1
|
Martins Y, Cerqueira e Costa MO, Palumbo MC, F. Do Porto D, Custódio FL, Trevizani R, Nicolás MF. PAPreC: A Pipeline for Antigenicity Prediction Comparison Methods across Bacteria. ACS OMEGA 2025; 10:5415-5429. [PMID: 39989760 PMCID: PMC11840615 DOI: 10.1021/acsomega.4c07147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 01/16/2025] [Accepted: 01/23/2025] [Indexed: 02/25/2025]
Abstract
Antigenicity prediction plays a crucial role in vaccine development, antibody-based therapies, and diagnostic assays, as this predictive approach helps assess the potential of molecular structures to induce and recruit immune cells and drive antibody production. Several existing prediction methods, which target complete proteins and epitopes identified through reverse vaccinology, face limitations regarding input data constraints, feature extraction strategies, and insufficient flexibility for model evaluation and interpretation. This work presents PAPreC (Pipeline for Antigenicity Prediction Comparison), an open-source, versatile workflow (available at https://github.com/YasCoMa/paprec_nx_workflow) designed to address these challenges. PAPreC systematically examines three key factors: the selection of training data sets, feature extraction methods (including physicochemical descriptors and ESM-2 encoder-derived embeddings), and diverse classifiers. It provides automated model evaluation, interpretability through SHapley Additive exPlanations (SHAP) analysis, and applicability domain assessments, enabling researchers to identify optimal configurations for their specific data sets. Applying PAPreC to IEDB data as a reference, we demonstrate its effectiveness across the ESKAPE pathogen group. A case study involving Pseudomonas aeruginosa and Staphylococcus aureus shows that specific feature configurations are more suitable for different sequence types, and that ESM-2 embeddings enhance model performance. Moreover, our results indicate that separate models for Gram-positive and Gram-negative bacteria are not required. PAPreC offers a comprehensive, adaptable, and robust framework to streamline and improve antigenicity prediction for diverse bacterial data sets.
Collapse
Affiliation(s)
- Yasmmin
C. Martins
- Bioinformatics
Laboratory, National Laboratory for Scientific
Computing, Av. Getúlio Vargas 333, 25651-075 Petrópolis, Brazil
- Department
of Biological Chemistry, Faculty of Exact and Natural Sciences, University of Buenos Aires - UBA, Av. Int. Cantilo, C1428 Buenos Aires, Argentina
| | - Maiana O. Cerqueira e Costa
- Bioinformatics
Laboratory, National Laboratory for Scientific
Computing, Av. Getúlio Vargas 333, 25651-075 Petrópolis, Brazil
| | - Miranda C. Palumbo
- Department
of Biological Chemistry, Faculty of Exact and Natural Sciences, University of Buenos Aires - UBA, Av. Int. Cantilo, C1428 Buenos Aires, Argentina
| | - Dario F. Do Porto
- Department
of Biological Chemistry, Faculty of Exact and Natural Sciences, University of Buenos Aires - UBA, Av. Int. Cantilo, C1428 Buenos Aires, Argentina
| | - Fábio L. Custódio
- Department
of Computational Mechanics, National Laboratory
for Scientific Computing, Av. Getúlio Vargas 333, 25651-075 Petrópolis, Brazil
| | - Raphael Trevizani
- Biotechnology, Oswaldo Cruz Foundation
- Fiocruz, Street São
José S/N, 61760-000 Eusébio, Brazil
| | - Marisa Fabiana Nicolás
- Bioinformatics
Laboratory, National Laboratory for Scientific
Computing, Av. Getúlio Vargas 333, 25651-075 Petrópolis, Brazil
| |
Collapse
|
2
|
Solyman SM, Kamal SA, Hanora AS. Protection of Mice Vaccinated with a New B Cell and T Cell Epitopes Cocktail from Staphylococcus aureus Challenge in Skin Infection Model. Curr Microbiol 2025; 82:128. [PMID: 39922982 DOI: 10.1007/s00284-025-04102-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 01/23/2025] [Indexed: 02/10/2025]
Abstract
Developing an effective vaccine against Staphylococcus aureus (S. aureus) is a key global health concern, especially with the increased reports of multidrug-resistant (MDR) S. aureus strains. Previous attempts for S. aureus vaccine development were unsuccessful. In this study, Manganese transport protein C (MABC) B cell epitopes, Nickel ABC transporter (NABC) B cell & T cell epitopes, and Phosphatidylinositol phosphodiesterase (PIc) B cell & T cell epitopes were used as a vaccine in mice skin infection model. Mice immunized with peptide mixture and MABC peptide group showed the best skin lesion healing results. The protection level was correlated with the highest IgG level, highest levels of interferon-gamma (INF γ), and lowest levels of interleukin-2 (IL-2). The peptide mixture group also showed the highest count of CD4/ CD8 cells. Results demonstrated that the inclusion of B cell and T cell epitopes of multiple genes improved both the humoral and cellular immunity and resulted in the best outcome in the skin infection mice model. A more expanded in-vivo study in different mice models is recommended for testing MABC, NABC, and PIc B cells and T cells peptides cocktail as promising S. aureus vaccine.
Collapse
Affiliation(s)
- Samar M Solyman
- Department of Microbiology & Immunology, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt.
- Department of Microbiology & Immunology, Faculty of Pharmacy, Sinai University, Elkantara Branch, Ismailia, Egypt.
| | - Shymaa A Kamal
- Department of Microbiology & Immunology, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt
| | - Amro S Hanora
- Department of Microbiology & Immunology, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt
- Department of Microbiology & Immunology, Faculty of Pharmacy, King Salman International University, Ras Sudr, Egypt
| |
Collapse
|
3
|
Das Mitra S, Kumar B, Rajegowda S, Bandopadhyay S, Karunakar P, Pais R. Reverse vaccinology & immunoinformatics approach to design a multiepitope vaccine (CV3Ag-antiMRSA) against methicillin resistant Staphylococcus aureus (MRSA) - a pathogen affecting both human and animal health. J Biomol Struct Dyn 2023; 42:11792-11811. [PMID: 37798927 DOI: 10.1080/07391102.2023.2265471] [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: 02/27/2023] [Accepted: 09/24/2023] [Indexed: 10/07/2023]
Abstract
Infections caused by drug resistant bacteria is a silent detrimental pandemic affecting the global health care profoundly. Methicillin resistant Staphylococcus aureus (MRSA) is a pathogen that causes serious infections in different settings (community, hospital & veterinary) whose treatment remains highly challenging due to its powerful characteristics (antibiotic resistance strategies, virulence factors). In this study, we used reverse vaccinology (RV) approach and designed an immunogenic multi epitope vaccine (CV3Ag-antiMRSA) targeting three potential antigen candidates viz., mecA encoding transpeptidase (PBP2a) protein responsible for conferring methicillin resistance and two virulence determinants - hlgA encoding gamma-hemolysin component A (a pore forming toxin) and isdB encoding iron regulated surface determinant B (heme transport component that allows S. aureus to scavenge iron from host hemoglobin and myoglobin). We employed an array of immunoinformatic tools/server to identify and use immunogenic epitopes (B cell and MHC class) to develop the chimeric subunit vaccine V4 (CV3Ag-antiMRSA) with immune modulating adjuvant and linkers. Based on different parameters, the vaccine construct V4 (CV3Ag-antiMRSA) was determined to be suitable vaccine (antigenic and non-allergen). Molecular docking and simulation of CV3Ag-antiMRSA with Toll Like Receptor (TLR2) predicted its immuno-stimulating potential. Finally, in silico cloning of CV3Ag-antiMRSA construct into pet28a and pet30 vector displayed its feasibility for the heterologous expression in the E. coli expression system. This vaccine candidate (CV3Ag-antiMRSA) designed based on the MRSA genomes obtained from both animal and human hosts can be experimentally validated and thereby contribute to vaccine development to impart protection to both animal and human health.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Susweta Das Mitra
- Department of Biotechnology, School of Basic & Applied Sciences, Dayananda Sagar University, Bangalore, Karnataka, India
| | - Bharat Kumar
- Department of Biotechnology, School of Basic & Applied Sciences, Dayananda Sagar University, Bangalore, Karnataka, India
| | - Sushmitha Rajegowda
- Department of Biotechnology, School of Basic & Applied Sciences, Dayananda Sagar University, Bangalore, Karnataka, India
| | - Satarupa Bandopadhyay
- Department of Biotechnology, School of Basic & Applied Sciences, Dayananda Sagar University, Bangalore, Karnataka, India
| | - Prashantha Karunakar
- Department of Biotechnology, Dayananda Sagar College of Engineering (Affiliated to Visvesvaraya Technological University, Belagavi), Bangalore, Karnataka, India
| | - Roshan Pais
- Department of Biotechnology, School of Basic & Applied Sciences, Dayananda Sagar University, Bangalore, Karnataka, India
| |
Collapse
|
4
|
Bear A, Locke T, Rowland-Jones S, Pecetta S, Bagnoli F, Darton TC. The immune evasion roles of Staphylococcus aureus protein A and impact on vaccine development. Front Cell Infect Microbiol 2023; 13:1242702. [PMID: 37829608 PMCID: PMC10565657 DOI: 10.3389/fcimb.2023.1242702] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 09/08/2023] [Indexed: 10/14/2023] Open
Abstract
While Staphylococcus aureus (S. aureus) bacteria are part of the human commensal flora, opportunistic invasion following breach of the epithelial layers can lead to a wide array of infection syndromes at both local and distant sites. Despite ubiquitous exposure from early infancy, the life-long risk of opportunistic infection is facilitated by a broad repertoire of S. aureus virulence proteins. These proteins play a key role in inhibiting development of a long-term protective immune response by mechanisms ranging from dysregulation of the complement cascade to the disruption of leukocyte migration. In this review we describe the recent progress made in dissecting S. aureus immune evasion, focusing on the role of the superantigen, staphylococcal protein A (SpA). Evasion of the normal human immune response drives the ability of S. aureus to cause infection, often recurrently, and is also thought to be a major hindrance in the development of effective vaccination strategies. Understanding the role of S. aureus virulence protein and determining methods overcoming or subverting these mechanisms could lead to much-needed breakthroughs in vaccine and monoclonal antibody development.
Collapse
Affiliation(s)
- Alex Bear
- Department of Infection, Immunity and Cardiovascular Disease, The University of Sheffield, Sheffield, United Kingdom
| | - Thomas Locke
- Department of Infection, Immunity and Cardiovascular Disease, The University of Sheffield, Sheffield, United Kingdom
| | - Sarah Rowland-Jones
- Department of Infection, Immunity and Cardiovascular Disease, The University of Sheffield, Sheffield, United Kingdom
| | | | | | - Thomas C. Darton
- Department of Infection, Immunity and Cardiovascular Disease, The University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
5
|
Khalid K, Poh CL. The Promising Potential of Reverse Vaccinology-Based Next-Generation Vaccine Development over Conventional Vaccines against Antibiotic-Resistant Bacteria. Vaccines (Basel) 2023; 11:1264. [PMID: 37515079 PMCID: PMC10385262 DOI: 10.3390/vaccines11071264] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 07/14/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
The clinical use of antibiotics has led to the emergence of multidrug-resistant (MDR) bacteria, leading to the current antibiotic resistance crisis. To address this issue, next-generation vaccines are being developed to prevent antimicrobial resistance caused by MDR bacteria. Traditional vaccine platforms, such as inactivated vaccines (IVs) and live attenuated vaccines (LAVs), were effective in preventing bacterial infections. However, they have shown reduced efficacy against emerging antibiotic-resistant bacteria, including MDR M. tuberculosis. Additionally, the large-scale production of LAVs and IVs requires the growth of live pathogenic microorganisms. A more promising approach for the accelerated development of vaccines against antibiotic-resistant bacteria involves the use of in silico immunoinformatics techniques and reverse vaccinology. The bioinformatics approach can identify highly conserved antigenic targets capable of providing broader protection against emerging drug-resistant bacteria. Multi-epitope vaccines, such as recombinant protein-, DNA-, or mRNA-based vaccines, which incorporate several antigenic targets, offer the potential for accelerated development timelines. This review evaluates the potential of next-generation vaccine development based on the reverse vaccinology approach and highlights the development of safe and immunogenic vaccines through relevant examples from successful preclinical and clinical studies.
Collapse
Affiliation(s)
- Kanwal Khalid
- Centre for Virus and Vaccine Research, School of Medical and Life Sciences, Sunway University, Bandar Sunway, Subang Jaya 47500, Malaysia
| | - Chit Laa Poh
- Centre for Virus and Vaccine Research, School of Medical and Life Sciences, Sunway University, Bandar Sunway, Subang Jaya 47500, Malaysia
| |
Collapse
|
6
|
Haag AF, Liljeroos L, Donato P, Pozzi C, Brignoli T, Bottomley MJ, Bagnoli F, Delany I. In Vivo Gene Expression Profiling of Staphylococcus aureus during Infection Informs Design of Stemless Leukocidins LukE and -D as Detoxified Vaccine Candidates. Microbiol Spectr 2023; 11:e0257422. [PMID: 36688711 PMCID: PMC9927290 DOI: 10.1128/spectrum.02574-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 01/02/2023] [Indexed: 01/24/2023] Open
Abstract
Staphylococcus aureus is a clinically important bacterial pathogen that has become resistant to treatment with most routinely used antibiotics. Alternative strategies, such as vaccination and phage therapy, are therefore actively being investigated to prevent or combat staphylococcal infections. Vaccination requires that vaccine targets are expressed at sufficient quantities during infection so that they can be targeted by the host's immune system. While our knowledge of in vitro expression levels of putative vaccine candidates is comprehensive, crucial in vivo expression data are scarce and promising vaccine candidates during in vitro assessment often prove ineffective in preventing S. aureus infection. Here, we show how a newly developed high-throughput quantitative reverse transcription-PCR (qRT-PCR) assay monitoring the expression of 84 staphylococcal genes encoding mostly virulence factors can inform the selection and design of effective vaccine candidates against staphylococcal infections. We show that this assay can accurately quantify mRNA expression levels of these genes in several host organs relying only on very limited amounts of bacterial mRNA in each sample. We selected two highly expressed genes, lukE and lukD, encoding pore-forming leukotoxins, to inform the design of detoxified recombinant proteins and showed that immunization with recombinant genetically detoxified LukED antigens conferred protection against staphylococcal skin infection in mice. Consequently, knowledge of in vivo-expressed virulence determinants can be successfully deployed to identify and select promising candidates for optimized design of effective vaccine antigens against S. aureus. Notably, this approach should be broadly applicable to numerous other pathogens. IMPORTANCE Vaccination is an attractive strategy for preventing bacterial infections in an age of increased antimicrobial resistance. However, vaccine development frequently suffers significant setbacks when candidate antigens that show promising results in in vitro experimentation fail to protect from disease. An alluring strategy is to focus resources on developing bacterial virulence factors that are expressed during disease establishment or maintenance and are critical for bacterial in-host survival as vaccine targets. While expression profiles of many virulence factors have been characterized in detail in vitro, our knowledge of their in vivo expression profiles is still scarce. Here, using a high-throughput qRT-PCR approach, we identified two highly expressed leukotoxins in a murine infection model and showed that genetically detoxified derivatives of these elicited a protective immune response in a murine skin infection model. Therefore, in vivo gene expression can inform the selection of promising candidates for the design of effective vaccine antigens.
Collapse
Affiliation(s)
- Andreas F. Haag
- GSK, Siena, Italy
- School of Medicine, University of St. Andrews, St. Andrews, United Kingdom
| | | | | | | | - Tarcisio Brignoli
- GSK, Siena, Italy
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | | | | | | |
Collapse
|
7
|
Shoaib M, Aqib AI, Muzammil I, Majeed N, Bhutta ZA, Kulyar MFEA, Fatima M, Zaheer CNF, Muneer A, Murtaza M, Kashif M, Shafqat F, Pu W. MRSA compendium of epidemiology, transmission, pathophysiology, treatment, and prevention within one health framework. Front Microbiol 2023; 13:1067284. [PMID: 36704547 PMCID: PMC9871788 DOI: 10.3389/fmicb.2022.1067284] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 12/19/2022] [Indexed: 01/12/2023] Open
Abstract
Staphylococcus aureus is recognized as commensal as well as opportunistic pathogen of humans and animals. Methicillin resistant strain of S. aureus (MRSA) has emerged as a major pathogen in hospitals, community and veterinary settings that compromises the public health and livestock production. MRSA basically emerged from MSSA after acquiring SCCmec element through gene transfer containing mecA gene responsible for encoding PBP-2α. This protein renders the MRSA resistant to most of the β-lactam antibiotics. Due to the continuous increasing prevalence and transmission of MRSA in hospitals, community and veterinary settings posing a major threat to public health. Furthermore, high pathogenicity of MRSA due to a number of virulence factors produced by S. aureus along with antibiotic resistance help to breach the immunity of host and responsible for causing severe infections in humans and animals. The clinical manifestations of MRSA consist of skin and soft tissues infection to bacteremia, septicemia, toxic shock, and scalded skin syndrome. Moreover, due to the increasing resistance of MRSA to number of antibiotics, there is need to approach alternatives ways to overcome economic as well as human losses. This review is going to discuss various aspects of MRSA starting from emergence, transmission, epidemiology, pathophysiology, disease patterns in hosts, novel treatment, and control strategies.
Collapse
Affiliation(s)
- Muhammad Shoaib
- Key Laboratory of New Animal Drug Project, Gansu Province/Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs/Lanzhou Institute of Husbandry and Pharmaceutical Sciences of the Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Amjad Islam Aqib
- Department of Medicine, Cholistan University of Veterinary and Animal Sciences, Bahawalpur, Pakistan
| | - Iqra Muzammil
- Department of Medicine, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Noreen Majeed
- Institute of Microbiology, University of Agriculture, Faisalabad, Pakistan
| | - Zeeshan Ahmad Bhutta
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | | | - Mahreen Fatima
- Faculty of Biosciences, Cholistan University of Veterinary and Animal Sciences, Bahawalpur, Pakistan
| | | | - Afshan Muneer
- Department of Zoology, Cholistan University of Veterinary and Animal Sciences, Bahawalpur, Pakistan
| | - Maheen Murtaza
- Department of Zoology, Cholistan University of Veterinary and Animal Sciences, Bahawalpur, Pakistan
| | - Muhammad Kashif
- Department of Microbiology, Cholistan University of Veterinary and Animal Sciences, Bahawalpur, Pakistan
| | - Furqan Shafqat
- Department of Microbiology, Cholistan University of Veterinary and Animal Sciences, Bahawalpur, Pakistan
| | - Wanxia Pu
- Key Laboratory of New Animal Drug Project, Gansu Province/Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs/Lanzhou Institute of Husbandry and Pharmaceutical Sciences of the Chinese Academy of Agricultural Sciences, Lanzhou, China
| |
Collapse
|
8
|
Jahantigh HR, Faezi S, Habibi M, Mahdavi M, Stufano A, Lovreglio P, Ahmadi K. The Candidate Antigens to Achieving an Effective Vaccine against Staphylococcus aureus. Vaccines (Basel) 2022; 10:vaccines10020199. [PMID: 35214658 PMCID: PMC8876328 DOI: 10.3390/vaccines10020199] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 01/20/2022] [Accepted: 01/26/2022] [Indexed: 12/11/2022] Open
Abstract
Staphylococcus aureus (S. aureus) is an opportunistic pathogen that causes various inflammatory local infections, from those of the skin to postinfectious glomerulonephritis. These infections could result in serious threats, putting the life of the patient in danger. Antibiotic-resistant S. aureus could lead to dramatic increases in human mortality. Antibiotic resistance would explicate the failure of current antibiotic therapies. So, it is obvious that an effective vaccine against S. aureus infections would significantly reduce costs related to care in hospitals. Bacterial vaccines have important impacts on morbidity and mortality caused by several common pathogens, however, a prophylactic vaccine against staphylococci has not yet been produced. During the last decades, the efforts to develop an S. aureus vaccine have faced two major failures in clinical trials. New strategies for vaccine development against S. aureus has supported the use of multiple antigens, the inclusion of adjuvants, and the focus on various virulence mechanisms. We aimed to present a compressive review of different antigens of S. aureus and also to introduce vaccine candidates undergoing clinical trials, from which can help us to choose a suitable and effective candidate for vaccine development against S. aureus.
Collapse
Affiliation(s)
- Hamid Reza Jahantigh
- Animal Health and Zoonosis, Department of Veterinary Medicine, University of Bari, 70010 Bari, Italy;
- Interdisciplinary Department of Medicine, Section of Occupational Medicine, University of Bari, 70010 Bari, Italy;
- Correspondence: (H.R.J.); (K.A.); Tel.: +39-3773827669 (H.R.J.)
| | - Sobhan Faezi
- Medical Biotechnology Research Center, School of Paramedicine, Guilan University of Medical Sciences, Rasht 41937, Iran;
| | - Mehri Habibi
- Department of Molecular Biology, Pasteur Institute of Iran, Pasteur Ave., Tehran 13164, Iran;
| | - Mehdi Mahdavi
- Advanced Therapy Medicinal Product (ATMP) Department, Breast Cancer Research Center, Motamed Cancer Institute, Academic Center for Education, Culture and Research (ACECR), Tehran 1517964311, Iran
- Recombinant Vaccine Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 13164, Iran;
| | - Angela Stufano
- Animal Health and Zoonosis, Department of Veterinary Medicine, University of Bari, 70010 Bari, Italy;
- Interdisciplinary Department of Medicine, Section of Occupational Medicine, University of Bari, 70010 Bari, Italy;
| | - Piero Lovreglio
- Interdisciplinary Department of Medicine, Section of Occupational Medicine, University of Bari, 70010 Bari, Italy;
| | - Khadijeh Ahmadi
- Infectious and Tropical Diseases Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas 79391, Iran
- Correspondence: (H.R.J.); (K.A.); Tel.: +39-3773827669 (H.R.J.)
| |
Collapse
|
9
|
Michalik M, Djahanschiri B, Leo JC, Linke D. An Update on "Reverse Vaccinology": The Pathway from Genomes and Epitope Predictions to Tailored, Recombinant Vaccines. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2412:45-71. [PMID: 34918241 DOI: 10.1007/978-1-0716-1892-9_4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
In this chapter, we review the computational approaches that have led to a new generation of vaccines in recent years. There are many alternative routes to develop vaccines based on the concept of reverse vaccinology. They all follow the same basic principles-mining available genome and proteome information for antigen candidates, and recombinantly expressing them for vaccine production. Some of the same principles have been used successfully for cancer therapy approaches. In this review, we focus on infectious diseases, describing the general workflow from bioinformatic predictions of antigens and epitopes down to examples where such predictions have been used successfully for vaccine development.
Collapse
Affiliation(s)
| | - Bardya Djahanschiri
- Institute of Cell Biology and Neuroscience, Goethe University, Frankfurt, Germany
| | - Jack C Leo
- Department of Biosciences, Nottingham Trent University, Nottingham, UK
| | - Dirk Linke
- Department of Biosciences, University of Oslo, Oslo, Norway.
| |
Collapse
|
10
|
König E, Gagliardi A, Riedmiller I, Andretta C, Tomasi M, Irene C, Frattini L, Zanella I, Berti F, Grandi A, Caproni E, Fantappiè L, Grandi G. Multi-Antigen Outer Membrane Vesicle Engineering to Develop Polyvalent Vaccines: The Staphylococcus aureus Case. Front Immunol 2021; 12:752168. [PMID: 34819933 PMCID: PMC8606680 DOI: 10.3389/fimmu.2021.752168] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 10/14/2021] [Indexed: 11/13/2022] Open
Abstract
Modification of surface antigens and differential expression of virulence factors are frequent strategies pathogens adopt to escape the host immune system. These escape mechanisms make pathogens a "moving target" for our immune system and represent a challenge for the development of vaccines, which require more than one antigen to be efficacious. Therefore, the availability of strategies, which simplify vaccine design, is highly desirable. Bacterial Outer Membrane Vesicles (OMVs) are a promising vaccine platform for their built-in adjuvanticity, ease of purification and flexibility to be engineered with foreign proteins. However, data on if and how OMVs can be engineered with multiple antigens is limited. In this work, we report a multi-antigen expression strategy based on the co-expression of two chimeras, each constituted by head-to-tail fusions of immunogenic proteins, in the same OMV-producing strain. We tested the strategy to develop a vaccine against Staphylococcus aureus, a Gram-positive human pathogen responsible for a large number of community and hospital-acquired diseases. Here we describe an OMV-based vaccine in which four S. aureus virulent factors, ClfAY338A, LukE, SpAKKAA and HlaH35L have been co-expressed in the same OMVs (CLSH-OMVsΔ60). The vaccine elicited antigen-specific antibodies with functional activity, as judged by their capacity to promote opsonophagocytosis and to inhibit Hla-mediated hemolysis, LukED-mediated leukocyte killing, and ClfA-mediated S. aureus binding to fibrinogen. Mice vaccinated with CLSH-OMVsΔ60 were robustly protected from S. aureus challenge in the skin, sepsis and kidney abscess models. This study not only describes a generalized approach to develop easy-to-produce and inexpensive multi-component vaccines, but also proposes a new tetravalent vaccine candidate ready to move to development.
Collapse
Affiliation(s)
- Enrico König
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | | | - Ilary Riedmiller
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Chiara Andretta
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Michele Tomasi
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Carmela Irene
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Luca Frattini
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Ilaria Zanella
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Francesco Berti
- Technical Research and Development, GlaxoSmithKline Vaccines, Siena, Italy
| | - Alberto Grandi
- ERC Vaccibiome Unit, Toscana Life Sciences Foundation, Siena, Italy.,Infectious Diseases and Cancer Immunotherapy Unit, BiOMViS Srl, Siena, Italy
| | - Elena Caproni
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Laura Fantappiè
- ERC Vaccibiome Unit, Toscana Life Sciences Foundation, Siena, Italy
| | - Guido Grandi
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| |
Collapse
|
11
|
Olatunji S, Bowen K, Huang CY, Weichert D, Singh W, Tikhonova IG, Scanlan EM, Olieric V, Caffrey M. Structural basis of the membrane intramolecular transacylase reaction responsible for lyso-form lipoprotein synthesis. Nat Commun 2021; 12:4254. [PMID: 34253723 PMCID: PMC8275575 DOI: 10.1038/s41467-021-24475-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 06/08/2021] [Indexed: 11/08/2022] Open
Abstract
Lipoproteins serve diverse functions in the bacterial cell and some are essential for survival. Some lipoproteins are adjuvants eliciting responses from the innate immune system of the host. The growing list of membrane enzymes responsible for lipoprotein synthesis includes the recently discovered lipoprotein intramolecular transacylase, Lit. Lit creates a lipoprotein that is less immunogenic, possibly enabling the bacteria to gain a foothold in the host by stealth. Here, we report the crystal structure of the Lit enzyme from Bacillus cereus and describe its mechanism of action. Lit consists of four transmembrane helices with an extracellular cap. Conserved residues map to the cap-membrane interface. They include two catalytic histidines that function to effect unimolecular transacylation. The reaction involves acyl transfer from the sn-2 position of the glyceryl moiety to the amino group on the N-terminal cysteine of the substrate via an 8-membered ring intermediate. Transacylation takes place in a confined aromatic residue-rich environment that likely evolved to bring distant moieties on the substrate into proximity and proper orientation for catalysis.
Collapse
Affiliation(s)
- Samir Olatunji
- Membrane Structural and Functional Biology Group, School of Medicine and School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Katherine Bowen
- School of Chemistry, Trinity College Dublin, Dublin, Ireland
| | - Chia-Ying Huang
- Swiss Light Source, Paul Scherrer Institute, Villigen, Switzerland
| | - Dietmar Weichert
- Membrane Structural and Functional Biology Group, School of Medicine and School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Warispreet Singh
- School of Pharmacy, Queen's University Belfast, Belfast, United Kingdom
- Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, United Kingdom
- Hub for Biotechnology in Build Environment, Newcastle upon Tyne, United Kingdom
| | - Irina G Tikhonova
- School of Pharmacy, Queen's University Belfast, Belfast, United Kingdom
| | - Eoin M Scanlan
- School of Chemistry, Trinity College Dublin, Dublin, Ireland
| | - Vincent Olieric
- Swiss Light Source, Paul Scherrer Institute, Villigen, Switzerland
| | - Martin Caffrey
- Membrane Structural and Functional Biology Group, School of Medicine and School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
12
|
Soltan MA, Magdy D, Solyman SM, Hanora A. Design of Staphylococcus aureus New Vaccine Candidates with B and T Cell Epitope Mapping, Reverse Vaccinology, and Immunoinformatics. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2021; 24:195-204. [PMID: 32286190 DOI: 10.1089/omi.2019.0183] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
An effective vaccine against Staphylococcus aureus infection is a major planetary heath priority, particularly with increasing antibiotic resistance worldwide. Previous efforts for a highly effective S. aureus vaccine were largely unsuccessful, in part, because the vaccine designs have tended to target mainly the B cell immunity and development of opsonic antibodies. In contrast, recent observations suggest that cell mediated immunity may be critical for protection against S. aureus. In addition, the S. aureus surface proteins are among the key immunodominant antigens because they are the first molecules to interact with the host organism cells and tissues. We report here an original vaccinomics study in which we used a reverse vaccinology and immunoinformatics in silico strategy integrated with genomics. After analyzing 2767 proteins, we defined 16 proteins of S. aureus as promising subunit vaccine candidates. Phosphatidylinositol phosphodiesterase (Plc) is secreted by extracellular pathogens such as S. aureus. We mapped the B and T cell epitopes for the Plc protein, tested the reactivity of the synthesized epitopes by Western blotting, and verified our findings in a pilot study of 10 patients with S. aureus infection. The peptides were then tested for their protective effect in groups of mice challenged with pathogenic S. aureus strain, which showed high protection level. These findings warrant further translational research for development of novel vaccines against S. aureus infection. Reverse vaccinology is an advanced approach that can be applied to identify new vaccine candidates against a host of microorganisms, including S. aureus.
Collapse
Affiliation(s)
- Mohamed A Soltan
- Department of Microbiology and Immunology, Faculty of Pharmacy, Sinai University, Ismailia, Egypt.,Department of Microbiology and Immunology, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt
| | - Dalia Magdy
- Department of Microbiology and Immunology, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt
| | - Samar M Solyman
- Department of Microbiology and Immunology, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt
| | - Amro Hanora
- Department of Microbiology and Immunology, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
13
|
Park B, Liu GY. Immune-Based Anti-Staphylococcal Therapeutic Approaches. Microorganisms 2021; 9:microorganisms9020328. [PMID: 33562054 PMCID: PMC7915210 DOI: 10.3390/microorganisms9020328] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 02/02/2021] [Accepted: 02/03/2021] [Indexed: 12/20/2022] Open
Abstract
Widespread methicillin-resistant Staphylococcus aureus (S. aureus) infections within community and healthcare settings are responsible for accelerated development of antibiotic resistance. As the antibiotic pipeline began drying up, alternative strategies were sought for future treatment of S. aureus infections. Here, we review immune-based anti-staphylococcal strategies that, unlike conventional antibiotics, target non-essential gene products elaborated by the pathogen. These strategies stimulate narrow or broad host immune mechanisms that are critical for anti-staphylococcal defenses. Alternative approaches aim to disrupt bacterial virulence mechanisms that enhance pathogen survival or induce immunopathology. Although immune-based therapeutics are unlikely to replace antibiotics in patient treatment in the near term, they have the potential to significantly improve upon the performance of antibiotics for treatment of invasive staphylococcal diseases.
Collapse
Affiliation(s)
- Bonggoo Park
- Cedars Sinai Medical Center, Division of Pediatric Infectious Diseases and the Immunobiology Research Institute, Los Angeles, CA 90048, USA;
| | - George Y. Liu
- Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA
- Correspondence:
| |
Collapse
|
14
|
Armentrout EI, Liu GY, Martins GA. T Cell Immunity and the Quest for Protective Vaccines against Staphylococcus aureus Infection. Microorganisms 2020; 8:microorganisms8121936. [PMID: 33291260 PMCID: PMC7762175 DOI: 10.3390/microorganisms8121936] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 11/27/2020] [Accepted: 11/30/2020] [Indexed: 12/14/2022] Open
Abstract
Staphylococcus aureus is a wide-spread human pathogen, and one of the top causative agents of nosocomial infections. The prevalence of antibiotic-resistant S. aureus strains, which are associated with higher mortality and morbidity rates than antibiotic-susceptible strains, is increasing around the world. Vaccination would be an effective preventive measure against S. aureus infection, but to date, every vaccine developed has failed in clinical trials, despite inducing robust antibody responses. These results suggest that induction of humoral immunity does not suffice to confer protection against the infection. Evidence from studies in murine models and in patients with immune defects support a role of T cell-mediated immunity in protective responses against S. aureus. Here, we review the current understanding of the mechanisms underlying adaptive immunity to S. aureus infections and discuss these findings in light of the recent S. aureus vaccine trial failures. We make the case for the need to develop anti-S. aureus vaccines that can specifically elicit robust and durable protective memory T cell subsets.
Collapse
Affiliation(s)
- Erin I. Armentrout
- Lung Institute, Cedars-Sinai Medical Center (CSMC), Los Angeles, CA 90048, USA;
- Division of Pulmonary and Critical Care Medicine, CSMC, Los Angeles, CA 90048, USA
| | - George Y. Liu
- Collaborative to Halt Antibiotic-Resistant Microbes, University of California, San Diego, La Jolla, CA 92161, USA;
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Gislâine A. Martins
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute (IBIRI), CSMC, Los Angeles, CA 90048, USA
- Department of Biomedical Sciences, Research Division of Immunology, CSMC, Los Angeles, CA 90048, USA
- Department of Medicine, Division of Gastroenterology, CSMC, Los Angeles, CA 90048, USA
- Correspondence:
| |
Collapse
|
15
|
Acapsular Staphylococcus aureus with a non-functional agr regains capsule expression after passage through the bloodstream in a bacteremia mouse model. Sci Rep 2020; 10:14108. [PMID: 32839485 PMCID: PMC7445255 DOI: 10.1038/s41598-020-70671-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 07/23/2020] [Indexed: 01/18/2023] Open
Abstract
Selection pressures exerted on Staphylococcus aureus by host factors during infection may lead to the emergence of regulatory phenotypes better adapted to the infection site. Traits convenient for persistence may be fixed by mutation thus turning these mutants into microevolution endpoints. The feasibility that stable, non-encapsulated S. aureus mutants can regain expression of key virulence factors for survival in the bloodstream was investigated. S. aureus agr mutant HU-14 (IS256 insertion in agrC) from a patient with chronic osteomyelitis was passed through the bloodstream using a bacteriemia mouse model and derivative P3.1 was obtained. Although IS256 remained inserted in agrC, P3.1 regained production of capsular polysaccharide type 5 (CP5) and staphyloxanthin. Furthermore, P3.1 expressed higher levels of asp23/SigB when compared with parental strain HU-14. Strain P3.1 displayed decreased osteoclastogenesis capacity, thus indicating decreased adaptability to bone compared with strain HU-14 and exhibited a trend to be more virulent than parental strain HU-14. Strain P3.1 exhibited the loss of one IS256 copy, which was originally located in the HU-14 noncoding region between dnaG (DNA primase) and rpoD (sigA). This loss may be associated with the observed phenotype change but the mechanism remains unknown. In conclusion, S. aureus organisms that escape the infected bone may recover the expression of key virulence factors through a rapid microevolution pathway involving SigB regulation of key virulence factors.
Collapse
|
16
|
Marchetti M, De Bei O, Bettati S, Campanini B, Kovachka S, Gianquinto E, Spyrakis F, Ronda L. Iron Metabolism at the Interface between Host and Pathogen: From Nutritional Immunity to Antibacterial Development. Int J Mol Sci 2020; 21:E2145. [PMID: 32245010 PMCID: PMC7139808 DOI: 10.3390/ijms21062145] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/16/2020] [Accepted: 03/17/2020] [Indexed: 02/08/2023] Open
Abstract
Nutritional immunity is a form of innate immunity widespread in both vertebrates and invertebrates. The term refers to a rich repertoire of mechanisms set up by the host to inhibit bacterial proliferation by sequestering trace minerals (mainly iron, but also zinc and manganese). This strategy, selected by evolution, represents an effective front-line defense against pathogens and has thus inspired the exploitation of iron restriction in the development of innovative antimicrobials or enhancers of antimicrobial therapy. This review focuses on the mechanisms of nutritional immunity, the strategies adopted by opportunistic human pathogen Staphylococcus aureus to circumvent it, and the impact of deletion mutants on the fitness, infectivity, and persistence inside the host. This information finally converges in an overview of the current development of inhibitors targeting the different stages of iron uptake, an as-yet unexploited target in the field of antistaphylococcal drug discovery.
Collapse
Affiliation(s)
- Marialaura Marchetti
- Interdepartmental Center Biopharmanet-TEC, University of Parma, 43124 Parma, Italy; (M.M.); (S.B.)
| | - Omar De Bei
- Department of Food and Drug, University of Parma, 43124 Parma, Italy; (O.D.B.); (B.C.)
| | - Stefano Bettati
- Interdepartmental Center Biopharmanet-TEC, University of Parma, 43124 Parma, Italy; (M.M.); (S.B.)
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
- Institute of Biophysics, National Research Council, 56124 Pisa, Italy
- National Institute of Biostructures and Biosystems, 00136 Rome, Italy
| | - Barbara Campanini
- Department of Food and Drug, University of Parma, 43124 Parma, Italy; (O.D.B.); (B.C.)
| | - Sandra Kovachka
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy; (S.K.); (E.G.); (F.S.)
| | - Eleonora Gianquinto
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy; (S.K.); (E.G.); (F.S.)
| | - Francesca Spyrakis
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy; (S.K.); (E.G.); (F.S.)
| | - Luca Ronda
- Interdepartmental Center Biopharmanet-TEC, University of Parma, 43124 Parma, Italy; (M.M.); (S.B.)
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
- Institute of Biophysics, National Research Council, 56124 Pisa, Italy
| |
Collapse
|
17
|
Cunha AF, Andrade HM, Souza FN, Fialho Júnior LC, Rosa DLSO, Ramos Sanchez EM, Gidlund M, Goto H, Brito MAVP, Guimarães AS, Lage AP, Reis LC, Della Libera AMMP, Heinemann MB, Cerqueira MMOP. Comparison of antibody repertories against Staphylococcus aureus in healthy and infected dairy cows with a distinct mastitis history and vaccinated with a polyvalent mastitis vaccine. J Dairy Sci 2020; 103:4588-4605. [PMID: 32113759 DOI: 10.3168/jds.2019-17084] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Accepted: 11/25/2019] [Indexed: 12/22/2022]
Abstract
Staphylococcus aureus is one of the pathogens most frequently isolated from cases of mastitis worldwide. To decrease the effect of S. aureus mastitis in dairy farming, alternative strategies for controlling mastitis are needed that depend on a better knowledge of cow-to-cow variations in S. aureus antibody production. The present study sought to explore the diversity of S. aureus antibodies produced by dairy cows with a distinct mastitis history and vaccinated with a polyvalent mastitis vaccine. We obtained protein extracts from S. aureus isolates derived from persistent subclinical mastitis. Proteins were fractionated using 2-dimensional gel electrophoresis and Western blotting. Then, Western blotting membranes were exposed to sera from 24 dairy cows that had been divided into the following groups: vaccinated dairy cows that were infected with S. aureus, further subdivided according to whether they (a) remained infected by S. aureus or (b) recovered from the intramammary infection; unvaccinated dairy cows infected with S. aureus; and vaccinated healthy dairy cows with no history of S. aureus mastitis. Proteins found to be reactive by Western blot were identified by mass spectrometry (MALDI/TOF-TOF). Our most important finding was that F0F1 ATP synthase subunit α, succinyl-diaminopimelate desuccinylase, and cysteinyl-tRNA synthetase were potential candidate proteins for the prevention of S. aureus mastitis. This study strengthens the notion that variations among animals should not be ignored and shows that the heterogeneity of antibody production against anti-staphylococcal antigens in animals may enable the identification of new immunotherapy targets.
Collapse
Affiliation(s)
- A F Cunha
- Departamento de Medicina Veterinária Preventiva, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte 31270-010, Brazil; Departamento de Tecnologia e Inspeção de Produtos de Origem Animal, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte 31270-010, Brazil.
| | - H M Andrade
- Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - F N Souza
- Departamento de Medicina Veterinária Preventiva, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte 31270-010, Brazil; Departamento de Tecnologia e Inspeção de Produtos de Origem Animal, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte 31270-010, Brazil; Veterinary Clinical Immunology Research Group, Departamento de Clínica Médica, Faculdade de Medicina Veterinária e Zootecnia, Universidade de São Paulo, São Paulo 05508-270, Brazil; Programa de Pós-graduação em Ciência Animal, Universidade Federal da Paraíba, Areia 58397-000, Brazil
| | - L C Fialho Júnior
- Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - D L S O Rosa
- Departamento de Medicina Veterinária Preventiva, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte 31270-010, Brazil; Departamento de Tecnologia e Inspeção de Produtos de Origem Animal, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte 31270-010, Brazil
| | - E M Ramos Sanchez
- Laboratório de Sorologia e Imunobiologia, Instituto de Medicina Tropical, Universidade de São Paulo, São Paulo 05403-000, Brazil; Departamento de Salud Publica, Facultad de Ciencias de La Salud, Universidad Nacional Toribio Rodriguez de Mendoza de Amazonas, Chachapoyas 01000, Peru
| | - M Gidlund
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-900, Brazil
| | - H Goto
- Laboratório de Sorologia e Imunobiologia, Instituto de Medicina Tropical, Universidade de São Paulo, São Paulo 05403-000, Brazil
| | - M A V P Brito
- EMBRAPA-Gado de Leite, Avenida Eugênio do Nascimento, 610, Juiz de Fora 36038-330, Brazil
| | - A S Guimarães
- Departamento de Medicina Veterinária, Universidade Federal de Lavras, Lavras 37200-000, Brazil
| | - A P Lage
- Departamento de Medicina Veterinária Preventiva, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte 31270-010, Brazil
| | - L C Reis
- Laboratório de Sorologia e Imunobiologia, Instituto de Medicina Tropical, Universidade de São Paulo, São Paulo 05403-000, Brazil
| | - A M M P Della Libera
- Veterinary Clinical Immunology Research Group, Departamento de Clínica Médica, Faculdade de Medicina Veterinária e Zootecnia, Universidade de São Paulo, São Paulo 05508-270, Brazil
| | - M B Heinemann
- Departamento de Medicina Veterinária Preventiva e Saúde Animal, Faculdade de Medicina Veterinária e Zootecnia, Universidade de São Paulo, São Paulo 05508-270, Brazil
| | - M M O P Cerqueira
- Departamento de Tecnologia e Inspeção de Produtos de Origem Animal, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte 31270-010, Brazil
| |
Collapse
|
18
|
Creech CB, Frenck RW, Fiquet A, Feldman R, Kankam MK, Pathirana S, Baber J, Radley D, Cooper D, Eiden J, Gruber WC, Jansen KU, Anderson AS, Gurtman A. Persistence of Immune Responses Through 36 Months in Healthy Adults After Vaccination With a Novel Staphylococcus aureus 4-Antigen Vaccine (SA4Ag). Open Forum Infect Dis 2019; 7:ofz532. [PMID: 31993453 PMCID: PMC6978999 DOI: 10.1093/ofid/ofz532] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 12/11/2019] [Indexed: 12/19/2022] Open
Abstract
Background Staphylococcus aureus causes serious health care- and community-associated disease, requiring improved preventive measures such as vaccines. The investigational S. aureus 4-antigen vaccine (SA4Ag), comprising capsular polysaccharide serotypes 5 and 8 (CP5 and CP8) conjugated to CRM197, recombinant mutant clumping factor A (rmClfA), and recombinant manganese transporter protein C (rP305A or rMntC), was well tolerated, inducing robust functional immune responses to all 4 antigens through 12 months postvaccination. This is a serological extension study through 36 months postvaccination. Methods In 2 previous studies, healthy adults received SA4Ag, SA3Ag (without rMntC), or placebo; serology was also assessed at ~24 and ~36 months postvaccination. Functional immune responses (antibody responses that facilitate killing of S. aureus or neutralize S. aureus virulence mechanisms) were assessed with opsonophagocytic activity killing assays (CP5 or CP8) and a fibrinogen-binding inhibition assay (ClfA). A competitive Luminex immunoassay assessed ClfA and rMntC responses. Adverse events within 48 hours of blood draw were recorded. Results Four hundred forty subjects (18-64 years old, 255; 65-85 years old, 185) were enrolled. At 24 and 36 months postvaccination, subjects receiving SA4Ag had substantially higher geometric mean titers (GMTs) for CP5, CP8, and ClfA vs baseline; geometric mean fold rises (GMFRs) from baseline to month 36 were 2.7-8.1. For rMntC, 36-month GMTs declined from peak levels but remained above baseline for all SA4Ag groups; GMFRs from baseline to month 36 were 1.8 and 1.5 in the younger and older cohorts, respectively. Conclusions Persistent functional immune responses to S. aureus antigens were observed through 36 months in healthy adults. ClinicalTrialsgov NCT01643941 and NCT01364571.
Collapse
Affiliation(s)
- C Buddy Creech
- Vanderbilt University School of Medicine and the Monroe Carell Jr. Children's Hospital, Nashville, Tennessee, USA
| | - Robert W Frenck
- Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Anne Fiquet
- Pfizer Vaccines Research & Development, Hurley, Berkshire, UK
| | - Robert Feldman
- QPS-MRA, Miami Research Associates LLC, Miami, Florida, USA
| | - Martin K Kankam
- Vince and Associates Clinical Research, Overland Park, Kansas, USA
| | - Sudam Pathirana
- Pfizer Vaccines Research & Development, Pearl River, New York, USA
| | - James Baber
- Pfizer Vaccines Research & Development, Sydney, New South Wales, Australia
| | - David Radley
- Pfizer Vaccines Research & Development, Pearl River, New York, USA
| | - David Cooper
- Pfizer Vaccines Research & Development, Pearl River, New York, USA
| | - Joseph Eiden
- Pfizer Vaccines Research & Development, Pearl River, New York, USA
| | - William C Gruber
- Pfizer Vaccines Research & Development, Pearl River, New York, USA
| | - Kathrin U Jansen
- Pfizer Vaccines Research & Development, Pearl River, New York, USA
| | | | | |
Collapse
|
19
|
Giersing BK, Vekemans J, Nava S, Kaslow DC, Moorthy V. Report from the World Health Organization's third Product Development for Vaccines Advisory Committee (PDVAC) meeting, Geneva, 8-10th June 2016. Vaccine 2019; 37:7315-7327. [PMID: 28262332 PMCID: PMC7131228 DOI: 10.1016/j.vaccine.2016.10.090] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 10/25/2016] [Indexed: 01/13/2023]
Abstract
The third meeting of WHO's Product Development for Vaccines Advisory Committee (PDVAC) was held in June 2016, with a remit to revisit the pathogen areas for which significant progress has occurred since recommendations from the 2015 meeting, as well as to consider new advances in the development of vaccines against other pathogens. Since the previous meeting, significant progress has been made with regulatory approvals of the first malaria and dengue vaccines, and the first phase III trials of a respiratory syncytial virus (RSV) vaccine candidate has started in the elderly and pregnant women. In addition, PDVAC has also supported vaccine development efforts against important emerging pathogens, including Middle Eastern Coronavirus (MERS CoV) and Zika virus. Trials of HIV and tuberculosis vaccine candidates are steadily progressing towards pivotal data points, and the leading norovirus vaccine candidate has entered a phase IIb efficacy study. WHO's Immunization, Vaccine and Biologicals (IVB) department is actively working in several pathogen areas on the recommendation of PDVAC, as well as continuing horizon scanning for advances in the development of vaccines that may benefit low and middle income countries (LMICs), such as the recent licensure of the enterovirus 71 (EV71) vaccine in China. Following on from discussions with WHO's Strategic Advisory Group of Experts (SAGE) on Immunization, PDVAC will also look beyond licensure and consider data needs for vaccine recommendation and implementation to reduce the delay between vaccine approval and vaccine impact.
Collapse
Affiliation(s)
- Birgitte K Giersing
- Initiative for Vaccine Research, World Health Organization, CH-1211 Geneva 27, Switzerland
| | - Johan Vekemans
- Initiative for Vaccine Research, World Health Organization, CH-1211 Geneva 27, Switzerland
| | - Samantha Nava
- University of Texas Medical Branch, Galveston, TX, USA
| | | | - Vasee Moorthy
- Initiative for Vaccine Research, World Health Organization, CH-1211 Geneva 27, Switzerland.
| |
Collapse
|
20
|
Affiliation(s)
- Stewart Sell
- Wadsworth Center, New York State Department of Health, Empire State Plaza, Albany, NY, USA
| |
Collapse
|
21
|
Bacterial outer membrane vesicles engineered with lipidated antigens as a platform for Staphylococcus aureus vaccine. Proc Natl Acad Sci U S A 2019; 116:21780-21788. [PMID: 31591215 PMCID: PMC6815149 DOI: 10.1073/pnas.1905112116] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Bacterial outer membrane vesicles (OMVs) represent an interesting vaccine platform for their built-in adjuvanticity and simplicity of production process. Moreover, OMVs can be decorated with foreign antigens using different synthetic biology approaches. However, the optimal OMV engineering strategy, which should guarantee the OMV compartmentalization of most heterologous antigens in quantities high enough to elicit protective immune responses, remains to be validated. In this work we exploited the lipoprotein transport pathway to engineer OMVs with foreign proteins. Using 5 Staphylococcus aureus protective antigens expressed in Escherichia coli as fusions to a lipoprotein leader sequence, we demonstrated that all 5 antigens accumulated in the vesicular compartment at a concentration ranging from 5 to 20% of total OMV proteins, suggesting that antigen lipidation could be a universal approach for OMV manipulation. Engineered OMVs elicited high, saturating antigen-specific antibody titers when administered to mice in quantities as low as 0.2 μg/dose. Moreover, the expression of lipidated antigens in E. coli BL21(DE3)ΔompAΔmsbBΔpagP was shown to affect the lipopolysaccharide structure, with the result that the TLR4 agonist activity of OMVs was markedly reduced. These results, together with the potent protective activity of engineered OMVs observed in mice challenged with S. aureus Newman strain, makes the 5-combo-OMVs a promising vaccine candidate to be tested in clinics.
Collapse
|
22
|
Foster TJ. Surface Proteins of Staphylococcus aureus. Microbiol Spectr 2019; 7:10.1128/microbiolspec.gpp3-0046-2018. [PMID: 31267926 PMCID: PMC10957221 DOI: 10.1128/microbiolspec.gpp3-0046-2018] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Indexed: 12/20/2022] Open
Abstract
The surface of Staphylococcus aureus is decorated with over 20 proteins that are covalently anchored to peptidoglycan by the action of sortase A. These cell wall-anchored (CWA) proteins can be classified into several structural and functional groups. The largest is the MSCRAMM family, which is characterized by tandemly repeated IgG-like folded domains that bind peptide ligands by the dock lock latch mechanism or the collagen triple helix by the collagen hug. Several CWA proteins comprise modules that have different functions, and some individual domains can bind different ligands, sometimes by different mechanisms. For example, the N-terminus of the fibronectin binding proteins comprises an MSCRAMM domain which binds several ligands, while the C-terminus is composed of tandem fibronectin binding repeats. Surface proteins promote adhesion to host cells and tissue, including components of the extracellular matrix, contribute to biofilm formation by stimulating attachment to the host or indwelling medical devices followed by cell-cell accumulation via homophilic interactions between proteins on neighboring cells, help bacteria evade host innate immune responses, participate in iron acquisition from host hemoglobin, and trigger invasion of bacteria into cells that are not normally phagocytic. The study of genetically manipulated strains using animal infection models has shown that many CWA proteins contribute to pathogenesis. Fragments of CWA proteins have the potential to be used in multicomponent vaccines to prevent S. aureus infections.
Collapse
|
23
|
de Souza PRK, Ferreira SS, Nunes FPB, Casagrande FB, Tessaro FHG, Silva MCF, Cruz JWMC, Mamizuka EM, Martins JO. Cytokine and Adhesion Molecule Expression Induced by Different Strains of Staphylococcus aureus in Type 1 Diabetic Rats: Role of Insulin. Front Immunol 2019; 9:3165. [PMID: 30705678 PMCID: PMC6344427 DOI: 10.3389/fimmu.2018.03165] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Accepted: 12/21/2018] [Indexed: 12/24/2022] Open
Abstract
Introduction: Staphylococcus aureus may provoke peritonitis and death, especially in immunocompromized individuals such as diabetic patients. We evaluated the role of insulin in S. aureus-induced peritoneal infection in diabetic and non-diabetic rats. Materials/Methods: Alloxan-diabetic male Wistar rats and their respective controls received intraperitoneal injections of different strains of S. aureus or sterile phosphate-buffered saline. After 3 days of infection, the first set of diabetic and non-diabetic rats received 4 and 1 IU, respectively, of neutral protamine Hagedorn insulin and were analyzed 8 h later. The second set of diabetic and non-diabetic rats received 4 and 1 IU, respectively, of insulin 2 h before intraperitoneal infection and a half dose of insulin at 5 p.m. for the next 2 days and were analyzed 16 h later. The following measurements were performed: (a) number of cells in the peritoneal lavage fluid (PeLF), white blood cell count, and blood glucose; (b) serum insulin and corticosterone; (c) cytokine levels in the PeLF; (d) expression of adhesion molecules in the vascular endothelium; and (e) microbicidal activity. Results: Diabetic rats showed an increased number of polymorphonuclear leukocytes (PMNs) and increased concentrations of CINC-1, IL-4, and IFN-γ in the PeLF after infection with the ATCC 25923 or N315 αHL+ strain. The mesenteric expression of PECAM-1 was increased after infection with the N315 HLA+ strain. ICAM-1 expression was increased with ATCC infection. Treatment of diabetic rats with a single dose of insulin restored CINC-1 levels in the PeLF for both strains; however, PMN migration, IL-4, and IFN-γ were restored in rats infected with the ATCC strain, whereas the PeLF concentrations of CINC-2, IL-1β, and IL-4 were increased in N315-infected animals. Insulin restored PMN migration and CINC-2 levels in the PeLF in ATCC-infected rats. After multiple treatments with insulin, the levels of IL-1β, IL-6, and IFN-γ were increased in the PeLF of diabetic rats after infection with either strain, and CINC-2 levels were restored in N315-infected animals. Conclusion: These results suggest that insulin distinctively modulates cytokine production or release, PMN leukocyte migration, and adhesion molecule expression during the course of peritonitis induced by different strains of S. aureus.
Collapse
Affiliation(s)
- Paula R Knox de Souza
- Laboratory of Immunoendocrinology, Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences (FCF), University of São Paulo (USP), São Paulo, Brazil.,Universidade Paulista, São Paulo, Brazil
| | - Sabrina S Ferreira
- Laboratory of Immunoendocrinology, Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences (FCF), University of São Paulo (USP), São Paulo, Brazil
| | - Fernanda P B Nunes
- Laboratory of Immunoendocrinology, Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences (FCF), University of São Paulo (USP), São Paulo, Brazil
| | - Felipe B Casagrande
- Laboratory of Immunoendocrinology, Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences (FCF), University of São Paulo (USP), São Paulo, Brazil
| | - Fernando H G Tessaro
- Laboratory of Immunoendocrinology, Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences (FCF), University of São Paulo (USP), São Paulo, Brazil
| | - Mariana C F Silva
- Laboratory of Immunoendocrinology, Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences (FCF), University of São Paulo (USP), São Paulo, Brazil
| | | | - Elsa M Mamizuka
- Laboratory of Microbiology, Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences (FCF), University of São Paulo (USP), São Paulo, Brazil
| | - Joilson O Martins
- Laboratory of Immunoendocrinology, Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences (FCF), University of São Paulo (USP), São Paulo, Brazil
| |
Collapse
|
24
|
Mohamed N, Timofeyeva Y, Jamrozy D, Rojas E, Hao L, Silmon de Monerri NC, Hawkins J, Singh G, Cai B, Liberator P, Sebastian S, Donald RGK, Scully IL, Jones CH, Creech CB, Thomsen I, Parkhill J, Peacock SJ, Jansen KU, Holden MTG, Anderson AS. Molecular epidemiology and expression of capsular polysaccharides in Staphylococcus aureus clinical isolates in the United States. PLoS One 2019; 14:e0208356. [PMID: 30641545 PMCID: PMC6331205 DOI: 10.1371/journal.pone.0208356] [Citation(s) in RCA: 20] [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: 08/25/2018] [Accepted: 11/15/2018] [Indexed: 12/14/2022] Open
Abstract
Staphylococcus aureus capsular polysaccharides (CP) are important virulence factors under evaluation as vaccine antigens. Clinical S. aureus isolates have the biosynthetic capability to express either CP5 or CP8 and an understanding of the relationship between CP genotype/phenotype and S. aureus epidemiology is valuable. Using whole genome sequencing, the clonal relatedness and CP genotype were evaluated for disease-associated S. aureus isolates selected from the Tigecycline Evaluation and Surveillance Trial (T.E.S.T) to represent different geographic regions in the United States (US) during 2004 and 2009–10. Thirteen prominent clonal complexes (CC) were identified, with CC5, 8, 30 and 45 representing >80% of disease isolates. CC5 and CC8 isolates were CP type 5 and, CC30 and CC45 isolates were CP type 8. Representative isolates from prevalent CC were susceptible to in vitro opsonophagocytic killing elicited by anti-CP antibodies, demonstrating that susceptibility to opsonic killing is not linked to the genetic lineage. However, as not all S. aureus isolates may express CP, isolates representing the diversity of disease isolates were assessed for CP production. While approximately 35% of isolates (primarily CC8) did not express CP in vitro, CP expression could be clearly demonstrated in vivo for 77% of a subset of these isolates (n = 20) despite the presence of mutations within the capsule operon. CP expression in vivo was also confirmed indirectly by measuring an increase in CP specific antibodies in mice infected with CP5 or CP8 isolates. Detection of antigen expression in vivo in relevant disease states is important to support the inclusion of these antigens in vaccines. Our findings confirm the validity of CP as vaccine targets and the potential of CP-based vaccines to contribute to S. aureus disease prevention.
Collapse
Affiliation(s)
- Naglaa Mohamed
- Pfizer Vaccine Research and Development, Pearl River, New York, United States of America
| | - Yekaterina Timofeyeva
- Pfizer Vaccine Research and Development, Pearl River, New York, United States of America
| | - Dorota Jamrozy
- The Wellcome Trust Sanger Institute, Cambridge, United Kingdom
| | - Eduardo Rojas
- Pfizer Vaccine Research and Development, Pearl River, New York, United States of America
| | - Li Hao
- Pfizer Vaccine Research and Development, Pearl River, New York, United States of America
| | | | - Julio Hawkins
- Pfizer Vaccine Research and Development, Pearl River, New York, United States of America
| | - Guy Singh
- Pfizer Vaccine Research and Development, Pearl River, New York, United States of America
| | - Bing Cai
- Pfizer Vaccine Research and Development, Pearl River, New York, United States of America
| | - Paul Liberator
- Pfizer Vaccine Research and Development, Pearl River, New York, United States of America
| | - Shite Sebastian
- Pfizer Vaccine Research and Development, Pearl River, New York, United States of America
| | - Robert G. K. Donald
- Pfizer Vaccine Research and Development, Pearl River, New York, United States of America
| | - Ingrid L. Scully
- Pfizer Vaccine Research and Development, Pearl River, New York, United States of America
| | - C. Hal Jones
- Pfizer Vaccine Research and Development, Pearl River, New York, United States of America
| | - C. Buddy Creech
- Vanderbilt Vaccine Research Program, Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Isaac Thomsen
- Vanderbilt Vaccine Research Program, Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Julian Parkhill
- The Wellcome Trust Sanger Institute, Cambridge, United Kingdom
| | - Sharon J. Peacock
- London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Kathrin U. Jansen
- Pfizer Vaccine Research and Development, Pearl River, New York, United States of America
| | | | - Annaliesa S. Anderson
- Pfizer Vaccine Research and Development, Pearl River, New York, United States of America
- * E-mail:
| |
Collapse
|
25
|
Manara S, Pasolli E, Dolce D, Ravenni N, Campana S, Armanini F, Asnicar F, Mengoni A, Galli L, Montagnani C, Venturini E, Rota-Stabelli O, Grandi G, Taccetti G, Segata N. Whole-genome epidemiology, characterisation, and phylogenetic reconstruction of Staphylococcus aureus strains in a paediatric hospital. Genome Med 2018; 10:82. [PMID: 30424799 PMCID: PMC6234625 DOI: 10.1186/s13073-018-0593-7] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 10/29/2018] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Staphylococcus aureus is an opportunistic pathogen and a leading cause of nosocomial infections. It can acquire resistance to all the antibiotics that entered the clinics to date, and the World Health Organization defined it as a high-priority pathogen for research and development of new antibiotics. A deeper understanding of the genetic variability of S. aureus in clinical settings would lead to a better comprehension of its pathogenic potential and improved strategies to contrast its virulence and resistance. However, the number of comprehensive studies addressing clinical cohorts of S. aureus infections by simultaneously looking at the epidemiology, phylogenetic reconstruction, genomic characterisation, and transmission pathways of infective clones is currently low, thus limiting global surveillance and epidemiological monitoring. METHODS We applied whole-genome shotgun sequencing (WGS) to 184 S. aureus isolates from 135 patients treated in different operative units of an Italian paediatric hospital over a timespan of 3 years, including both methicillin-resistant S. aureus (MRSA) and methicillin-sensitive S. aureus (MSSA) from different infection types. We typed known and unknown clones from their genomes by multilocus sequence typing (MLST), Staphylococcal Cassette Chromosome mec (SCCmec), Staphylococcal protein A gene (spa), and Panton-Valentine Leukocidin (PVL), and we inferred their whole-genome phylogeny. We explored the prevalence of virulence and antibiotic resistance genes in our cohort, and the conservation of genes encoding vaccine candidates. We also performed a timed phylogenetic investigation for a potential outbreak of a newly emerging nosocomial clone. RESULTS The phylogeny of the 135 single-patient S. aureus isolates showed a high level of diversity, including 80 different lineages, and co-presence of local, global, livestock-associated, and hypervirulent clones. Five of these clones do not have representative genomes in public databases. Variability in the epidemiology is mirrored by variability in the SCCmec cassettes, with some novel variants of the type IV cassette carrying extra antibiotic resistances. Virulence and resistance genes were unevenly distributed across different clones and infection types, with highly resistant and lowly virulent clones showing strong association with chronic diseases, and highly virulent strains only reported in acute infections. Antigens included in vaccine formulations undergoing clinical trials were conserved at different levels in our cohort, with only a few highly prevalent genes fully conserved, potentially explaining the difficulty of developing a vaccine against S. aureus. We also found a recently diverged ST1-SCCmecIV-t127 PVL- clone suspected to be hospital-specific, but time-resolved integrative phylogenetic analysis refuted this hypothesis and suggested that this quickly emerging lineage was acquired independently by patients. CONCLUSIONS Whole genome sequencing allowed us to study the epidemiology and genomic repertoire of S. aureus in a clinical setting and provided evidence of its often underestimated complexity. Some virulence factors and clones are specific of disease types, but the variability and dispensability of many antigens considered for vaccine development together with the quickly changing epidemiology of S. aureus makes it very challenging to develop full-coverage therapies and vaccines. Expanding WGS-based surveillance of S. aureus to many more hospitals would allow the identification of specific strains representing the main burden of infection and therefore reassessing the efforts for the discovery of new treatments and clinical practices.
Collapse
Affiliation(s)
- Serena Manara
- Centre for Integrative Biology, University of Trento, Trento, Italy
| | - Edoardo Pasolli
- Centre for Integrative Biology, University of Trento, Trento, Italy
| | - Daniela Dolce
- Cystic Fibrosis Center, Interdisciplinary Specialist Department, Anna Meyer Children's University Hospital, Florence, Italy
| | - Novella Ravenni
- Cystic Fibrosis Center, Interdisciplinary Specialist Department, Anna Meyer Children's University Hospital, Florence, Italy
| | - Silvia Campana
- Cystic Fibrosis Center, Interdisciplinary Specialist Department, Anna Meyer Children's University Hospital, Florence, Italy
| | | | | | - Alessio Mengoni
- Department of Biology, University of Florence, Florence, Italy
| | - Luisa Galli
- Department of Health Sciences, University of Florence, Florence, Italy
- Infectious Diseases Unit, Anna Meyer Children's University Hospital, Florence, Italy
| | - Carlotta Montagnani
- Infectious Diseases Unit, Anna Meyer Children's University Hospital, Florence, Italy
| | - Elisabetta Venturini
- Infectious Diseases Unit, Anna Meyer Children's University Hospital, Florence, Italy
| | - Omar Rota-Stabelli
- Department of Sustainable Agro-Ecosystems and Bioresources, Fondazione Edmund Mach, San Michele all'Adige, Italy
| | - Guido Grandi
- Centre for Integrative Biology, University of Trento, Trento, Italy
| | - Giovanni Taccetti
- Cystic Fibrosis Center, Interdisciplinary Specialist Department, Anna Meyer Children's University Hospital, Florence, Italy
| | - Nicola Segata
- Centre for Integrative Biology, University of Trento, Trento, Italy.
| |
Collapse
|
26
|
Wang Q, Wang X, Wang X, Feng R, Luo Q, Huang J. Generation of a novel Streptococcus agalactiae ghost vaccine and examination of its immunogenicity against virulent challenge in tilapia. FISH & SHELLFISH IMMUNOLOGY 2018; 81:49-56. [PMID: 29969706 DOI: 10.1016/j.fsi.2018.06.055] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 06/20/2018] [Accepted: 06/30/2018] [Indexed: 06/08/2023]
Abstract
Streptococcus agalactiae (S. agalactiae) is a gram-positive pathogen that causes a wide range of infections in fish and other animals including humans. Bacterial ghosts (BGs) are nonliving, empty cell envelopes and are well represented as novel vaccine candidates. In this study, we examined the immunogenicity and protective efficacy of S. agalactiae ghosts (SAG) against a virulent challenge in tilapia. Nonliving SAG was generated by a culture with Penicillin and Streptolysin, and then treated with the MIC of sodium hydroxide. The formation of a transmembrane lysis tunnel structure in SAG was visualized by electron microscopy. To investigate the SAG as a vaccine candidate, fish were divided into three groups, A (SAG immunized), B [Formalin-inactivated S. agalactiae (FSA) immunized] and C (phosphate-buffered saline, PBS-immunized control). The IgM antibody responses were significantly stronger in the SAG-immunized group than in FSA-immunized group, which was higher than in the non-immunized control group (P < 0.05). Moreover, phagocytic activity (percent phagocytes, PP) was significantly higher (p < 0.05) in the SAG-immunized group than in FSA-immunized group, which was higher than in the non-immunized control group (P < 0.05). In addition, non-specific immune immunity, such as lysozyme and superoxide dismutase activities, in the SAG-immunized fish showed significantly higher activities than FSA-immunized fish and the control group fish (P < 0.05). Also, fish immunized with SAG and FSA showed significantly higher (p < 0.05) gene expression of IL-1β, TNF-α, IFN-γ and TGF-β in the head kidney and spleen than fish treated with PBS during the whole observed period. In addition, fish immunized with SAG showed significantly higher gene expression of L-1β, TNF-α, and TGF-β in the spleen than in the FSA-immunized fish. Although there was no significant (P > 0.05) difference of survival rate (SR) or relative percent survival (RPS) between SAG and FSA immunized groups, they were all significantly more protected against the S. agalactiae challenge (SR: 86.67%, RPS: 76.395) and (SR: 80.00%, RPS: 67.50%) respectively, compared to the PBS-treated group (SR: 33.33%). These results suggest that immunization with SAG induces immune responses and provides protection against a virulent S. agalactiae challenge.
Collapse
Affiliation(s)
- Qishuo Wang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention & Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, Taian, 271018, PR China; Huaian research center, Institute of Hydrobiology, Chinese Academy of Sciences, Huaian 223000, PR China
| | - Xuepeng Wang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention & Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, Taian, 271018, PR China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 272000, PR China.
| | - Xuemei Wang
- Shandong Yisheng Livestock Veterinary Science Institute, 264000, Yantai, PR China
| | - Ruijuan Feng
- Jiangsu Tianshen Co., Ltd., 223000, Huai'an, PR China
| | - Qian Luo
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention & Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, Taian, 271018, PR China
| | - Jinjing Huang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention & Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, Taian, 271018, PR China
| |
Collapse
|
27
|
Gurtman A, Begier E, Mohamed N, Baber J, Sabharwal C, Haupt RM, Edwards H, Cooper D, Jansen KU, Anderson AS. The development of a staphylococcus aureus four antigen vaccine for use prior to elective orthopedic surgery. Hum Vaccin Immunother 2018; 15:358-370. [PMID: 30215582 DOI: 10.1080/21645515.2018.1523093] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Staphylococcus aureus (S. aureus) is a challenging bacterial pathogen which can cause a range of diseases, from mild skin infections, to more serious and invasive disease including deep or organ space surgical site infections, life-threatening bacteremia, and sepsis. S. aureus rapidly develops resistance to antibiotic treatments. Despite current infection control measures, the burden of disease remains high. The most advanced vaccine in clinical development is a 4 antigen S. aureus vaccine (SA4Ag) candidate that is being evaluated in a phase 2b/3 efficacy study in patients undergoing elective spinal fusion surgery (STaphylococcus aureus suRgical Inpatient Vaccine Efficacy [STRIVE]). SA4Ag has been shown in early phase clinical trials to be generally safe and well tolerated, and to induce high levels of bactericidal antibodies in healthy adults. In this review we discuss the design of SA4Ag, as well as the proposed clinical development plan supporting licensure of SA4Ag for the prevention of invasive disease caused by S. aureus in elective orthopedic surgical populations. We also explore the rationale for the generalizability of the results of the STRIVE efficacy study (patients undergoing elective open posterior multilevel instrumented spinal fusion surgery) to a broad elective orthopedic surgery population due to the common pathophysiology of invasive S. aureus disease and commonalties of patient and procedural risk factors for developing postoperative S. aureus surgical site infections.
Collapse
Affiliation(s)
- A Gurtman
- a Pfizer Vaccine Research and Development , Pfizer, Inc ., Pearl River , NY , USA
| | - E Begier
- a Pfizer Vaccine Research and Development , Pfizer, Inc ., Pearl River , NY , USA
| | - N Mohamed
- a Pfizer Vaccine Research and Development , Pfizer, Inc ., Pearl River , NY , USA
| | - J Baber
- b Pfizer Vaccine Research and Development , Sydney , NSW , Australia
| | - C Sabharwal
- a Pfizer Vaccine Research and Development , Pfizer, Inc ., Pearl River , NY , USA
| | - R M Haupt
- c Medical Development, Scientific and Clinical Affairs , Pfizer, Inc ., Collegeville , PA , USA
| | - H Edwards
- d World Wide Regulatory Affairs , Pfizer Inc ., Walton Oaks , UK
| | - D Cooper
- a Pfizer Vaccine Research and Development , Pfizer, Inc ., Pearl River , NY , USA
| | - K U Jansen
- a Pfizer Vaccine Research and Development , Pfizer, Inc ., Pearl River , NY , USA
| | - A S Anderson
- a Pfizer Vaccine Research and Development , Pfizer, Inc ., Pearl River , NY , USA
| |
Collapse
|
28
|
Liu Q, Mazhar M, Miller LS. Immune and Inflammatory Reponses to Staphylococcus aureus Skin Infections. CURRENT DERMATOLOGY REPORTS 2018; 7:338-349. [PMID: 30989002 DOI: 10.1007/s13671-018-0235-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Purpose of Review There have been recent advances in our understanding of cutaneous immune responses to the important human skin pathogen, Staphylococcus aureus (S. aureus). This review will highlight these insights into innate and adaptive immune mechanisms in host defense and cutaneous inflammation in response to S. aureus skin infections. Recent Findings Antimicrobial peptides, pattern recognition receptors and inflammasome activation function in innate immunity as well as T cells and their effector cytokines play a key role in adaptive immunity against S. aureus skin infections. In addition, certain mechanisms by which S. aureus contributes to aberrant cutaneous inflammation, such as in flares of the inflammatory skin disease atopic dermatitis have also been identified. Summary These cutaneous immune mechanisms could provide new targets for future vaccines and immune-based therapies to combat skin infections and cutaneous inflammation caused by S. aureus.
Collapse
Affiliation(s)
- Qi Liu
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Momina Mazhar
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Lloyd S Miller
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| |
Collapse
|
29
|
Nagy E, Nagy G, Power CA, Badarau A, Szijártó V. Anti-bacterial Monoclonal Antibodies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1053:119-153. [PMID: 29549638 DOI: 10.1007/978-3-319-72077-7_7] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The failing efficacy of antibiotics and the high mortality rate among high-risk patients calls for new treatment modalities for bacterial infections. Due to the vastly divergent pathogenesis of human pathogens, each microbe requires a tailored approach. The main modes of action of anti-bacterial antibodies are virulence factor neutralization, complement-mediated bacterial lysis and enhancement of opsonophagocytic uptake and killing (OPK). Gram-positive bacteria cannot be lysed by complement and their pathogenesis often involves secreted toxins, therefore typically toxin-neutralization and OPK activity are required to prevent and ameliorate disease. In fact, the success stories in terms of approved products, in the anti-bacterial mAb field are based on toxin neutralization (Bacillus anthracis, Clostridium difficile). In contrast, Gram-negative bacteria are vulnerable to antibody-dependent complement-mediated lysis, while their pathogenesis rarely relies on secreted exotoxins, and involves the pro-inflammatory endotoxin (lipopolysaccharide). Given the complexity of bacterial pathogenesis, antibody therapeutics are expected to be most efficient upon targeting more than one virulence factor and/or combining different modes of action. The improved understanding of bacterial pathogenesis combined with the versatility and maturity of antibody discovery technologies available today are pivotal for the design of novel anti-bacterial therapeutics. The intensified research generating promising proof-of-concept data, and the increasing number of clinical programs with anti-bacterial mAbs, indicate that the field is ready to fulfill its promise in the coming years.
Collapse
Affiliation(s)
- Eszter Nagy
- Arsanis Biosciences GmbH/Arsanis, Inc, Vienna, Austria.
| | - Gábor Nagy
- Arsanis Biosciences GmbH/Arsanis, Inc, Vienna, Austria
| | | | | | | |
Collapse
|
30
|
Abstract
Staphylococcus aureus causes severe disease in humans for which no licensed vaccine exists. A novel vaccine is in development that targets multiple elements of the bacteria since single-component vaccines have not shown efficacy to date. How these multiple components alter the immune response raised by the vaccine is not well studied. We found that the addition of two protein components did not alter substantially the antibody responses raised with respect to function or mobilization of B cells. There was also not a substantial change in the activity of T cells, another part of the adaptive response. This study showed that protection by this vaccine may be mediated primarily by antibody protection. Staphylococcus aureus causes severe disease in humans for which no licensed vaccine exists. A novel S. aureus vaccine (SA4Ag) is in development, targeting the capsular polysaccharides (CPs) and two virulence-associated surface proteins. Vaccine-elicited antibody responses to CPs are efficacious against serious infection by other encapsulated bacteria. Studies of natural S. aureus infection have also shown a role for TH17 and/or TH1 responses in protection. Single-antigen vaccines, including CPs, have not been effective against S. aureus; a multiantigen vaccine approach is likely required. However, the impact of addition of protein antigens on the immune response to CPs has not been studied. Here, the immune response induced by a bivalent CP conjugate vaccine (to model the established mechanism of action of vaccine-induced protection against Gram-positive pathogens) was compared to the response induced by SA4Ag, which contains both CP conjugates and protein antigens, in cynomolgus macaques. Microengraving, flow cytometry, opsonophagocytic assays, and Luminex technology were used to analyze the B-cell, T-cell, functional antibody, and innate immune responses. Both the bivalent CP vaccine and SA4Ag induced cytokine production from naive cells and antigen-specific memory B-cell and functional antibody responses. Increases in levels of circulating, activated T cells were not apparent following vaccination, nor was a TH17 or TH1 response evident. However, our data are consistent with a vaccine-induced recruitment of T follicular helper (TFH) cells to lymph nodes. Collectively, these data suggest that the response to SA4Ag is primarily mediated by B cells and antibodies that abrogate important S. aureus virulence mechanisms. IMPORTANCEStaphylococcus aureus causes severe disease in humans for which no licensed vaccine exists. A novel vaccine is in development that targets multiple elements of the bacteria since single-component vaccines have not shown efficacy to date. How these multiple components alter the immune response raised by the vaccine is not well studied. We found that the addition of two protein components did not alter substantially the antibody responses raised with respect to function or mobilization of B cells. There was also not a substantial change in the activity of T cells, another part of the adaptive response. This study showed that protection by this vaccine may be mediated primarily by antibody protection.
Collapse
|
31
|
Abstract
The problem of antimicrobial resistance (AMR) and the associated morbidity and mortality due to antibiotic resistant bacterial pathogens is not new. However, AMR has been increasing at an alarming rate with appearances of diseases caused by bacteria exhibiting resistance to not just one but multiple classes of antibiotics. The World Health Organization (WHO) supported by governments, health ministries and health agencies has formulated global action plans to combat the rise in AMR, supporting a number of proven initiatives such as antimicrobial stewardship, investments in development of new classes of antibiotics, and educational programs designed to eliminate inappropriate antibiotic use. Vaccines as tools to reduce AMR have historically been under-recognized, yet the positive effect in reducing AMR has been well established. For example Haemophilus influenzae type B (Hib) as well as Streptococcus pneumoniae (pneumococcal) conjugate vaccines have impressive track records in not only preventing life threatening diseases caused by these bacteria, but also reducing antibiotic use and AMR. This paper will describe the drivers of antibiotic use and subsequent development of AMR; it will make the case how existing vaccines are already participating in combatting AMR, describe future prospects for the role of new vaccines in development to reduce AMR, and highlight challenges associated with future vaccine development to combat AMR.
Collapse
Affiliation(s)
- Kathrin U Jansen
- a Pfizer Vaccine Research and Development , Pearl River , NY , USA
| | | |
Collapse
|
32
|
Misra N, Wines TF, Knopp CL, Hermann R, Bond L, Mitchell B, McGuire MA, Tinker JK. Immunogenicity of a Staphylococcus aureus-cholera toxin A 2/B vaccine for bovine mastitis. Vaccine 2018; 36:3513-3521. [PMID: 29739718 DOI: 10.1016/j.vaccine.2018.04.067] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 04/09/2018] [Accepted: 04/22/2018] [Indexed: 01/10/2023]
Abstract
Staphylococcus aureus causes a chronic, contagious disease of the udder, or mastitis, in dairy cows. This infection is often refractory to antibiotic treatment, and has a significant economic impact on milk production worldwide. An effective vaccine to prevent S. aureus mastitis would improve animal health, reduce antibiotic dependence and inform human vaccine approaches. The iron-regulated surface determinant A (IsdA) and clumping factor A (ClfA) are conserved S. aureus extracellular-matrix adhesins and target vaccine antigens. Here we report the results of two bovine immunogenicity trials using purified IsdA and ClfA-cholera toxin A2/B chimeras (IsdA-CTA2/B and ClfA-CTA2/B). Cows were intranasally inoculated with IsdA-CTA2/B + ClfA-CTA2/B at dry off and followed for 70 days. Trial 1 utilized three groups with one or two booster doses at a total concentration of 600 or 900 μg. Trial 2 utilized two groups with one booster at a total concentration of 1200 μg. Humoral immune responses in serum and milk were examined by ELISA. Responses in serum were significant between groups and provide evidence of antigen-specific IgG induction after vaccination in both trials. Cellular proliferation was detected by flow cytometry using antigen-stimulated PBMCs from day 60 of Trial 2 and revealed an increase in CD4+ T cells from vaccinated cows. IsdA and ClfA stimulation induced IL-4 expression, but not IFN-γ or IL-17, in PBMCs from day 60 as determined by cytokine expression analysis. Opsonophagocytosis of S. aureus confirmed the functional in vitro activity of anti-IsdA antibodies from Trial 2 serum and milk. The vaccine was well tolerated and safe, and results support the potential of mucosally-delivered CTA2/B chimeras to protect cows from mastitis caused by S. aureus.
Collapse
Affiliation(s)
- N Misra
- Biomolecular Ph.D. Program, Boise State University, Boise, ID, USA
| | - T F Wines
- Department of Biological Sciences, Boise State University, Boise, ID, USA
| | - C L Knopp
- Department of Biological Sciences, Boise State University, Boise, ID, USA
| | - R Hermann
- Department of Biological Sciences, Boise State University, Boise, ID, USA; Biomolecular Ph.D. Program, Boise State University, Boise, ID, USA
| | - L Bond
- Biomolecular Research Center, Boise State University, Boise, ID, USA
| | - B Mitchell
- DairyTeam Nutrition and Veterinary Consulting, Boise, ID, USA
| | - M A McGuire
- Department of Animal and Veterinary Science, University of Idaho, Moscow, ID, USA
| | - J K Tinker
- Department of Biological Sciences, Boise State University, Boise, ID, USA; Biomolecular Ph.D. Program, Boise State University, Boise, ID, USA.
| |
Collapse
|
33
|
Evaluation of the humoral immune response to a multicomponent recombinant vaccine against S. aureus in healthy pregnant heifers. Vet J 2018; 235:47-53. [DOI: 10.1016/j.tvjl.2018.03.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 03/12/2018] [Accepted: 03/16/2018] [Indexed: 11/19/2022]
|
34
|
Abstract
PURPOSE OF REVIEW Multidrug resistance of bacterial pathogens has confronted physicians around the world with the threat of inefficacy of the antibiotic regime, which is particularly important for patients with sepsis. Antibiotic resistance has revived search for alternative nonantibiotic strategies. Among them, prophylaxis by vaccination is an appealing concept. RECENT FINDINGS This review provides a compact overview on available vaccines against community-acquired pathogens such as pneumococci (in synergy with influenza) and meningococci and provides an overview on the ongoing developments of vaccines targeting typical nosocomial pathogens such as Clostridium difficile, Staphylococcus aureus, Acintetobacter baumannii, Klebsiella pneumonia, and Pseudomonas aeruginosa. SUMMARY The effects achieved by some conjugated vaccines (e.g. against Haemophilus influenzae B and Streptococcus pneumoniae) are encouraging. Their widespread use has resulted in a decrease or almost elimination of invasive diseases by the covered pneumococcal serotypes or Haemophilus influenzae B, respectively. These vaccines confer not only individual protection but also exploit herd protection effects. However, a multitude of failures reflects the obstacles on the way to effective and well tolerated bacterial vaccines. Regional differences in strain prevalence and variability of antigens that limit cross-protectivity remain major obstacles. However, promising candidates are in clinical development.
Collapse
|
35
|
Affiliation(s)
- Dane Parker
- a Department of Pediatrics , Columbia University , New York , NY , USA
| |
Collapse
|
36
|
Furukawa M, Yoneyama H, Hata E, Iwano H, Higuchi H, Ando T, Sato M, Hayashi T, Kiku Y, Nagasawa Y, Niimi K, Usami K, Ito K, Watanabe K, Nochi T, Aso H. Identification of a novel mechanism of action of bovine IgG antibodies specific for Staphylococcus aureus. Vet Res 2018; 49:22. [PMID: 29482613 PMCID: PMC5828400 DOI: 10.1186/s13567-018-0517-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 01/21/2018] [Indexed: 11/12/2022] Open
Abstract
Staphylococcus aureus is a major pathogen that causes subclinical mastitis associated with huge economic losses to the dairy industry. A few vaccines for bovine mastitis are available, and they are expected to induce the production of S. aureus-specific antibodies that prevent bacterial adherence to host cells or promote opsonization by phagocytes. However, the efficacy of such vaccines are still under debate; therefore, further research focusing on improving the current vaccines by seeking additional mechanisms of action is required to reduce economic losses due to mastitis in the dairy industry. Here, we generated S. aureus-specific bovine IgG antibodies (anti-S. aureus) that directly inhibited bacterial growth in vitro. Inhibition depended on specificity for anti-S. aureus, not the interaction between Protein A and the fragment crystallizable region of the IgG antibodies or bacterial agglutination. An in vitro culture study using S. aureus strain JE2 and its deletion mutant JE2ΔSrtA, which lacks the gene encoding sortase A, revealed that the effect of anti-S. aureus was sortase-A-independent. Sortase A is involved in the synthesis of cell-wall-associated proteins. Thus, other surface molecules, such as membrane proteins, cell surface polysaccharides, or both, may trigger the inhibition of bacterial growth by anti-S. aureus. Together, our findings contribute insights into developing new strategies to further improve the available mastitis vaccine by designing a novel antigen on the surface of S. aureus to induce inhibitory signals that prevent bacterial growth.
Collapse
Affiliation(s)
- Mutsumi Furukawa
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Sendai, Miyagi, 980-0845, Japan
| | - Hiroshi Yoneyama
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Sendai, Miyagi, 980-0845, Japan
| | - Eiji Hata
- National Institute of Animal Health, National Agriculture and Food Research Organization, Sapporo, Hokkaido, 062-0045, Japan
| | - Hidetomo Iwano
- School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Hokkaido, 069-8501, Japan
| | - Hidetoshi Higuchi
- School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Hokkaido, 069-8501, Japan
| | - Tasuke Ando
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Sendai, Miyagi, 980-0845, Japan
| | - Mika Sato
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Sendai, Miyagi, 980-0845, Japan
| | - Tomohito Hayashi
- National Institute of Animal Health, National Agriculture and Food Research Organization, Sapporo, Hokkaido, 062-0045, Japan
| | - Yoshio Kiku
- National Institute of Animal Health, National Agriculture and Food Research Organization, Sapporo, Hokkaido, 062-0045, Japan
| | - Yuya Nagasawa
- National Institute of Animal Health, National Agriculture and Food Research Organization, Sapporo, Hokkaido, 062-0045, Japan
| | - Kanae Niimi
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Sendai, Miyagi, 980-0845, Japan
| | - Katsuki Usami
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Sendai, Miyagi, 980-0845, Japan
| | - Kumiko Ito
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Sendai, Miyagi, 980-0845, Japan
| | - Kouichi Watanabe
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Sendai, Miyagi, 980-0845, Japan
| | - Tomonori Nochi
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Sendai, Miyagi, 980-0845, Japan. .,International Research and Development Center for Mucosal Vaccine, The University of Tokyo, Tokyo, 108-8639, Japan.
| | - Hisashi Aso
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Sendai, Miyagi, 980-0845, Japan
| |
Collapse
|
37
|
Li T, Huang M, Song Z, Zhang H, Chen C. Biological characteristics and conjugated antigens of ClfA A-FnBPA and CP5 in Staphylococcus aureus. CANADIAN JOURNAL OF VETERINARY RESEARCH = REVUE CANADIENNE DE RECHERCHE VETERINAIRE 2018; 82:48-54. [PMID: 29382968 PMCID: PMC5764038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 02/27/2017] [Indexed: 06/07/2023]
Abstract
To obtain immunogenic conjugate antigens, adipic acid dihydrazide (ADH), as a bridge, and 1-ethyl-3-(3-dimethylaminopropyl) carbodiimidehydrochloride (EDAC), as a coupling agent, were used to conjugate the purified fusion protein, clumping factor A-fibronectin binding protein ClfA A-FnBPA, and type 5 capsular polysaccharide (CP5). The conjugates were mixed with an adjuvant, and mice were immunized 3 times and challenged with Staphylococcus aureus 1 week later. Antibody titers were determined by indirect enzyme-linked immunosorbent assay (ELISA). At 14 days after the first immunization, antibodies against the purified protein and conjugate were detected; after 28 days, antibody levels increased; and a week after the third immunization, antibody levels continued to increase. However, the conjugate antibody titers were higher than those of the purified protein during the study, and no IgG antibodies against purified CP5 were detected during the entire experiment. The protection rate increased to 90% in the conjugate group, indicating that the conjugate imparts a relatively higher protective efficacy than the purified protein and purified CP5.
Collapse
Affiliation(s)
| | | | | | | | - Chuangfu Chen
- Address all correspondence to Dr. Chuangfu Chen; telephone: +86-0993-2058002; fax: +86-0993-2058002; e-mail:
| |
Collapse
|
38
|
Multidrug-Resistant Organisms: A Review of Transmission and Control. JOURNAL OF ACUTE CARE PHYSICAL THERAPY 2018. [DOI: 10.1097/jat.0000000000000072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
39
|
Scully IL, Pavliak V, Timofeyeva Y, Liu Y, Singer C, Anderson AS. O-Acetylation is essential for functional antibody generation against Staphylococcus aureus capsular polysaccharide. Hum Vaccin Immunother 2017; 14:81-84. [PMID: 29182428 PMCID: PMC5791590 DOI: 10.1080/21645515.2017.1386360] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Staphylococcus aureus produces an antiphagocytic polysaccharide capsule to evade neutrophil-mediated killing. Many vaccines against encapsulated bacterial pathogens require generation of functional anti-capsular antibodies to mediate protection against infection and disease. Here it is shown that the generation of such antibody responses to S. aureus in vivo and in vitro requires the presence of O-acetyl modifications on the capsular polysaccharides. O-acetylation of S. aureus capsular polysaccharide therefore should be monitored carefully during vaccine development and production. This finding may provide additional insight into the previous failure of a S. aureus capsular polysaccharide conjugate vaccine.
Collapse
Affiliation(s)
| | | | | | - Yongdong Liu
- a Pfizer Vaccine Research , Pearl River , NY , USA
| | | | | |
Collapse
|
40
|
Immunization Intraductal Fibronectin and Whole Cells of Staphylococcus aureus, Escherichia coli and Streptococcus agalactiae, in Cows to Dry in Puebla-Mexico. JOURNAL OF PURE AND APPLIED MICROBIOLOGY 2017. [DOI: 10.22207/jpam.11.3.20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
41
|
Yu W, Wang L, Wang M, Liu S, Li W, Wang X, Li X, Yu S, Yao D, Ma J, Yu L, Chen J, Feng Z, Cui Y. Identification and characterization of CD4 + T cell epitopes on manganese transport protein C of Staphylococcus aureus. Microb Pathog 2017; 112:30-37. [PMID: 28942173 DOI: 10.1016/j.micpath.2017.09.045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 08/26/2017] [Accepted: 09/19/2017] [Indexed: 11/29/2022]
Abstract
Manganese transport protein C (MntC) of Staphylococcus aureus represents an excellent vaccine-candidate antigen. The important role of CD4+ T cells in effective immunity against S. aureus infection was shown; however, CD4+ T cell-specific epitopes on S. aureus MntC have not been well identified. Here, we used bioinformatics prediction algorithms to evaluate and identify nine candidate epitopes within MntC. Our results showed that peptide M8 emulsified in Freund's adjuvant induced a much higher cell-proliferation rate as compared with controls. Additionally, CD4+ T cells stimulated with peptide M8 secreted significantly higher levels of interferon-γ and interleukin-17A. These results suggested that peptide M8 represented an H-2d (I-E)-restricted Th17-specific epitope.
Collapse
Affiliation(s)
- Wei Yu
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang Province, China
| | - Lizi Wang
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang Province, China
| | - Mengyao Wang
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang Province, China
| | - Shuo Liu
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang Province, China
| | - Wanyu Li
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang Province, China
| | - Xintong Wang
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang Province, China
| | - Xiaoting Li
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang Province, China
| | - Simiao Yu
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang Province, China
| | - Di Yao
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang Province, China
| | - Jinzhu Ma
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang Province, China
| | - Liquan Yu
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang Province, China
| | - Jing Chen
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang Province, China
| | - Zhenyue Feng
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang Province, China
| | - Yudong Cui
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang Province, China.
| |
Collapse
|
42
|
Mohamed N, Wang MY, Le Huec JC, Liljenqvist U, Scully IL, Baber J, Begier E, Jansen KU, Gurtman A, Anderson AS. Vaccine development to prevent Staphylococcus aureus surgical-site infections. Br J Surg 2017; 104:e41-e54. [PMID: 28121039 DOI: 10.1002/bjs.10454] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 10/27/2016] [Accepted: 11/06/2016] [Indexed: 12/12/2022]
Abstract
BACKGROUND Staphylococcus aureus surgical-site infections (SSIs) are a major cause of poor health outcomes, including mortality, across surgical specialties. Despite current advances as a result of preventive interventions, the disease burden of S. aureus SSI remains high, and increasing antibiotic resistance continues to be a concern. Prophylactic S. aureus vaccines may represent an opportunity to prevent SSI. METHODS A review of SSI pathophysiology was undertaken in the context of evaluating new approaches to developing a prophylactic vaccine to prevent S. aureus SSI. RESULTS A prophylactic vaccine ideally would provide protective immunity at the time of the surgical incision to prevent initiation and progression of infection. Although the pathogenicity of S. aureus is attributed to many virulence factors, previous attempts to develop S. aureus vaccines targeted only a single virulence mechanism. The field has now moved towards multiple-antigen vaccine strategies, and promising results have been observed in early-phase clinical studies that supported the recent initiation of an efficacy trial to prevent SSI. CONCLUSION There is an unmet medical need for novel S. aureus SSI prevention measures. Advances in understanding of S. aureus SSI pathophysiology could lead to the development of effective and safe prophylactic multiple-antigen vaccines to prevent S. aureus SSI.
Collapse
Affiliation(s)
- N Mohamed
- Pfizer Vaccine Research and Development, Pearl River, New York, USA
| | - M Y Wang
- Departments of Neurological Surgery and Rehabilitation Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - J-C Le Huec
- Spine Unit 2, Surgical Research Laboratory, Bordeaux University Hospital, Bordeaux, France
| | - U Liljenqvist
- Department of Spine Surgery, St Franziskus Hospital Muenster, Münster, Germany
| | - I L Scully
- Pfizer Vaccine Research and Development, Pearl River, New York, USA
| | - J Baber
- Pfizer Vaccine Clinical Research and Development, Sydney, New South Wales, Australia
| | - E Begier
- Pfizer Vaccine Clinical Research and Development, Pearl River, New York, USA
| | - K U Jansen
- Pfizer Vaccine Research and Development, Pearl River, New York, USA
| | - A Gurtman
- Pfizer Vaccine Clinical Research and Development, Pearl River, New York, USA
| | - A S Anderson
- Pfizer Vaccine Research and Development, Pearl River, New York, USA
| |
Collapse
|
43
|
Begier E, Seiden DJ, Patton M, Zito E, Severs J, Cooper D, Eiden J, Gruber WC, Jansen KU, Anderson AS, Gurtman A. SA4Ag, a 4-antigen Staphylococcus aureus vaccine, rapidly induces high levels of bacteria-killing antibodies. Vaccine 2017; 35:1132-1139. [PMID: 28143674 DOI: 10.1016/j.vaccine.2017.01.024] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 12/12/2016] [Accepted: 01/10/2017] [Indexed: 10/20/2022]
Abstract
BACKGROUND Staphylococcus aureus is a leading cause of healthcare-associated infections. No preventive vaccine is currently licensed. SA4Ag is an investigational 4-antigen S. aureus vaccine, composed of capsular polysaccharide conjugates of serotypes 5 and 8 (CP5 and CP8), recombinant surface protein clumping factor A (rmClfA), and recombinant manganese transporter protein C (rMntC). This Phase 1 study aimed to confirm the safety and immunogenicity of SA4Ag produced by the final manufacturing process before efficacy study initiation in a surgical population. METHODS Healthy adults (18-<65years) received one intramuscular SA4Ag injection. Serum functional antibodies were measured at baseline and Day 29 post-vaccination. An opsonophagocytic activity (OPA) assay measured the ability of vaccine-induced antibodies to CP5 and CP8 to kill S. aureus clinical isolates. For MntC and ClfA, antigen-specific immunogenicity was assessed via competitive Luminex® immunoassay (cLIA) and via fibrinogen-binding inhibition (FBI) assay for ClfA only. Reactogenicity and adverse event data were collected. RESULTS One hundred participants were vaccinated. SA4Ag was well tolerated, with a satisfactory safety profile. On Day 29, OPA geometric mean titers (GMTs) were 45,738 (CP5, 95% CI: 38,078-54,940) and 42,652 (CP8, 95% CI: 32,792-55,477), consistent with 69.2- and 28.9-fold rises in bacteria-killing antibodies, respectively; cLIA GMTs were 2064.4 (MntC, 95% CI: 1518.2-2807.0) and 3081.4 (ClfA, 95% CI: 2422.2-3920.0), consistent with 19.6- and 12.3-fold rises, respectively. Similar to cLIA results, ClfA FBI titers rose 11.0-fold (GMT: 672.2, 95% CI: 499.8-904.2). The vast majority of participants achieved the pre-defined biologically relevant thresholds: CP5: 100%; CP8: 97.9%, ClfA: 87.8%; and MntC 96.9%. CONCLUSIONS SA4Ag was safe, well tolerated, and rapidly induced high levels of bacteria-killing antibodies in healthy adults. A Phase 2B efficacy trial in adults (18-85years) undergoing elective spinal fusion is ongoing to assess SA4Ag's ability to prevent postoperative invasive surgical site and bloodstream infections caused by S. aureus. Clinicaltrials.gov Identifier: NCT02364596.
Collapse
Affiliation(s)
- Elizabeth Begier
- Pfizer Vaccine Clinical Research & Development, Pearl River, NY, USA.
| | | | - Michael Patton
- Pfizer Vaccine Clinical Research & Development, Maidenhead, UK
| | | | - Joseph Severs
- Pfizer Vaccine Clinical Research & Development, Pearl River, NY, USA
| | - David Cooper
- Vaccine Research & Development, Pearl River, NY, USA
| | - Joseph Eiden
- Pfizer Vaccine Clinical Research & Development, Pearl River, NY, USA
| | - William C Gruber
- Pfizer Vaccine Clinical Research & Development, Pearl River, NY, USA
| | | | | | - Alejandra Gurtman
- Pfizer Vaccine Clinical Research & Development, Pearl River, NY, USA
| |
Collapse
|
44
|
Safety, tolerability, and immunogenicity of a 4-antigen Staphylococcus aureus vaccine (SA4Ag): Results from a first-in-human randomised, placebo-controlled phase 1/2 study. Vaccine 2016; 35:375-384. [PMID: 27916408 DOI: 10.1016/j.vaccine.2016.11.010] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 11/01/2016] [Accepted: 11/02/2016] [Indexed: 11/21/2022]
Abstract
BACKGROUND A prophylactic Staphylococcus aureus four-antigen vaccine (SA4Ag) is under development for prevention of invasive S. aureus disease. A preliminary S. aureus three-antigen vaccine (SA3Ag) was reformulated to include a novel manganese transporter protein (MntC or rP305A). This study describes the first-in-human dose-finding, safety, and immunogenicity results for SA4Ag. METHODS In this double-blind, sponsor-unblind, placebo-controlled, phase 1/2 study, 454 healthy adults aged 18-64years were randomised to receive a single dose of one of three formulations of SA4Ag with escalating dose levels of rP305A or placebo. Functional immune responses were measured using opsonophagocytic activity (OPA) killing and fibrinogen-binding inhibition (FBI) assays; antigen-specific immunogenicity was assessed using a four-plex competitive Luminex® immunoassay (cLIA). RESULTS A high proportion of SA4Ag recipients met the pre-defined antibody thresholds for each antigen at Day 29. A substantial and dose-level dependent immune response was observed for rP305A, with up to 18-fold rises in cLIA titres at Day 29. Robust functional responses were demonstrated, with >80-fold and >20-fold rises in OPA assay titres at Day 29 using S. aureus strains expressing capsular polysaccharide serotypes 5 and 8, respectively. Durable antibody responses were observed through month 12, gradually waning from peak levels achieved by days 11-15. SA4Ag was well tolerated, and no vaccine-related serious adverse events were reported. CONCLUSIONS Single-dose vaccination of SA4Ag in healthy adults aged 18-64years safely induced rapid and robust functional immune responses that were durable through month 12, supporting further development of this vaccine. TRIAL REGISTRATION NUMBER NCT01364571.
Collapse
|
45
|
Missiakas D, Schneewind O. Staphylococcus aureus vaccines: Deviating from the carol. J Exp Med 2016; 213:1645-53. [PMID: 27526714 PMCID: PMC4995089 DOI: 10.1084/jem.20160569] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 07/29/2016] [Indexed: 12/27/2022] Open
Abstract
Staphylococcus aureus, a commensal of the human nasopharynx and skin, also causes invasive disease, most frequently skin and soft tissue infections. Invasive disease caused by drug-resistant strains, designated MRSA (methicillin-resistant S. aureus), is associated with failure of antibiotic therapy and elevated mortality. Here we review polysaccharide-conjugate and subunit vaccines that were designed to prevent S. aureus infection in patients at risk of bacteremia or surgical wound infection but failed to reach their clinical endpoints. We also discuss vaccines with ongoing trials for combinations of polysaccharide-conjugates and subunits. S. aureus colonization and invasive disease are not associated with the development of protective immune responses, which is attributable to a large spectrum of immune evasion factors. Two evasive strategies, assembly of protective fibrin shields via coagulases and protein A-mediated B cell superantigen activity, are discussed as possible vaccine targets. Although correlates for protective immunity are not yet known, opsonophagocytic killing of staphylococci by phagocytic cells offers opportunities to establish such criteria.
Collapse
Affiliation(s)
| | - Olaf Schneewind
- Department of Microbiology, University of Chicago, Chicago, IL 60637
| |
Collapse
|
46
|
Giersing BK, Dastgheyb SS, Modjarrad K, Moorthy V. Status of vaccine research and development of vaccines for Staphylococcus aureus. Vaccine 2016; 34:2962-2966. [DOI: 10.1016/j.vaccine.2016.03.110] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 03/30/2016] [Indexed: 01/08/2023]
|
47
|
Dayan GH, Mohamed N, Scully IL, Cooper D, Begier E, Eiden J, Jansen KU, Gurtman A, Anderson AS. Staphylococcus aureus: the current state of disease, pathophysiology and strategies for prevention. Expert Rev Vaccines 2016; 15:1373-1392. [PMID: 27118628 DOI: 10.1080/14760584.2016.1179583] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Staphylococcus aureus is both a commensal organism and also an important opportunistic human pathogen, causing a variety of community and hospital-associated pathologies, such as bacteremia-sepsis, endocarditis, pneumonia, osteomyelitis, arthritis and skin diseases. The resurgence of S. aureus during the last decade in many settings has been facilitated not only by bacterial antibiotic resistance mechanisms but also by the emergence of new S. aureus clonal types with increased expression of virulence factors and the capacity to neutralize the host immune response. Prevention of the spread of S. aureus infection relies on the use of contact precautions and adequate procedures for infection control that so far have not been fully effective. Prevention using a prophylactic vaccine would complement these processes, having the potential to bring additional, significant progress toward decreasing invasive disease due to S. aureus.
Collapse
Affiliation(s)
- Gustavo H Dayan
- a Pfizer Vaccine Research and Development , Pearl River , NY , USA
| | - Naglaa Mohamed
- a Pfizer Vaccine Research and Development , Pearl River , NY , USA
| | - Ingrid L Scully
- a Pfizer Vaccine Research and Development , Pearl River , NY , USA
| | - David Cooper
- a Pfizer Vaccine Research and Development , Pearl River , NY , USA
| | - Elizabeth Begier
- a Pfizer Vaccine Research and Development , Pearl River , NY , USA
| | - Joseph Eiden
- a Pfizer Vaccine Research and Development , Pearl River , NY , USA
| | - Kathrin U Jansen
- a Pfizer Vaccine Research and Development , Pearl River , NY , USA
| | | | | |
Collapse
|
48
|
Yang L, Cai C, Feng Q, Shi Y, Zuo Q, Yang H, Jing H, Wei C, Zhuang Y, Zou Q, Zeng H. Protective efficacy of the chimeric Staphylococcus aureus vaccine candidate IC in sepsis and pneumonia models. Sci Rep 2016; 6:20929. [PMID: 26865417 PMCID: PMC4750066 DOI: 10.1038/srep20929] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 01/13/2016] [Indexed: 12/16/2022] Open
Abstract
Staphylococcus aureus causes serious sepsis and necrotic pneumonia worldwide. Due to the spread of multidrug-resistant strains, developing an effective vaccine is the most promising method for combating S. aureus infection. In this study, based on the immune-dominant areas of the iron surface determinant B (IsdB) and clumping factor A (ClfA), we designed the novel chimeric vaccine IsdB151-277ClfA33-213 (IC). IC formulated with the AlPO4 adjuvant induced higher protection in an S. aureus sepsis model compared with the single components alone and showed broad immune protection against several clinical S. aureus isolates. Immunisation with IC induced strong antibody responses. The protective effect of antibodies was demonstrated through the opsonophagocytic assay (OPA) and passive immunisation experiment. Moreover, this new chimeric vaccine induced Th1/Th17-skewed cellular immune responses based on cytokine profiles and CD4+ T cell stimulation tests. Neutralisation of IL-17A alone (but not IFN-γ) resulted in a significant decrease in vaccine immune protection. Finally, we found that IC showed protective efficacy in a pneumonia model. Taken together, these data provide evidence that IC is a potentially promising vaccine candidate for combating S. aureus sepsis and pneumonia.
Collapse
Affiliation(s)
- Liuyang Yang
- National Engineering Research Center of Immunological Products &Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, P.R. China
| | - Changzhi Cai
- National Engineering Research Center of Immunological Products &Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, P.R. China
| | - Qiang Feng
- Department of Biological and Chemical Engineering, Chongqing University of Education, Chongqing 400067, P.R. China
| | - Yun Shi
- National Engineering Research Center of Immunological Products &Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, P.R. China
| | - Qianfei Zuo
- National Engineering Research Center of Immunological Products &Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, P.R. China
| | - Huijie Yang
- National Engineering Research Center of Immunological Products &Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, P.R. China
| | - Haiming Jing
- National Engineering Research Center of Immunological Products &Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, P.R. China
| | - Chao Wei
- National Engineering Research Center of Immunological Products &Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, P.R. China
| | - Yuan Zhuang
- National Engineering Research Center of Immunological Products &Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, P.R. China
| | - Quanming Zou
- National Engineering Research Center of Immunological Products &Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, P.R. China
| | - Hao Zeng
- National Engineering Research Center of Immunological Products &Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, P.R. China
| |
Collapse
|
49
|
Delfani S, Mohabati Mobarez A, Imani Fooladi AA, Amani J, Emaneini M. Protection of mice against Staphylococcus aureus infection by a recombinant protein ClfA-IsdB-Hlg as a vaccine candidate. Med Microbiol Immunol 2016; 205:47-55. [PMID: 26155981 DOI: 10.1007/s00430-015-0425-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 06/24/2015] [Indexed: 10/23/2022]
Abstract
Staphylococcus aureus is one of the most important causes of nosocomial infections. An effective vaccine to prevent S. aureus infections is urgently required due to the dramatic increase in the number of antibiotic-resistant strains. In this report, we evaluated a newly recombinant protein composed of selected antigenic regions of clumping factor A (ClfA), iron surface determinant B (IsdB) and gamma hemolysin B (HlgB) of S. aureus and sequence coding for hydrophobic linkers between three domains. The recombinant gene was constructed in pET-28a (+) and expressed in Escherichia coli BL21. In addition, sequence coding for a His(6)-tag was added followed by a hybrid procedure of nickel chelate protein purification. Immunization of BALB/c mice with the recombinant protein ClfA-IsdB-Hlg evoked antigen-specific antibodies that could opsonize S. aureus cells, enhancing in vitro phagocytosis by macrophages. Vaccination with the recombinant protein also reduced the bacterial load recovered from mice spleen samples and increased survival following the intraperitoneal challenge with pathogenic S. aureus compared to the control mice. Our results showed that the recombinant protein ClfA-IsdB-Hlg is a promising vaccine candidate for the prevention of S. aureus bacteremia infections.
Collapse
Affiliation(s)
- Somayeh Delfani
- Department of Bacteriology, Faculty of Medical Sciences, Tarbiat Modares University, P.O. Box 14111-115, Tehran, Iran
| | - Ashraf Mohabati Mobarez
- Department of Bacteriology, Faculty of Medical Sciences, Tarbiat Modares University, P.O. Box 14111-115, Tehran, Iran.
| | - Abbas Ali Imani Fooladi
- Applied Microbiology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Jafar Amani
- Applied Microbiology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mohammad Emaneini
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
50
|
Weaver LG, Singh Y, Burn PL, Blanchfield JT. The synthesis and ring-opening metathesis polymerization of glycomonomers. RSC Adv 2016. [DOI: 10.1039/c5ra25732h] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The synthesis of a series of short poly(norbornene)s displaying pendant disaccharides is reported.
Collapse
Affiliation(s)
- Lucy G. Weaver
- The School of Chemistry & Molecular Biosciences
- University of Queensland
- St Lucia
- Australia
| | - Yogendra Singh
- The School of Chemistry & Molecular Biosciences
- University of Queensland
- St Lucia
- Australia
| | - Paul L. Burn
- Centre for Organic Photonics & Electronics
- University of Queensland
- St Lucia
- Australia
| | - Joanne T. Blanchfield
- The School of Chemistry & Molecular Biosciences
- University of Queensland
- St Lucia
- Australia
| |
Collapse
|