. MASS SPECTROMETRY REVIEWS 2021; 40:408-565. [PMID: 33725404 DOI: 10.1002/mas.21651] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 07/24/2020] [Indexed: 06/12/2023]
Abstract
This review is the ninth update of the original article published in 1999 on the application of matrix-assisted laser desorption/ionization (MALDI) mass spectrometry to the analysis of carbohydrates and glycoconjugates and brings coverage of the literature to the end of 2016. Also included are papers that describe methods appropriate to analysis by MALDI, such as sample preparation techniques, even though the ionization method is not MALDI. Topics covered in the first part of the review include general aspects such as theory of the MALDI process, matrices, derivatization, MALDI imaging, fragmentation and arrays. The second part of the review is devoted to applications to various structural types such as oligo- and poly-saccharides, glycoproteins, glycolipids, glycosides and biopharmaceuticals. Much of this material is presented in tabular form. The third part of the review covers medical and industrial applications of the technique, studies of enzyme reactions and applications to chemical synthesis. The reported work shows increasing use of combined new techniques such as ion mobility and the enormous impact that MALDI imaging is having. MALDI, although invented over 30 years ago is still an ideal technique for carbohydrate analysis and advancements in the technique and range of applications show no sign of deminishing. © 2020 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- David J Harvey
- Nuffield Department of Medicine, Target Discovery Institute, University of Oxford, Roosevelt Drive, Oxford, OX3 7FZ, United Kingdom
| |
Collapse
|
33
|
Neisseria meningitidis Factor H Binding Protein Surface Exposure on Salmonella Typhimurium GMMA Is Critical to Induce an Effective Immune Response against Both Diseases. Pathogens 2021; 10:pathogens10060726. [PMID: 34207575 PMCID: PMC8229706 DOI: 10.3390/pathogens10060726] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/26/2021] [Accepted: 06/03/2021] [Indexed: 11/18/2022] Open
Abstract
GMMA, outer membrane vesicles resulting from hyperblebbing mutated bacterial strains, are a versatile vaccine platform for displaying both homologous and heterologous antigens. Periplasmic expression is a popular technique for protein expression in the lumen of the blebs. However, the ability of internalized antigens to induce antibody responses has not been extensively investigated. Herein, the Neisseria meningitidis factor H binding protein (fHbp) was heterologously expressed in the lumen of O-antigen positive (OAg+) and O-antigen negative (OAg−) Salmonella Typhimurium GMMA. Only the OAg− GMMA induced an anti-fHbp IgG response in mice if formulated on Alum, although it was weak and much lower compared to the recombinant fHbp. The OAg− GMMA on Alum showed partial instability, with possible exposure of fHbp to the immune system. When we chemically conjugated fHbp to the surface of both OAg+ and OAg− GMMA, these constructs induced a stronger functional response compared to the fHbp immunization alone. Moreover, the OAg+ GMMA construct elicited a strong response against both the target antigens (fHbp and OAg), with no immune interference observed. This result suggests that antigen localization on GMMA surface can play a critical role in the induction of an effective immune response and can encourage the development of GMMA based vaccines delivering key protective antigens on their surface.
Collapse
|
34
|
Zingl FG, Leitner DR, Thapa HB, Schild S. Outer membrane vesicles as versatile tools for therapeutic approaches. MICROLIFE 2021; 2:uqab006. [PMID: 37223254 PMCID: PMC10117751 DOI: 10.1093/femsml/uqab006] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/05/2021] [Indexed: 05/25/2023]
Abstract
Budding of the bacterial surface results in the formation and secretion of outer membrane vesicles, which is a conserved phenomenon observed in Gram-negative bacteria. Recent studies highlight that these sphere-shaped facsimiles of the donor bacterium's surface with enclosed periplasmic content may serve multiple purposes for their host bacterium. These include inter- and intraspecies cell-cell communication, effector delivery to target cells and bacterial adaptation strategies. This review provides a concise overview of potential medical applications to exploit outer membrane vesicles for therapeutic approaches. Due to the fact that outer membrane vesicles resemble the surface of their donor cells, they represent interesting nonliving candidates for vaccine development. Furthermore, bacterial donor species can be genetically engineered to display various proteins and glycans of interest on the outer membrane vesicle surface or in their lumen. Outer membrane vesicles also possess valuable bioreactor features as they have the natural capacity to protect, stabilize and enhance the activity of luminal enzymes. Along these features, outer membrane vesicles not only might be suitable for biotechnological applications but may also enable cell-specific delivery of designed therapeutics as they are efficiently internalized by nonprofessional phagocytes. Finally, outer membrane vesicles are potent modulators of our immune system with pro- and anti-inflammatory properties. A deeper understanding of immunoregulatory effects provoked by different outer membrane vesicles is the basis for their possible future applications ranging from inflammation and immune response modulation to anticancer therapy.
Collapse
Affiliation(s)
- Franz G Zingl
- Institute of Molecular Biosciences, University of Graz, Humboldtstrasse 50, 8010 Graz, Austria
| | - Deborah R Leitner
- Institute of Molecular Biosciences, University of Graz, Humboldtstrasse 50, 8010 Graz, Austria
| | - Himadri B Thapa
- 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, Austria
- Field of Excellence BioHealth, University of Graz, 8010 Graz, Austria
| |
Collapse
|
35
|
Fiorino F, Pettini E, Koeberling O, Ciabattini A, Pozzi G, Martin LB, Medaglini D. Long-Term Anti-Bacterial Immunity against Systemic Infection by Salmonella enterica Serovar Typhimurium Elicited by a GMMA-Based Vaccine. Vaccines (Basel) 2021; 9:495. [PMID: 34065899 PMCID: PMC8150838 DOI: 10.3390/vaccines9050495] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/04/2021] [Accepted: 05/05/2021] [Indexed: 12/27/2022] Open
Abstract
Salmonella Typhimurium (STm) represents the most prevalent cause of invasive non-typhoidal Salmonella (iNTS) disease, and currently no licensed vaccine is available. In this work we characterized the long-term anti-bacterial immunity elicited by a STm vaccine based on Generalized Modules of Membrane Antigens (GMMA) delivering O:4,5 antigen, using a murine model of systemic infection. Subcutaneous immunization of mice with STmGMMA/Alhydrogel elicited rapid, high, and persistent antigen-specific serum IgG and IgM responses. The serum was bactericidal in vitro. O:4,5-specific IgG were also detected in fecal samples after immunization and positively correlated with IgG observed in intestinal washes. Long-lived plasma cells and O:4,5-specific memory B cells were detected in spleen and bone marrow. After systemic STm challenge, a significant reduction of bacterial load in blood, spleen, and liver, as well as a reduction of circulating neutrophils and G-CSF glycoprotein was observed in STmGMMA/Alhydrogel immunized mice compared to untreated animals. Taken together, these data support the development of a GMMA-based vaccine for prevention of iNTS disease.
Collapse
Affiliation(s)
- Fabio Fiorino
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (F.F.); (E.P.); (A.C.); (G.P.)
| | - Elena Pettini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (F.F.); (E.P.); (A.C.); (G.P.)
| | - Oliver Koeberling
- GSK Vaccines Institute for Global Health S.r.l., 53100 Siena, Italy;
| | - Annalisa Ciabattini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (F.F.); (E.P.); (A.C.); (G.P.)
| | - Gianni Pozzi
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (F.F.); (E.P.); (A.C.); (G.P.)
| | - Laura B. Martin
- GSK Vaccines Institute for Global Health S.r.l., 53100 Siena, Italy;
| | - Donata Medaglini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (F.F.); (E.P.); (A.C.); (G.P.)
| |
Collapse
|
36
|
Salmonella Paratyphi A Outer Membrane Vesicles Displaying Vi Polysaccharide as a Multivalent Vaccine against Enteric Fever. Infect Immun 2021; 89:IAI.00699-20. [PMID: 33318138 DOI: 10.1128/iai.00699-20] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 12/07/2020] [Indexed: 12/23/2022] Open
Abstract
Typhoid and paratyphoid fevers have a high incidence worldwide and coexist in many geographical areas, especially in low-middle-income countries (LMIC) in South and Southeast Asia. There is extensive consensus on the urgent need for better and affordable vaccines against systemic Salmonella infections. Generalized modules for membrane antigens (GMMA), outer membrane exosomes shed by Salmonella bacteria genetically manipulated to increase blebbing, resemble the bacterial surface where protective antigens are displayed in their native environment. Here, we engineered S Paratyphi A using the pDC5-viaB plasmid to generate GMMA displaying the heterologous S Typhi Vi antigen together with the homologous O:2 O antigen. The presence of both Vi and O:2 was confirmed by flow cytometry on bacterial cells, and their amount was quantified on the resulting vesicles through a panel of analytical methods. When tested in mice, such GMMA induced a strong antibody response against both Vi and O:2, and these antibodies were functional in a serum bactericidal assay. Our approach yielded a bivalent vaccine candidate able to induce immune responses against different Salmonella serovars, which could benefit LMIC residents and travelers.
Collapse
|
37
|
Palmieri E, Arato V, Oldrini D, Ricchetti B, Aruta MG, Pansegrau W, Marchi S, Giusti F, Ferlenghi I, Rossi O, Alfini R, Giannelli C, Gasperini G, Necchi F, Micoli F. Stability of Outer Membrane Vesicles-Based Vaccines, Identifying the Most Appropriate Methods to Detect Changes in Vaccine Potency. Vaccines (Basel) 2021; 9:229. [PMID: 33800727 PMCID: PMC7998687 DOI: 10.3390/vaccines9030229] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/01/2021] [Accepted: 03/02/2021] [Indexed: 01/15/2023] Open
Abstract
Ensuring the stability of vaccines is crucial to successfully performing global immunization programs. Outer Membrane Vesicles (OMV) are receiving great attention as vaccine platforms. OMV are complex molecules and few data have been collected so far on their stability. OMV produced by bacteria, genetically modified to increase their spontaneous release, simplifying their production, are also known as Generalized Modules for Membrane Antigens (GMMA). We have performed accelerated stability studies on GMMA from different pathogens and verified the ability of physico-chemical and immunological methods to detect possible changes. High-temperature conditions (100 °C for 40 min) did not affect GMMA stability and immunogenicity in mice, in contrast to the effect of milder temperatures for a longer period of time (37 °C or 50 °C for 4 weeks). We identified critical quality attributes to monitor during stability assessment that could impact vaccine efficacy. In particular, specific recognition of antigens by monoclonal antibodies through competitive ELISA assays may replace in vivo tests for the potency assessment of GMMA-based vaccines.
Collapse
Affiliation(s)
- Elena Palmieri
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., Via Fiorentina 1, 53100 Siena, Italy; (E.P.); (V.A.); (D.O.); (B.R.); (M.G.A.); (O.R.); (R.A.); (C.G.); (G.G.); (F.N.)
| | - Vanessa Arato
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., Via Fiorentina 1, 53100 Siena, Italy; (E.P.); (V.A.); (D.O.); (B.R.); (M.G.A.); (O.R.); (R.A.); (C.G.); (G.G.); (F.N.)
| | - Davide Oldrini
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., Via Fiorentina 1, 53100 Siena, Italy; (E.P.); (V.A.); (D.O.); (B.R.); (M.G.A.); (O.R.); (R.A.); (C.G.); (G.G.); (F.N.)
| | - Beatrice Ricchetti
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., Via Fiorentina 1, 53100 Siena, Italy; (E.P.); (V.A.); (D.O.); (B.R.); (M.G.A.); (O.R.); (R.A.); (C.G.); (G.G.); (F.N.)
| | - Maria Grazia Aruta
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., Via Fiorentina 1, 53100 Siena, Italy; (E.P.); (V.A.); (D.O.); (B.R.); (M.G.A.); (O.R.); (R.A.); (C.G.); (G.G.); (F.N.)
| | - Werner Pansegrau
- GSK, Via Fiorentina 1, 53100 Siena, Italy; (W.P.); (S.M.); (F.G.); (I.F.)
| | - Sara Marchi
- GSK, Via Fiorentina 1, 53100 Siena, Italy; (W.P.); (S.M.); (F.G.); (I.F.)
| | - Fabiola Giusti
- GSK, Via Fiorentina 1, 53100 Siena, Italy; (W.P.); (S.M.); (F.G.); (I.F.)
| | - Ilaria Ferlenghi
- GSK, Via Fiorentina 1, 53100 Siena, Italy; (W.P.); (S.M.); (F.G.); (I.F.)
| | - Omar Rossi
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., Via Fiorentina 1, 53100 Siena, Italy; (E.P.); (V.A.); (D.O.); (B.R.); (M.G.A.); (O.R.); (R.A.); (C.G.); (G.G.); (F.N.)
| | - Renzo Alfini
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., Via Fiorentina 1, 53100 Siena, Italy; (E.P.); (V.A.); (D.O.); (B.R.); (M.G.A.); (O.R.); (R.A.); (C.G.); (G.G.); (F.N.)
| | - Carlo Giannelli
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., Via Fiorentina 1, 53100 Siena, Italy; (E.P.); (V.A.); (D.O.); (B.R.); (M.G.A.); (O.R.); (R.A.); (C.G.); (G.G.); (F.N.)
| | - Gianmarco Gasperini
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., Via Fiorentina 1, 53100 Siena, Italy; (E.P.); (V.A.); (D.O.); (B.R.); (M.G.A.); (O.R.); (R.A.); (C.G.); (G.G.); (F.N.)
| | - Francesca Necchi
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., Via Fiorentina 1, 53100 Siena, Italy; (E.P.); (V.A.); (D.O.); (B.R.); (M.G.A.); (O.R.); (R.A.); (C.G.); (G.G.); (F.N.)
| | - Francesca Micoli
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., Via Fiorentina 1, 53100 Siena, Italy; (E.P.); (V.A.); (D.O.); (B.R.); (M.G.A.); (O.R.); (R.A.); (C.G.); (G.G.); (F.N.)
| |
Collapse
|
38
|
Sears KT, Galen JE, Tennant SM. Advances in the development of Salmonella-based vaccine strategies for protection against Salmonellosis in humans. J Appl Microbiol 2021; 131:2640-2658. [PMID: 33665941 PMCID: PMC9292744 DOI: 10.1111/jam.15055] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/18/2021] [Accepted: 03/02/2021] [Indexed: 11/29/2022]
Abstract
Salmonella spp. are important human pathogens globally causing millions of cases of typhoid fever and non‐typhoidal salmonellosis annually. There are only a few vaccines licensed for use in humans which all target Salmonella enterica serovar Typhi. Vaccine development is hampered by antigenic diversity between the thousands of serovars capable of causing infection in humans. However, a number of attenuated candidate vaccine strains are currently being developed. As facultative intracellular pathogens with multiple systems for transporting effector proteins to host cells, attenuated Salmonella strains can also serve as ideal tools for the delivery of foreign antigens to create multivalent live carrier vaccines for simultaneous immunization against several unrelated pathogens. Further, the ease with which Salmonella can be genetically modified and the extensive knowledge of the virulence mechanisms of this pathogen means that this bacterium has often served as a model organism to test new approaches. In this review we focus on (1) recent advances in live attenuated Salmonella vaccine development, (2) improvements in expression of foreign antigens in carrier vaccines and (3) adaptation of attenuated strains as sources of purified antigens and vesicles that can be used for subunit and conjugate vaccines or together with attenuated vaccine strains in heterologous prime‐boosting immunization strategies. These advances have led to the development of new vaccines against Salmonella which have or will soon be tested in clinical trials.
Collapse
Affiliation(s)
- K T Sears
- Center for Vaccine Development and Global Health, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - J E Galen
- Center for Vaccine Development and Global Health, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - S M Tennant
- Center for Vaccine Development and Global Health, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
39
|
Sokaribo AS, Perera SR, Sereggela Z, Krochak R, Balezantis LR, Xing X, Lam S, Deck W, Attah-Poku S, Abbott DW, Tamuly S, White AP. A GMMA-CPS-Based Vaccine for Non-Typhoidal Salmonella. Vaccines (Basel) 2021; 9:vaccines9020165. [PMID: 33671372 PMCID: PMC7922415 DOI: 10.3390/vaccines9020165] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 02/10/2021] [Accepted: 02/11/2021] [Indexed: 11/16/2022] Open
Abstract
Non-typhoidal Salmonella are a major cause of gastroenteritis worldwide, as well as causing bloodstream infections in sub-Saharan Africa with a high fatality rate. No vaccine is currently available for human use. Current vaccine development strategies are focused on capsular polysaccharides (CPS) present on the surface of non-typhoidal Salmonella. This study aimed to boost the amount of CPS purified from S. Typhimurium for immunization trials. Random mutagenesis with Tn10 transposon increased the production of CPS colanic acid, by 10-fold compared to wildtype. Immunization with colanic acid or colanic acid conjugated to truncated glycoprotein D or inactivated diphtheria toxin did not induce a protective immune response in mice. However, immunization with Generalized Modules for Membrane Antigens (GMMAs) isolated from colanic acid overproducing isolates reduced Salmonella colonization in mice. Our results support the development of a GMMA-CPS-based vaccine against non-typhoidal Salmonella.
Collapse
Affiliation(s)
- Akosiererem S. Sokaribo
- Vaccine and Infectious Disease Organization-International Vaccine Centre, Saskatoon, SK S7N5E3, Canada; (A.S.S.); (S.R.P.); (Z.S.); (R.K.); (L.R.B.); (S.L.); (W.D.); (S.A.-P.)
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, SK S7N5E5, Canada
| | - Sumudu R. Perera
- Vaccine and Infectious Disease Organization-International Vaccine Centre, Saskatoon, SK S7N5E3, Canada; (A.S.S.); (S.R.P.); (Z.S.); (R.K.); (L.R.B.); (S.L.); (W.D.); (S.A.-P.)
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, SK S7N5E5, Canada
| | - Zoe Sereggela
- Vaccine and Infectious Disease Organization-International Vaccine Centre, Saskatoon, SK S7N5E3, Canada; (A.S.S.); (S.R.P.); (Z.S.); (R.K.); (L.R.B.); (S.L.); (W.D.); (S.A.-P.)
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, SK S7N5E5, Canada
| | - Ryan Krochak
- Vaccine and Infectious Disease Organization-International Vaccine Centre, Saskatoon, SK S7N5E3, Canada; (A.S.S.); (S.R.P.); (Z.S.); (R.K.); (L.R.B.); (S.L.); (W.D.); (S.A.-P.)
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, SK S7N5E5, Canada
| | - Lindsay R. Balezantis
- Vaccine and Infectious Disease Organization-International Vaccine Centre, Saskatoon, SK S7N5E3, Canada; (A.S.S.); (S.R.P.); (Z.S.); (R.K.); (L.R.B.); (S.L.); (W.D.); (S.A.-P.)
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, SK S7N5E5, Canada
| | - Xiaohui Xing
- Agriculture and Agri-Food Canada, Lethbridge Research and Development Centre, Lethbridge, AB T1J4B1, Canada; (X.X.); (D.W.A.)
| | - Shirley Lam
- Vaccine and Infectious Disease Organization-International Vaccine Centre, Saskatoon, SK S7N5E3, Canada; (A.S.S.); (S.R.P.); (Z.S.); (R.K.); (L.R.B.); (S.L.); (W.D.); (S.A.-P.)
| | - William Deck
- Vaccine and Infectious Disease Organization-International Vaccine Centre, Saskatoon, SK S7N5E3, Canada; (A.S.S.); (S.R.P.); (Z.S.); (R.K.); (L.R.B.); (S.L.); (W.D.); (S.A.-P.)
| | - Sam Attah-Poku
- Vaccine and Infectious Disease Organization-International Vaccine Centre, Saskatoon, SK S7N5E3, Canada; (A.S.S.); (S.R.P.); (Z.S.); (R.K.); (L.R.B.); (S.L.); (W.D.); (S.A.-P.)
| | - Dennis Wade Abbott
- Agriculture and Agri-Food Canada, Lethbridge Research and Development Centre, Lethbridge, AB T1J4B1, Canada; (X.X.); (D.W.A.)
| | - Shantanu Tamuly
- Department of Veterinary Biochemistry, College of Veterinary Science, Assam Agricultural University, Khanapara, Guwahati 781022, Assam, India;
| | - Aaron P. White
- Vaccine and Infectious Disease Organization-International Vaccine Centre, Saskatoon, SK S7N5E3, Canada; (A.S.S.); (S.R.P.); (Z.S.); (R.K.); (L.R.B.); (S.L.); (W.D.); (S.A.-P.)
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, SK S7N5E5, Canada
- Correspondence: ; Tel.: +01-306-966-7485
| |
Collapse
|
40
|
Zanella I, König E, Tomasi M, Gagliardi A, Frattini L, Fantappiè L, Irene C, Zerbini F, Caproni E, Isaac SJ, Grigolato M, Corbellari R, Valensin S, Ferlenghi I, Giusti F, Bini L, Ashhab Y, Grandi A, Grandi G. Proteome-minimized outer membrane vesicles from Escherichia coli as a generalized vaccine platform. J Extracell Vesicles 2021; 10:e12066. [PMID: 33643549 PMCID: PMC7886703 DOI: 10.1002/jev2.12066] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 12/26/2020] [Accepted: 01/13/2021] [Indexed: 02/01/2023] Open
Abstract
Because of their potent adjuvanticity, ease of manipulation and simplicity of production Gram‐negative Outer Membrane Vesicles OMVs have the potential to become a highly effective vaccine platform. However, some optimization is required, including the reduction of the number of endogenous proteins, the increase of the loading capacity with respect to heterologous antigens, the enhancement of productivity in terms of number of vesicles per culture volume. In this work we describe the use of Synthetic Biology to create Escherichia coli BL21(DE3)Δ60, a strain releasing OMVs (OMVsΔ60) deprived of 59 endogenous proteins. The strain produces large quantities of vesicles (> 40 mg/L under laboratory conditions), which can accommodate recombinant proteins to a level ranging from 5% to 30% of total OMV proteins. Moreover, also thanks to the absence of immune responses toward the inactivated endogenous proteins, OMVsΔ60 decorated with heterologous antigens/epitopes elicit elevated antigens/epitopes‐specific antibody titers and high frequencies of epitope‐specific IFN‐γ‐producing CD8+ T cells. Altogether, we believe that E. coli BL21(DE3)Δ60 have the potential to become a workhorse factory for novel OMV‐based vaccines.
Collapse
Affiliation(s)
- Ilaria Zanella
- Department of Cellular, Computational and Integrative Biology (CIBIO) Laboratory of Synthetic and Structural Vaccinology University of Trento Trento Italy
| | - Enrico König
- Department of Cellular, Computational and Integrative Biology (CIBIO) Laboratory of Synthetic and Structural Vaccinology University of Trento Trento Italy
| | - Michele Tomasi
- Department of Cellular, Computational and Integrative Biology (CIBIO) Laboratory of Synthetic and Structural Vaccinology University of Trento Trento Italy
| | - Assunta Gagliardi
- Department of Cellular, Computational and Integrative Biology (CIBIO) Laboratory of Synthetic and Structural Vaccinology University of Trento Trento Italy
| | - Luca Frattini
- Department of Cellular, Computational and Integrative Biology (CIBIO) Laboratory of Synthetic and Structural Vaccinology University of Trento Trento Italy
| | | | - Carmela Irene
- Department of Cellular, Computational and Integrative Biology (CIBIO) Laboratory of Synthetic and Structural Vaccinology University of Trento Trento Italy
| | - Francesca Zerbini
- Department of Cellular, Computational and Integrative Biology (CIBIO) Laboratory of Synthetic and Structural Vaccinology University of Trento Trento Italy
| | - Elena Caproni
- Department of Cellular, Computational and Integrative Biology (CIBIO) Laboratory of Synthetic and Structural Vaccinology University of Trento Trento Italy
| | - Samine J Isaac
- Department of Cellular, Computational and Integrative Biology (CIBIO) Laboratory of Synthetic and Structural Vaccinology University of Trento Trento Italy
| | - Martina Grigolato
- Department of Cellular, Computational and Integrative Biology (CIBIO) Laboratory of Synthetic and Structural Vaccinology University of Trento Trento Italy
| | - Riccardo Corbellari
- Department of Cellular, Computational and Integrative Biology (CIBIO) Laboratory of Synthetic and Structural Vaccinology University of Trento Trento Italy
| | | | | | | | - Luca Bini
- Department of Life Sciences Functional Proteomics Laboratories University of Siena Siena Italy
| | - Yaqoub Ashhab
- Palestine-Korea Biotechnology Center Palestine Polytechnic University Hebron Palestine
| | - Alberto Grandi
- Toscana Life Sciences Foundation Siena Italy.,BiOMViS Srl Siena Italy
| | - Guido Grandi
- Department of Cellular, Computational and Integrative Biology (CIBIO) Laboratory of Synthetic and Structural Vaccinology University of Trento Trento Italy
| |
Collapse
|
41
|
Mancini F, Gasperini G, Rossi O, Aruta MG, Raso MM, Alfini R, Biagini M, Necchi F, Micoli F. Dissecting the contribution of O-Antigen and proteins to the immunogenicity of Shigella sonnei generalized modules for membrane antigens (GMMA). Sci Rep 2021; 11:906. [PMID: 33441861 PMCID: PMC7806729 DOI: 10.1038/s41598-020-80421-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 12/11/2020] [Indexed: 01/11/2023] Open
Abstract
GMMA are exosomes released from engineered Gram-negative bacteria resembling the composition of outer membranes. We applied the GMMA technology for the development of an O-Antigen (OAg) based vaccine against Shigella sonnei, the most epidemiologically relevant cause of shigellosis. S. sonnei OAg has been identified as a key antigen for protective immunity, and GMMA are able to induce anti-OAg-specific IgG response in animal models and healthy adults. The contribution of protein-specific antibodies induced upon vaccination with GMMA has never been fully elucidated. Anti-protein antibodies are induced in mice upon immunization with either OAg-negative and OAg-positive GMMA. Here we demonstrated that OAg chains shield the bacteria from anti-protein antibody binding and therefore anti-OAg antibodies were the main drivers of bactericidal activity against OAg-positive bacteria. Interestingly, antibodies that are not targeting the OAg are functional against OAg-negative bacteria. The immunodominant protein antigens were identified by proteomic analysis. Our study confirms a critical role of the OAg on the immune response induced by S. sonnei GMMA. However, little is known about OAg length and density regulation during infection and, therefore, protein exposure. Hence, the presence of protein antigens on S. sonnei GMMA represents an added value for GMMA vaccines compared to other OAg-based formulations.
Collapse
Affiliation(s)
- Francesca Mancini
- 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
| | - Omar Rossi
- GSK Vaccines Institute for Global Health (GVGH), via Fiorentina 1, 53100, Siena, Italy
| | - Maria Grazia Aruta
- GSK Vaccines Institute for Global Health (GVGH), via Fiorentina 1, 53100, Siena, Italy
| | - Maria Michelina Raso
- 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
| | | | - Francesca Necchi
- 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
|
42
|
Micoli F, MacLennan CA. Outer membrane vesicle vaccines. Semin Immunol 2020; 50:101433. [PMID: 33309166 DOI: 10.1016/j.smim.2020.101433] [Citation(s) in RCA: 125] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 11/16/2020] [Accepted: 11/19/2020] [Indexed: 10/22/2022]
Abstract
Outer Membrane Vesicles (OMV) have received increased attention in recent years as a vaccine platform against bacterial pathogens. OMV from Neisseria meningitidis serogroup B have been extensively explored. Following the success of the MeNZB OMV vaccine in controlling an outbreak of N. meningitidis B in New Zealand, additional research and development resulted in the licensure of the OMV-containing four-component 4CMenB vaccine, Bexsero. This provided broader protection against multiple meningococcal B strains. Advances in the field of genetic engineering have permitted further improvements in the platform resulting in increased yields, reduced endotoxicity and decoration with homologous and heterologous antigens to enhance immuno genicity and provide broader protection. The OMV vaccine platform has been extended to many other pathogens. In this review, we discuss progress in the development of the OMV vaccine delivery platform, highlighting successful applications, together with potential challenges and gaps.
Collapse
Affiliation(s)
| | - Calman A MacLennan
- Bill & Melinda Gates Foundation, 62 Buckingham Gate, London, United Kingdom; Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
43
|
Antibodies and Protection in Systemic Salmonella Infections: Do We Still Have More Questions than Answers? Infect Immun 2020; 88:IAI.00219-20. [PMID: 32601109 DOI: 10.1128/iai.00219-20] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Salmonella causes grave systemic infections in humans and other animals and provides a paradigm for other diseases in which the bacteria have both intracellular and extracellular lifestyles. New generations of vaccines rely on the essential contribution of the antibody responses for their protection. The quality, antigen specificity, and functions associated with antibody responses to this pathogen have been elusive for a long time. Recent approaches that combine studies in humans and genetically manipulated experimental models and that exploit awareness of the location and within-host life cycle of the pathogen are shedding light on how humoral immunity to Salmonella operates. However, this area of research remains full of controversy and discrepancies. The overall scenario indicates that antibodies are essential for resistance against systemic Salmonella infections and can express the highest protective function when operating in conjunction with cell-mediated immunity. Antigen specificity, isotype profile, Fc-gamma receptor usage, and complement activation are all intertwined factors that still arcanely influence antibody-mediated protection to Salmonella.
Collapse
|
44
|
Micoli F, Alfini R, Di Benedetto R, Necchi F, Schiavo F, Mancini F, Carducci M, Palmieri E, Balocchi C, Gasperini G, Brunelli B, Costantino P, Adamo R, Piccioli D, Saul A. GMMA Is a Versatile Platform to Design Effective Multivalent Combination Vaccines. Vaccines (Basel) 2020; 8:E540. [PMID: 32957610 PMCID: PMC7564227 DOI: 10.3390/vaccines8030540] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/12/2020] [Accepted: 09/14/2020] [Indexed: 01/21/2023] Open
Abstract
Technology platforms are an important strategy to facilitate the design, development and implementation of vaccines to combat high-burden diseases that are still a threat for human populations, especially in low- and middle-income countries, and to address the increasing number and global distribution of pathogens resistant to antimicrobial drugs. Generalized Modules for Membrane Antigens (GMMA), outer membrane vesicles derived from engineered Gram-negative bacteria, represent an attractive technology to design affordable vaccines. Here, we show that GMMA, decorated with heterologous polysaccharide or protein antigens, leads to a strong and effective antigen-specific humoral immune response in mice. Importantly, GMMA promote enhanced immunogenicity compared to traditional formulations (e.g., recombinant proteins and glycoconjugate vaccines), without negative impact to the anti-GMMA immune response. Our findings support the use of GMMA as a "plug and play" technology for the development of effective combination vaccines targeting different bugs at the same time.
Collapse
Affiliation(s)
- Francesca Micoli
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., 53100 Siena, Italy; (R.A.); (R.D.B.); (F.N.); (F.S.); (F.M.); (M.C.); (E.P.); (G.G.); (A.S.)
| | - Renzo Alfini
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., 53100 Siena, Italy; (R.A.); (R.D.B.); (F.N.); (F.S.); (F.M.); (M.C.); (E.P.); (G.G.); (A.S.)
| | - Roberta Di Benedetto
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., 53100 Siena, Italy; (R.A.); (R.D.B.); (F.N.); (F.S.); (F.M.); (M.C.); (E.P.); (G.G.); (A.S.)
| | - Francesca Necchi
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., 53100 Siena, Italy; (R.A.); (R.D.B.); (F.N.); (F.S.); (F.M.); (M.C.); (E.P.); (G.G.); (A.S.)
| | - Fabiola Schiavo
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., 53100 Siena, Italy; (R.A.); (R.D.B.); (F.N.); (F.S.); (F.M.); (M.C.); (E.P.); (G.G.); (A.S.)
| | - Francesca Mancini
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., 53100 Siena, Italy; (R.A.); (R.D.B.); (F.N.); (F.S.); (F.M.); (M.C.); (E.P.); (G.G.); (A.S.)
| | - Martina Carducci
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., 53100 Siena, Italy; (R.A.); (R.D.B.); (F.N.); (F.S.); (F.M.); (M.C.); (E.P.); (G.G.); (A.S.)
| | - Elena Palmieri
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., 53100 Siena, Italy; (R.A.); (R.D.B.); (F.N.); (F.S.); (F.M.); (M.C.); (E.P.); (G.G.); (A.S.)
| | | | - Gianmarco Gasperini
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., 53100 Siena, Italy; (R.A.); (R.D.B.); (F.N.); (F.S.); (F.M.); (M.C.); (E.P.); (G.G.); (A.S.)
| | | | | | - Roberto Adamo
- GSK, 53100 Siena, Italy; (C.B.); (B.B.); (P.C.); (R.A.); (D.P.)
| | - Diego Piccioli
- GSK, 53100 Siena, Italy; (C.B.); (B.B.); (P.C.); (R.A.); (D.P.)
| | - Allan Saul
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., 53100 Siena, Italy; (R.A.); (R.D.B.); (F.N.); (F.S.); (F.M.); (M.C.); (E.P.); (G.G.); (A.S.)
| |
Collapse
|
45
|
Rossi O, Aruta MG, Acquaviva A, Mancini F, Micoli F, Necchi F. Characterization of Competitive ELISA and Formulated Alhydrogel Competitive ELISA (FAcE) for Direct Quantification of Active Ingredients in GMMA-Based Vaccines. Methods Protoc 2020; 3:mps3030062. [PMID: 32878036 PMCID: PMC7563494 DOI: 10.3390/mps3030062] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 08/27/2020] [Accepted: 08/28/2020] [Indexed: 11/17/2022] Open
Abstract
Generalized modules for membrane antigens (GMMA) represent a technology particularly attractive for designing affordable vaccines against Gram-negative bacteria. We explored such technology for the development of O-antigen-based vaccines against Shigella and nontyphoidal Salmonella. Adsorption of GMMA on Alhydrogel was required for abrogation of pyrogenicity in rabbits, and Shigella sonnei GMMA on Alhydrogel was well tolerated and immunogenic in humans. Quantification of key antigens in formulated vaccines was fundamental for release and to check stability overtime. Traditionally, the direct quantification of antigens adsorbed on aluminum salts has been challenging, and the quantification of each active ingredient in multicomponent formulated vaccines has been even more complicated. To directly quantify each active ingredient and unbound drug substances in formulated vaccines, we developed the Formulated Alhydrogel competitive ELISA (FAcE) and the competitive ELISA method, respectively. The methods were both fully characterized, assessing specificity, repeatability, intermediate precision, and accuracy, for S. sonnei OAg quantification, both in a single component or multicomponent GMMA formulation also containing S. flexneri GMMA. The developed immunological methods allowed us to fully characterize Shigella GMMA drug products, supporting their preclinical and clinical development. The same methods, already extended to GMMA from nontyphoidal Salmonella and Neisseria meningitidis, could be potentially extended to any antigen formulated on Alhydrogel.
Collapse
|
46
|
Baliban SM, Lu YJ, Malley R. Overview of the Nontyphoidal and Paratyphoidal Salmonella Vaccine Pipeline: Current Status and Future Prospects. Clin Infect Dis 2020; 71:S151-S154. [PMID: 32725233 PMCID: PMC7388718 DOI: 10.1093/cid/ciaa514] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Nontyphoidal Salmonella and Salmonella Paratyphi are responsible for significant morbidity and mortality worldwide. To date, no vaccine has been licensed against these organisms. The development of effective vaccines remains an urgent priority. In this review, the rationale for and current status of various vaccine candidates against S. Paratyphi and nontyphoidal Salmonella are presented, with a focus on the research findings from the 2019 International Conference on Typhoid and Other Invasive Salmonelloses. Additionally, other vaccine candidates that are currently undergoing clinical development are highlighted. Future approaches, which may include antigens that are genetically conserved across Salmonella and confer broad, non-serotype-specific protection, are also discussed.
Collapse
Affiliation(s)
- Scott M Baliban
- Center for Vaccine Development and Global Health, University of Maryland, School of Medicine, Baltimore, Maryland, USA
| | - Ying-Jie Lu
- Division of Infectious Diseases, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Richard Malley
- Division of Infectious Diseases, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
47
|
Rossi O, Citiulo F, Mancini F. Outer membrane vesicles: moving within the intricate labyrinth of assays that can predict risks of reactogenicity in humans. Hum Vaccin Immunother 2020; 17:601-613. [PMID: 32687736 PMCID: PMC7899674 DOI: 10.1080/21645515.2020.1780092] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Outer membrane vesicles (OMV) are exosomes naturally released from the surface of Gram-negative bacteria. Since the ’80s, OMVs have been proposed as powerful vaccine platforms due to their intrinsic self-adjuvanticity and ability to present multiple antigens in natural conformation. However, the presence of several pathogen-associated molecular patterns (PAMPs), especially lipid A, has raised concerns about potential systemic reactogenicity in humans. Recently, chemical and genetic approaches allowed to efficiently modulate the balance between reactogenicity and immunogenicity for the use of OMV in humans. Several assays (monocyte activation test, rabbit pyrogenicity test, limulus amebocyte lysate, human transfectant cells, and toxicology studies) were developed to test, with highly predictive potential, the risk of reactogenicity in humans before moving to clinical use. In this review, we provide a historical perspective on how different assays were and can be used to successfully evaluate systemic reactogenicity during clinical development and after licensure.
Collapse
Affiliation(s)
- Omar Rossi
- GSK Vaccines Institute for Global Health S.r.l (GVGH) , Siena, Italy
| | - Francesco Citiulo
- GSK Vaccines Institute for Global Health S.r.l (GVGH) , Siena, Italy
| | - Francesca Mancini
- GSK Vaccines Institute for Global Health S.r.l (GVGH) , Siena, Italy
| |
Collapse
|
48
|
OMV Vaccines and the Role of TLR Agonists in Immune Response. Int J Mol Sci 2020; 21:ijms21124416. [PMID: 32575921 PMCID: PMC7352230 DOI: 10.3390/ijms21124416] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 06/17/2020] [Accepted: 06/18/2020] [Indexed: 12/26/2022] Open
Abstract
Outer Membrane Vesicles (OMVs) are bacterial nanoparticles that are spontaneously released during growth both in vitro and in vivo by Gram-negative bacteria. They are spherical, bilayered membrane nanostructures that contain many components found within the external surface of the parent bacterium. Naturally, OMVs serve the bacteria as a mechanism to deliver DNA, RNA, proteins, and toxins, as well as to promote biofilm formation and remodel the outer membrane during growth. On the other hand, as OMVs possess the optimal size to be uptaken by immune cells, and present a range of surface-exposed antigens in native conformation and Toll-like receptor (TLR) activating components, they represent an attractive and powerful vaccine platform able to induce both humoral and cell-mediated immune responses. This work reviews the TLR-agonists expressed on OMVs and their capability to trigger individual TLRs expressed on different cell types of the immune system, and then focuses on their impact on the immune responses elicited by OMVs compared to traditional vaccines.
Collapse
|
49
|
Intra-Laboratory Evaluation of Luminescence Based High-Throughput Serum Bactericidal Assay (L-SBA) to Determine Bactericidal Activity of Human Sera against Shigella. High Throughput 2020; 9:ht9020014. [PMID: 32521658 PMCID: PMC7361673 DOI: 10.3390/ht9020014] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 06/01/2020] [Accepted: 06/04/2020] [Indexed: 12/31/2022] Open
Abstract
Despite the huge decrease in deaths caused by Shigella worldwide in recent decades, shigellosis still causes over 200,000 deaths every year. No vaccine is currently available, and the morbidity of the disease coupled with the rise of antimicrobial resistance renders the introduction of an effective vaccine extremely urgent. Although a clear immune correlate of protection against shigellosis has not yet been established, the demonstration of the bactericidal activity of antibodies induced upon vaccination may provide one means of the functionality of antibodies induced in protecting against Shigella. The method of choice to evaluate the complement-mediated functional activity of vaccine-induced antibodies is the Serum Bactericidal Assay (SBA). Here we present the development and intra-laboratory characterization of a high-throughput luminescence-based SBA (L-SBA) method, based on the detection of ATP as a proxy of surviving bacteria, to evaluate the complement-mediated killing of human sera. We demonstrated the high specificity of the assay against a homologous strain without any heterologous aspecificity detected against species-related and non-species-related strains. We assessed the linearity, repeatability and reproducibility of L-SBA on human sera. This work will guide the bactericidal activity assessment of clinical sera raised against S. sonnei. The method has the potential of being applicable with similar performances to determine the bactericidal activity of any non-clinical and clinical sera that rely on complement-mediated killing.
Collapse
|
50
|
Berti F, Micoli F. Improving efficacy of glycoconjugate vaccines: from chemical conjugates to next generation constructs. Curr Opin Immunol 2020; 65:42-49. [PMID: 32361591 DOI: 10.1016/j.coi.2020.03.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/24/2020] [Accepted: 03/23/2020] [Indexed: 01/10/2023]
Abstract
Glycoconjugate vaccines are among the safest and most successful vaccines developed during the past 30 years. Since the first semisynthetic chemical conjugate vaccine licensed in the 1980's to protect human against Haemophilus influenzae type b infection, conjugate vaccines against Neisseria meningitidis and Streptococcus pneumoniae have been developed and registered using the same approach (i.e. bacterial growth to produce capsular polysaccharide antigen and chemical coupling to carrier protein). Other types of conjugate vaccines have been recently developed and tested in clinical trials, prepared by coupling chemically synthesized oligosaccharides to proteins, by engineering Escherichia coli to directly produce bioconjugates or by delivering the native carbohydrate antigen in engineered membrane vesicles (i.e. Generalized Modules for Membrane Antigens, GMMA). Through this review, the reader will have an insight regarding the history and the characteristics of different types of conjugate vaccines, and the attributes that might affect their immunogenicity and their potential for future applications.
Collapse
|