1
|
Sorieul C, Dolce M, Romano MR, Codée J, Adamo R. Glycoconjugate vaccines against antimicrobial resistant pathogens. Expert Rev Vaccines 2023; 22:1055-1078. [PMID: 37902243 DOI: 10.1080/14760584.2023.2274955] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 10/20/2023] [Indexed: 10/31/2023]
Abstract
INTRODUCTION Antimicrobial resistance (AMR) is responsible for the death of millions worldwide and stands as a major threat to our healthcare systems, which are heavily reliant on antibiotics to fight bacterial infections. The development of vaccines against the main pathogens involved is urgently required as prevention remains essential against the rise of AMR. AREAS COVERED A systematic research review was conducted on MEDLINE database focusing on the six AMR pathogens defined as ESKAPE (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Escherichia coli), which are considered critical or high priority pathogens by the World Health Organization (WHO) and the Centers for Disease Control and Prevention (CDC). The analysis was intersecated with the terms carbohydrate, glycoconjugate, bioconjugate, glyconanoparticle, and multiple presenting antigen system vaccines. EXPERT OPINION Glycoconjugate vaccines have been successful in preventing meningitis and pneumoniae, and there are high expectations that they will play a key role in fighting AMR. We herein discuss the recent technological, preclinical, and clinical advances, as well as the challenges associated with the development of carbohydrate-based vaccines against leading AMR bacteria, with focus on the ESKAPE pathogens. The need of innovative clinical and regulatory approaches to tackle these targets is also highlighted.
Collapse
Affiliation(s)
- Charlotte Sorieul
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Marta Dolce
- GSK, Via Fiorentina 1, Siena, Italy
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | | | - Jeroen Codée
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | | |
Collapse
|
2
|
Amandine GB, Gagnaire J, Pelissier C, Philippe B, Elisabeth BN. Vaccines for healthcare associated infections without vaccine prevention to date. Vaccine X 2022; 11:100168. [PMID: 35600984 PMCID: PMC9118472 DOI: 10.1016/j.jvacx.2022.100168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 03/28/2022] [Accepted: 04/25/2022] [Indexed: 11/28/2022] Open
Abstract
In spite of the widespread implementation of preventive strategies, the prevalence of healthcare-associated infections (HAIs) remains high. The prevalence of multidrug resistant organisms is high in HAIs. In 2019, the World Health Organization retained antimicrobial resistance as one of the ten issues for global health. The development of vaccines may contribute to the fight against antimicrobial resistance to reduce the burden of HAIs. Staphylococcus aureus, Gram negative bacteria and Clostridium difficile are the most frequent pathogens reported in HAIs. Consequently, the development of vaccines against these pathogens is crucial. At this stage, the goal of obtaining effective vaccines against S.aureus and Gram negative bacteria has not yet been achieved. However, we can expect in the near future availability of a vaccine against C. difficile. In addition, identifying populations who may benefit from these vaccines is complex, as at-risk patients are not great responders to vaccines, or as vaccination may occur too late, when they are already confronted to the risk. Vaccinating healthcare workers (HCWs) against these pathogens may have an impact only if HCWs play a role in the transmission and in the pathogens acquisition in patients, if the vaccine is effective to reduce pathogens carriage and if vaccine coverage is sufficient to protect patients. Acceptance of these potential vaccines should be evaluated and addressed in patients and in HCWs.
Collapse
Affiliation(s)
- Gagneux-Brunon Amandine
- Inserm, CIC 1408, I-REIVAC, University Hospital of Saint-Etienne, 42055 Saint-Etienne, France.,CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Univ Lyon, Université Jean Monnet, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, F42023 Saint-Etienne, France.,Department of Infectious Diseases, University Hospital of Saint-Etienne, 42055 Saint-Etienne, France
| | - Julie Gagnaire
- Department of Infectious Diseases, University Hospital of Saint-Etienne, 42055 Saint-Etienne, France.,Infection Control Unit, University Hospital of Saint-Etienne, 42055 Saint-Etienne, France
| | - Carole Pelissier
- Occupational Health Department, University Hospital of Saint-Etienne, 42055 Saint-Etienne, France
| | - Berthelot Philippe
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Univ Lyon, Université Jean Monnet, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, F42023 Saint-Etienne, France.,Department of Infectious Diseases, University Hospital of Saint-Etienne, 42055 Saint-Etienne, France.,Infection Control Unit, University Hospital of Saint-Etienne, 42055 Saint-Etienne, France
| | - Botelho-Nevers Elisabeth
- Inserm, CIC 1408, I-REIVAC, University Hospital of Saint-Etienne, 42055 Saint-Etienne, France.,CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Univ Lyon, Université Jean Monnet, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, F42023 Saint-Etienne, France.,Department of Infectious Diseases, University Hospital of Saint-Etienne, 42055 Saint-Etienne, France
| |
Collapse
|
3
|
Staphylococcus aureus-A Known Opponent against Host Defense Mechanisms and Vaccine Development-Do We Still Have a Chance to Win? Int J Mol Sci 2022; 23:ijms23020948. [PMID: 35055134 PMCID: PMC8781139 DOI: 10.3390/ijms23020948] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/11/2022] [Accepted: 01/13/2022] [Indexed: 02/07/2023] Open
Abstract
The main purpose of this review is to present justification for the urgent need to implement specific prophylaxis of invasive Staphylococcus aureus infections. We emphasize the difficulties in achieving this goal due to numerous S. aureus virulence factors important for the process of infection and the remarkable ability of these bacteria to avoid host defense mechanisms. We precede these considerations with a brief overview of the global necessitiy to intensify the use of vaccines against other pathogens as well, particularly in light of an impasse in antibiotic therapy. Finally, we point out global trends in research into modern technologies used in the field of molecular microbiology to develop new vaccines. We focus on the vaccines designed to fight the infections caused by S. aureus, which are often resistant to the majority of available therapeutic options.
Collapse
|
4
|
Vaccine Composition Formulated with a Novel Lactobacillus-Derived Exopolysaccharides Adjuvant Provided High Protection against Staphylococcus aureus. Vaccines (Basel) 2021; 9:vaccines9070775. [PMID: 34358191 PMCID: PMC8310297 DOI: 10.3390/vaccines9070775] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/01/2021] [Accepted: 07/06/2021] [Indexed: 12/28/2022] Open
Abstract
A vaccine that effectively targets methicillin-resistant Staphylococcus aureus (MRSA) is urgently needed, and has been the focus of studies by numerous research groups, but with limited success to date. Recently, our team found that exopolysaccharides derived from probiotic Lactobacilluscasei strain WXD030 as an adjuvant-formulated OVA could upregulate IFN-γ and IL-17 expression in CD4+ T cells. In this study, we developed a vaccine (termed rMntC-EPS) composed of S. aureus antigen MntC and Lactobacillus casei exopolysaccharides, which conferred high levels of protection against S. aureus infection. Methods: Six–eight-week-old female mice were vaccinated with purified rMntC-EPS30. The immune protection function of rMntC-EPS30 was assessed by the protective effect of rMntC-EPS30 to S. aureus-induced pulmonary and cutaneous infection in mice, bacterial loads and H&E in injury site, and ELISA for inflammation-related cytokines. The protective mechanism of rMntC-EPS30 was assessed by ELISA for IgG in serum, cytokines in the spleen and lungs of vaccinated mice. In addition, flow cytometry was used for analyzing cellular immune response induced by rMntC-EPS30. For confirmation of our findings, three kinds of mice were used in this study: IL-17A knockout mice, IFN-γ knockout mice and TCRγ/δ knockout mice. Results: rMntC-EPS30 conferred up to 90% protection against S. aureus pulmonary infection and significantly reduced the abscess size in the S. aureus cutaneous model, with clearance of the pathogen. The rMntC-EPS vaccine could induce superior humoral immunity as well as significantly increase IL-17A and IFN-γ production. In addition, we found that rMntC-EPS vaccination induced robust Th 17/γδ T 17 primary and recall responses. Interestingly, this protective effect was distinctly reduced in the IL-17A knockout mice but not in IFN-γ knockout mice. Moreover, in TCRγ/δ knockout mice, rMntC-EPS vaccination neither increased IL-17A secretion nor provided effective protection against S. aureus infection. Conclusions: These data demonstrated that the rMntC formulated with a novel Lactobacillus-derived Exopolysaccharides adjuvant provided high protection against Staphylococcus aureus. The rMntC-EPS vaccine induced γδ T cells and IL-17A might play substantial roles in anti-S. aureus immunity. Our findings provided direct evidence that rMntC-EPS vaccine is a promising candidate for future clinical application against S. aureus-induced pulmonary and cutaneous infection.
Collapse
|
5
|
Scully IL, Timofeyeva Y, Illenberger A, Lu P, Liberator PA, Jansen KU, Anderson AS. Performance of a Four-Antigen Staphylococcus aureus Vaccine in Preclinical Models of Invasive S. aureus Disease. Microorganisms 2021; 9:microorganisms9010177. [PMID: 33467609 PMCID: PMC7830931 DOI: 10.3390/microorganisms9010177] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/07/2021] [Accepted: 01/07/2021] [Indexed: 01/16/2023] Open
Abstract
A Staphylococcus aureus four-antigen vaccine (SA4Ag) was designed for the prevention of invasive disease in surgical patients. The vaccine is composed of capsular polysaccharide type 5 and type 8 CRM197 conjugates, a clumping factor A mutant (Y338A-ClfA) and manganese transporter subunit C (MntC). S. aureus pathogenicity is characterized by an ability to rapidly adapt to the host environment during infection, which can progress from a local infection to sepsis and invasion of distant organs. To test the protective capacity of the SA4Ag vaccine against progressive disease stages of an invasive S. aureus infection, a deep tissue infection mouse model, a bacteremia mouse model, a pyelonephritis model, and a rat model of infectious endocarditis were utilized. SA4Ag vaccination significantly reduced the bacterial burden in deep tissue infection, in bacteremia, and in the pyelonephritis model. Complete prevention of infection was demonstrated in a clinically relevant endocarditis model. Unfortunately, these positive preclinical findings with SA4Ag did not prove the clinical utility of SA4Ag in the prevention of surgery-associated invasive S. aureus infection.
Collapse
|
6
|
|
7
|
Abstract
: The role of Staphylococcus aureus (SA) in the pathogenesis and management in atopic dermatitis is rapidly evolving. The modern understanding of SA in atopic dermatitis now includes an expanded array of virulence factors, the interplay of clonal and temporal shifts in SA populations, and host factors such as filaggrin and natural moisturizing factor. New, emerging therapies that focus on long-term, targeted elimination of SA colonization are currently under investigation (Br J Dermatol 2017;17(1)63-71). Herein, we discuss and review the latest staphylococcal and microbiome-modifying therapies including topical antibiotics, topical natural oil fatty acids, anti-SA vaccines, microbial transplantation, vitamin D supplementation, dupilumab and proposed future investigative directions.
Collapse
|
8
|
Visansirikul S, Kolodziej SA, Demchenko AV. Staphylococcus aureuscapsular polysaccharides: a structural and synthetic perspective. Org Biomol Chem 2020; 18:783-798. [DOI: 10.1039/c9ob02546d] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
This review surveys known structures of staphylococcal polysaccharides and summarizes all synthetic efforts to obtain these sequences.
Collapse
Affiliation(s)
- Satsawat Visansirikul
- Department of Chemistry and Biochemistry
- University of Missouri – St Louis
- One University Boulevard
- St Louis
- USA
| | | | - Alexei V. Demchenko
- Department of Chemistry and Biochemistry
- University of Missouri – St Louis
- One University Boulevard
- St Louis
- USA
| |
Collapse
|
9
|
Ma Y, Wang C, Li Y, Li J, Wan Q, Chen J, Tay FR, Niu L. Considerations and Caveats in Combating ESKAPE Pathogens against Nosocomial Infections. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1901872. [PMID: 31921562 PMCID: PMC6947519 DOI: 10.1002/advs.201901872] [Citation(s) in RCA: 161] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 10/04/2019] [Indexed: 05/19/2023]
Abstract
ESKAPE pathogens (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter species) are among the most common opportunistic pathogens in nosocomial infections. ESKAPE pathogens distinguish themselves from normal ones by developing a high level of antibiotic resistance that involves multiple mechanisms. Contemporary therapeutic strategies which are potential options in combating ESKAPE bacteria need further investigation. Herein, a broad overview of the antimicrobial research on ESKAPE pathogens over the past five years is provided with prospective clinical applications.
Collapse
Affiliation(s)
- Yu‐Xuan Ma
- State Key Laboratory of Military StomatologyNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical University145 Changle West RoadXi'anShaanxi710032P. R. China
| | - Chen‐Yu Wang
- State Key Laboratory of Military StomatologyNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical University145 Changle West RoadXi'anShaanxi710032P. R. China
| | - Yuan‐Yuan Li
- State Key Laboratory of Military StomatologyNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical University145 Changle West RoadXi'anShaanxi710032P. R. China
| | - Jing Li
- State Key Laboratory of Military StomatologyNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical University145 Changle West RoadXi'anShaanxi710032P. R. China
| | - Qian‐Qian Wan
- State Key Laboratory of Military StomatologyNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical University145 Changle West RoadXi'anShaanxi710032P. R. China
| | - Ji‐Hua Chen
- State Key Laboratory of Military StomatologyNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical University145 Changle West RoadXi'anShaanxi710032P. R. China
| | - Franklin R. Tay
- State Key Laboratory of Military StomatologyNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical University145 Changle West RoadXi'anShaanxi710032P. R. China
- The Graduate SchoolAugusta University1430, John Wesley Gilbert DriveAugustaGA30912‐1129USA
| | - Li‐Na Niu
- State Key Laboratory of Military StomatologyNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical University145 Changle West RoadXi'anShaanxi710032P. R. China
- The Graduate SchoolAugusta University1430, John Wesley Gilbert DriveAugustaGA30912‐1129USA
| |
Collapse
|
10
|
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
|
11
|
Behera A, Rai D, Kulkarni SS. Total Syntheses of Conjugation-Ready Trisaccharide Repeating Units of Pseudomonas aeruginosa O11 and Staphylococcus aureus Type 5 Capsular Polysaccharide for Vaccine Development. J Am Chem Soc 2019; 142:456-467. [DOI: 10.1021/jacs.9b11309] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Archanamayee Behera
- Department of Chemistry, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Diksha Rai
- Department of Chemistry, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Suvarn S. Kulkarni
- Department of Chemistry, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| |
Collapse
|
12
|
Marshall HS, Baber J, Richmond P, Nissen M, Shakib S, Kreiswirth BN, Zito ET, Severs J, Eiden J, Gruber W, Jansen KU, Jones CH, Anderson AS. S. aureus colonization in healthy Australian adults receiving an investigational S. aureus 3-antigen vaccine. J Infect 2019; 79:582-592. [PMID: 31585191 DOI: 10.1016/j.jinf.2019.09.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 09/09/2019] [Accepted: 09/27/2019] [Indexed: 12/14/2022]
Abstract
OBJECTIVES Assess Staphylococcus aureus (S. aureus) colonization in healthy Australian adults receiving an investigational S. aureus 3-antigen vaccine (SA3Ag). METHODS In this phase 1, double-blind, sponsor-unblinded study, participants were randomized to receive a single dose (1 of 3 dose levels) of SA3Ag or placebo and a booster dose or placebo at 6 months. S. aureus isolates from nasal, perineal, and oropharyngeal swabs before and through 12 months post-vaccination were identified. RESULTS Baseline S. aureus colonization prevalence was 30.6% (any site), with nasal carriage (27.0%) more common than oropharyngeal/perineal (3.2% each). Following initial vaccination (low-dose: 102; mid-dose: 101; high-dose: 101; placebo: 102) and booster (low-dose: 45; mid-dose: 44; high-dose: 27; placebo: 181), placebo and SA3Ag groups showed similar S. aureus carriage through 12 months. Most colonized participants (74.0%) were colonized by single spa types. Placebo and SA3Ag groups had similar persistence of colonization, with 19.6-30.7% due to single spa types. Acquisition was observed in mid- and high-dose recipients (∼20%) and low-dose and placebo recipients (∼12%). Vaccination resulted in substantial increases in antibodies to all 3 antigens, irrespective of carriage status. CONCLUSIONS Based on descriptive analyses of this small study, SA3Ag vaccination did not impact S. aureus acquisition or carriage. Carriage status did not impact antibody responses to SA3Ag.
Collapse
Affiliation(s)
- Helen S Marshall
- Vaccinology and Immunology Research Trials Unit, Women's and Children's Hospital and Robinson Research Institute and Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia.
| | - James Baber
- Pfizer Australia Pty Ltd, Sydney, NSW, Australia
| | - Peter Richmond
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, University of Western Australia Division of Paediatrics & Vaccine Trials Group, Perth, WA, Australia
| | - Michael Nissen
- Queensland Paediatric Infectious Diseases Laboratory, Children's Health Research Centre, University of Queensland, Queensland Children's Hospital, South Brisbane, Qld, Australia
| | - Sepehr Shakib
- Department of Clinical Pharmacology, University of Adelaide, Adelaide, SA, Australia
| | | | - Edward T Zito
- Pfizer Vaccine Research and Development, Collegeville, PA, USA
| | - Joseph Severs
- Pfizer Vaccine Research and Development, Pearl River, NY, USA
| | - Joseph Eiden
- Pfizer Vaccine Research and Development, Pearl River, NY, USA
| | - William Gruber
- Pfizer Vaccine Research and Development, Pearl River, NY, USA
| | | | - C Hal Jones
- Pfizer Vaccine Research and Development, Pearl River, NY, USA
| | | |
Collapse
|
13
|
Ansari S, Jha RK, Mishra SK, Tiwari BR, Asaad AM. Recent advances in Staphylococcus aureus infection: focus on vaccine development. Infect Drug Resist 2019; 12:1243-1255. [PMID: 31190912 PMCID: PMC6526327 DOI: 10.2147/idr.s175014] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 04/19/2019] [Indexed: 12/17/2022] Open
Abstract
Staphylococcus aureus normally colonizes the nasal cavity and pharynx. After breaching the normal habitat, the organism is able to cause a number of infections at any site of the body. The development of antibiotic resistance has created a global challenge for treating infections. Therefore, protection by vaccines may provide valuable measures. Currently, several vaccine candidates have been prepared which are either in preclinical phase or in early clinical phase, whereas several candidates have failed to show a protective efficacy in human subjects. Approaches have also been made in the development of monoclonal or polyclonal antibodies for passive immunization to protect from S. aureus infections. Therefore, in this review we have summarized the findings of recently published scientific literature to make a concise report.
Collapse
Affiliation(s)
- Shamshul Ansari
- Department of Microbiology, Chitwan Medical College and Teaching Hospital, Bharatpur, Chitwan, Nepal
| | - Rajesh Kumar Jha
- Department of Systems and Diseases (Pharmacology), Saba University School of Medicine, Saba, Dutch Caribbean
| | - Shyam Kumar Mishra
- Department of Microbiology, Maharajgunj Medical Campus, Institute of Medicine, Tribhuvan University, Kathmandu, Nepal
| | | | - Ahmed Morad Asaad
- Department of Microbiology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
14
|
Haynes J, Perry V, Benson E, Meeks A, Watts G, Watkins H, Braun R. In Depth Breadth Analyses of Human Blockade Responses to Norovirus and Response to Vaccination. Viruses 2019; 11:v11050392. [PMID: 31035476 PMCID: PMC6563306 DOI: 10.3390/v11050392] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 04/12/2019] [Accepted: 04/23/2019] [Indexed: 12/19/2022] Open
Abstract
To evaluate and understand the efficacy of vaccine candidates, supportive immunological measures are needed. Critical attributes for a norovirus vaccine are the strength and breadth of antibody responses against the many different genotypes. In the absence of suitable neutralization assays to test samples from vaccine clinical trials, blockade assays offer a method that can measure functional antibodies specific for many of the different norovirus strains. This paper describes development and optimization of blockade assays for an extended panel of 20 different norovirus strains that can provide robust and reliable data needed for vaccine assessment. The blockade assays were used to test a panel of human clinical samples taken before and after vaccination with the Takeda TAK-214 norovirus vaccine. Great variability was evident in the repertoire of blocking antibody responses prevaccination and postvaccination among individuals. Following vaccination with TAK-214, blocking antibody levels were enhanced across a wide spectrum of different genotypes. The results indicate that adults may have multiple exposures to norovirus and that the magnitude and breadth of the complex preexisting antibody response can be boosted and expanded by vaccination.
Collapse
Affiliation(s)
- Joel Haynes
- Vaccines Discovery Research, Takeda Pharmaceuticals, Cambridge, MA 02139, USA.
| | - Virginia Perry
- Vaccines Discovery Research, Takeda Pharmaceuticals, Cambridge, MA 02139, USA.
| | - Evelyn Benson
- Vaccines Discovery Research, Takeda Pharmaceuticals, Cambridge, MA 02139, USA.
| | - Alisa Meeks
- Vaccines Discovery Research, Takeda Pharmaceuticals, Cambridge, MA 02139, USA.
| | - Gayle Watts
- Vaccines Discovery Research, Takeda Pharmaceuticals, Cambridge, MA 02139, USA.
| | - Heather Watkins
- Vaccines Discovery Research, Takeda Pharmaceuticals, Cambridge, MA 02139, USA.
| | - Ralph Braun
- Vaccines Discovery Research, Takeda Pharmaceuticals, Cambridge, MA 02139, USA.
| |
Collapse
|
15
|
Connolly R, Denton MD, Humphreys H, McLoughlin RM. Would hemodialysis patients benefit from a Staphylococcus aureus vaccine? Kidney Int 2019; 95:518-525. [PMID: 30691691 DOI: 10.1016/j.kint.2018.10.023] [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: 07/06/2018] [Revised: 09/17/2018] [Accepted: 10/05/2018] [Indexed: 11/19/2022]
Abstract
Staphylococcus aureus bloodstream infection can have potentially catastrophic consequences for patients on hemodialysis. Consequently, an effective vaccine to prevent S aureus infection would have a significant influence on morbidity and mortality in this group. To date, however, efforts to develop a vaccine have been unsuccessful. Previous antibody-inducing vaccine candidates did not prevent or attenuate S aureus infection in clinical trials. Recent advances have helped to elucidate the role of specific T-cell subsets, notably T-helper cell 1 and T-helper cell 17, in the immune response to S aureus. These cells are essential for coordinating an effective phagocytic response via cytokine production, indirectly leading to destruction of the organism. It is now widely accepted that next-generation S aureus vaccines must also induce effective T-cell-mediated immunity. However, there remains a gap in our knowledge: how will an S aureus vaccine drive these responses in those patients most at risk? Given that patients on hemodialysis are an immunocompromised population, in particular with specific T-cell defects, including defects in T-helper cell subsets, this is likely to affect their ability to respond to an S aureus vaccine. We urgently need a better understanding of T-cell-mediated immunity in this cohort if an efficacious vaccine is ever to be realized for these patients.
Collapse
Affiliation(s)
- Roisin Connolly
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Dublin, Ireland; Department of Clinical Microbiology, Royal College of Surgeons in Ireland, Dublin, Ireland; Department of Microbiology, Beaumont Hospital, Dublin, Ireland
| | - Mark D Denton
- Beaumont Kidney Centre, Beaumont Hospital, Dublin, Ireland
| | - Hilary Humphreys
- Department of Clinical Microbiology, Royal College of Surgeons in Ireland, Dublin, Ireland; Department of Microbiology, Beaumont Hospital, Dublin, Ireland
| | - Rachel M McLoughlin
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Dublin, Ireland.
| |
Collapse
|
16
|
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
|
17
|
Micoli F, Costantino P, Adamo R. Potential targets for next generation antimicrobial glycoconjugate vaccines. FEMS Microbiol Rev 2018; 42:388-423. [PMID: 29547971 PMCID: PMC5995208 DOI: 10.1093/femsre/fuy011] [Citation(s) in RCA: 123] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 03/13/2018] [Indexed: 12/21/2022] Open
Abstract
Cell surface carbohydrates have been proven optimal targets for vaccine development. Conjugation of polysaccharides to a carrier protein triggers a T-cell-dependent immune response to the glycan moiety. Licensed glycoconjugate vaccines are produced by chemical conjugation of capsular polysaccharides to prevent meningitis caused by meningococcus, pneumococcus and Haemophilus influenzae type b. However, other classes of carbohydrates (O-antigens, exopolysaccharides, wall/teichoic acids) represent attractive targets for developing vaccines. Recent analysis from WHO/CHO underpins alarming concern toward antibiotic-resistant bacteria, such as the so called ESKAPE pathogens (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa and Enterobacter spp.) and additional pathogens such as Clostridium difficile and Group A Streptococcus. Fungal infections are also becoming increasingly invasive for immunocompromised patients or hospitalized individuals. Other emergencies could derive from bacteria which spread during environmental calamities (Vibrio cholerae) or with potential as bioterrorism weapons (Burkholderia pseudomallei and mallei, Francisella tularensis). Vaccination could aid reducing the use of broad-spectrum antibiotics and provide protection by herd immunity also to individuals who are not vaccinated. This review analyzes structural and functional differences of the polysaccharides exposed on the surface of emerging pathogenic bacteria, combined with medical need and technological feasibility of corresponding glycoconjugate vaccines.
Collapse
Affiliation(s)
- Francesca Micoli
- GSK Vaccines Institute for Global Health (GVGH), Via Fiorentina 1, 53100 Siena
| | | | | |
Collapse
|
18
|
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
|
19
|
Clowry J, Irvine AD, McLoughlin RM. Next-generation anti-Staphylococcus aureus vaccines: A potential new therapeutic option for atopic dermatitis? J Allergy Clin Immunol 2018; 143:78-81. [PMID: 30218675 DOI: 10.1016/j.jaci.2018.08.038] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 08/01/2018] [Accepted: 08/26/2018] [Indexed: 01/18/2023]
Affiliation(s)
- Julianne Clowry
- Pediatric Dermatology, Our Lady's Children's Hospital Crumlin, Dublin, Ireland; National Children's Research Centre, Our Lady's Children's Hospital Crumlin, Dublin, Ireland; Clinical Medicine, Trinity College Dublin, Dublin, Ireland
| | - Alan D Irvine
- Pediatric Dermatology, Our Lady's Children's Hospital Crumlin, Dublin, Ireland; National Children's Research Centre, Our Lady's Children's Hospital Crumlin, Dublin, Ireland; Clinical Medicine, Trinity College Dublin, Dublin, Ireland.
| | - Rachel M McLoughlin
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Dublin, Ireland.
| |
Collapse
|
20
|
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
|
21
|
Inoue M, Yonemura T, Baber J, Shoji Y, Aizawa M, Cooper D, Eiden J, Gruber WC, Jansen KU, Anderson AS, Gurtman A. Safety, tolerability, and immunogenicity of a novel 4-antigen Staphylococcus aureus vaccine (SA4Ag) in healthy Japanese adults. Hum Vaccin Immunother 2018; 14:2682-2691. [PMID: 30084709 PMCID: PMC6314418 DOI: 10.1080/21645515.2018.1496764] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 06/06/2018] [Accepted: 06/22/2018] [Indexed: 11/29/2022] Open
Abstract
A novel Staphylococcus aureus 4-antigen vaccine (SA4Ag) is under development, comprising capsular polysaccharide serotypes 5 and 8 (CP5 and CP8) conjugated to CRM197, recombinant protein clumping factor A (rmClfA), and recombinant manganese transporter protein C (MntC). We evaluated SA4Ag safety, tolerability, and immunogenicity in Japanese adults aged 20 to 64 and 65 to 85 years. A total of 136 healthy Japanese adults (68 per age group) were randomized 1:1 to receive single-dose SA4Ag or placebo intramuscularly (Day 1). Safety assessments included reactogenicity and adverse events. The ability of the vaccine to induce immune responses that are considered functional due to their ability to facilitate the killing of S. aureus or neutralize S. aureus virulence mechanisms was assessed using 5 different antigen-specific assays. SA4Ag was well tolerated in both age groups, with no safety concerns. At Day 29, > 85% of SA4Ag recipients in each age group achieved predefined thresholds for each antigen. Antibody geometric mean-fold rises from baseline to Day 29 in SA4Ag groups were: > 80 and > 30 for CP5 and CP8 (opsonophagocytic activity assay), > 10 for ClfA (fibrinogen-binding inhibition assay), and > 15 and > 7 for ClfA and MntC (competitive Luminex® immunoassay), respectively. Antibody titers decreased through Month 12 but remained well above baseline and placebo levels. SA4Ag had an acceptable safety profile and induced rapid and robust functional immune responses in both age groups. These results support ongoing development of SA4Ag for the prevention of invasive S. aureus disease in elective-surgery patients in Japan, North America, and Europe.
Collapse
Affiliation(s)
| | | | - James Baber
- Pfizer Vaccine Research and Development, Sydney, NSW, Australia
| | - Yasuko Shoji
- Pfizer Vaccine Research and Development, Tokyo, Japan
| | | | - David Cooper
- Pfizer Vaccine Research and Development, Pearl River, NY, USA
| | - Joseph Eiden
- Pfizer Vaccine Research and Development, Pearl River, NY, USA
| | | | | | | | | |
Collapse
|
22
|
Gagneux-Brunon A, Lucht F, Launay O, Berthelot P, Botelho-Nevers E. Vaccines for healthcare-associated infections: present, future, and expectations. Expert Rev Vaccines 2018; 17:421-433. [DOI: 10.1080/14760584.2018.1470507] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Amandine Gagneux-Brunon
- Inserm, CIC 1408, I-REIVAC, University Hospital of Saint-Etienne, Saint-Etienne, France
- GIMAP EA 3064, University of Lyon, Saint-Etienne, France
| | - Frédéric Lucht
- Inserm, CIC 1408, I-REIVAC, University Hospital of Saint-Etienne, Saint-Etienne, France
- GIMAP EA 3064, University of Lyon, Saint-Etienne, France
| | - Odile Launay
- Inserm CIC 1417, I-REIVAC, University of Paris-Descartes, University Hospital of Cochin-Broca-Hôtel-Dieu, Paris, France
| | - Philippe Berthelot
- GIMAP EA 3064, University of Lyon, Saint-Etienne, France
- Infection control unit, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Elisabeth Botelho-Nevers
- Inserm, CIC 1408, I-REIVAC, University Hospital of Saint-Etienne, Saint-Etienne, France
- GIMAP EA 3064, University of Lyon, Saint-Etienne, France
| |
Collapse
|
23
|
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
|
24
|
Juergens C, Trammel J, Shoji Y, Patterson S, Watson W, Webber C, Gruber WC, Scott DA, Schmoele-Thoma B. Late onset of injection site reactions after vaccination with the 13-valent pneumococcal conjugate vaccine in adult study populations. Hum Vaccin Immunother 2018. [PMID: 29543583 PMCID: PMC6149808 DOI: 10.1080/21645515.2018.1452576] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Injection site reactions (ISRs; redness, swelling and pain) commonly occur within 1–2 days after vaccination. After administration of toxoid vaccines including diphtheria toxoid, a later onset of ISRs has also been observed. As the serotype capsular polysaccharides in the 13-valent pneumococcal conjugate vaccine (PCV13) are conjugated to cross-reactive material 197 (CRM197), a nontoxic variant of diphtheria toxin, the onset of ISRs over 14 days was explored in 8 adult studies with 19 cohorts. Subjects received PCV13 with aluminum phosphate (AlPO4, n = 5667) or without AlPO4 (n = 304); 1097 subjects received 23-valent pneumococcal polysaccharide vaccine (PPSV23). Late ISRs with onset between days 6–14 were observed in 8/8 cohorts aged ≥65 years after PCV13 with AlPO4 (incidence across cohorts for redness, 2.3%-19.6%; swelling, 0.9%-10.8%; pain, 1.6%-10.0%) and in 1/1 cohort after PCV13 without AlPO4 (redness 10.5%; swelling 7.5%; pain 12.3%); and in 2/4 cohorts aged 50 to 64 years after PCV13 (redness 3.1%-4.8%; swelling 1.0%-3.2%; pain 3.7%-5%). Late ISRs were not generally observed in 1/1 cohort aged 18 to 49 years after PCV13; in 2/2 cohorts aged ≥53 years after PCV13 revaccination; and in 3/3 cohorts aged ≥60 years who received PPSV23, which does not contain CRM197. Post hoc analysis demonstrated numerically higher pneumococcal immune responses in subgroups with late ISRs versus those without. In conclusion, causality of late ISRs is likely multifactorial, with age and the PCV13 carrier protein CRM197 potentially associated. AlPO4, a vaccine adjuvant, did not appear causally related. Observations do not affect the favorable risk-benefit profile of PCV13.
Collapse
Affiliation(s)
- Christine Juergens
- a Pfizer Vaccine Clinical Research and Development , Pfizer Pharma GmbH , Berlin , Germany
| | - James Trammel
- b Biostatistics, Syneos Health , Stonewall , LA , USA
| | - Yasuko Shoji
- c Pfizer Vaccine Clinical Research and Development , Tokyo , Japan
| | - Scott Patterson
- d Pfizer Vaccines Clinical Research & Development , Collegeville , PA , USA
| | - Wendy Watson
- d Pfizer Vaccines Clinical Research & Development , Collegeville , PA , USA
| | - Chris Webber
- e Pfizer Vaccines Clinical Research & Development , Pearl River , NY , USA
| | - William C Gruber
- e Pfizer Vaccines Clinical Research & Development , Pearl River , NY , USA
| | - Daniel A Scott
- d Pfizer Vaccines Clinical Research & Development , Collegeville , PA , USA
| | - Beate Schmoele-Thoma
- a Pfizer Vaccine Clinical Research and Development , Pfizer Pharma GmbH , Berlin , Germany
| |
Collapse
|
25
|
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
|
26
|
Xu X, Zhu H, Lv H. Safety of Staphylococcus aureus four-antigen and three-antigen vaccines in healthy adults: A meta-analysis of randomized controlled trials. Hum Vaccin Immunother 2017; 14:314-321. [PMID: 29064736 DOI: 10.1080/21645515.2017.1395540] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
PURPOSE Two new Staphylococcus aureus vaccines, S. aureus four-antigen (SA4Ag) and three-antigen (SA3Ag) vaccines, have good immunogenicity and tolerance. However, the safety of these vaccines is worth exploring. Here, we performed a meta-analysis to investigate the safety of SA3Ag and SA4Ag by evaluating systemic and local adverse events. METHODS The Medline, EMBASE, and Cochrane databases were searched for randomized clinical trials confirming the safety of SA4Ag and SA3Ag. Two investigators independently selected suitable trials, assessed trial quality, and extracted data. RESULTS Three studies comprising a total of 1,148 participants were included in this review. The two S. aureus vaccines did not increase systemic adverse events (relative ratio 1.1 [95% confidence interval 0.98, 1.24]), but increased the incidence of local adverse events (2.89 [2.15, 3.90]). However, the incidence of severe local adverse events (4.06 [0.78, 21.24]) did not rise significantly. CONCLUSIONS SA4Ag and SA3Ag have acceptable safety in adults.
Collapse
Affiliation(s)
- Xiaoqun Xu
- a Department of Clinical Laboratory, Centre of Laboratory Medicine , Zhejiang Provincial People's Hospital, The Affiliated People's Hospital of Hangzhou Medical College , Hangzhou , Zhejiang , China.,b Zhejiang Chinese Medical University , Hangzhou , Zhejiang , China
| | - Houyong Zhu
- c Department of Cardiology , Hangzhou Hospital of Traditional Chinese Medicine; Hangzhou Dingqiao Hospital , Hangzhou , Zhejiang , China
| | - Huoyang Lv
- a Department of Clinical Laboratory, Centre of Laboratory Medicine , Zhejiang Provincial People's Hospital, The Affiliated People's Hospital of Hangzhou Medical College , Hangzhou , Zhejiang , China
| |
Collapse
|
27
|
Gagneux-Brunon A, Lucht F, Launay O, Berthelot P, Botelho-Nevers E. Les vaccins dans la prévention des infections associées aux soins. JOURNAL DES ANTI-INFECTIEUX 2017. [PMCID: PMC7148680 DOI: 10.1016/j.antinf.2017.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Les infections associées aux soins (IAS) constituent un véritable problème de santé publique. Escherichia coli, Staphylococcus aureus, Clostridium difficile sont les plus souvent à l’origine des IAS. L’antibiorésistance fréquente complique encore la prise en charge et des impasses thérapeutiques existent à présent. Les mesures d’hygiène hospitalière bien qu’essentielles sont insuffisantes pour diminuer drastiquement les IAS. Ainsi, des stratégies alternatives à l’antibiothérapie s’avèrent nécessaires pour prévenir et traiter les IAS. Parmi celles-ci, la vaccination et l’immunisation passive sont probablement les plus prometteuses. Nous avons fait une mise au point sur les vaccins disponibles et en développement clinique pour lutter contre les IAS, chez les patients à risque d’IAS et les soignants. L’intérêt de la vaccination grippale et rotavirus chez les patients pour prévenir ces IAS virales a été examiné. Le développement d’un vaccin anti-S. aureus, déjà émaillé de 2 échecs est complexe. Toutefois, ces échecs ont permis d’améliorer les connaissances sur l’immunité anti-S. aureus. La mise à disposition d’un vaccin préventif anti-C. difficile semble plus proche. Pour les autres bactéries gram négatif responsables d’IAS, le développement est moins avancé. La vaccination des patients à risques d’IAS pose également des problèmes de réponse vaccinale qu’il faudra résoudre pour utiliser cette stratégie. Ainsi, la vaccination des soignants, de par l’effet de groupe permet également de prévenir les IAS. Nous faisons ici le point sur l’intérêt de la vaccination des soignants contre la rougeole, la coqueluche, la grippe, la varicelle, l’hépatite B pour réduire les IAS avec des vaccins déjà disponibles.
Collapse
|
28
|
Scully IL, McNeil LK, Pathirana S, Singer CL, Liu Y, Mullen S, Girgenti D, Gurtman A, Pride MW, Jansen KU, Huang PL, Anderson AS. Neutrophil killing of Staphylococcus aureus in diabetes, obesity and metabolic syndrome: a prospective cellular surveillance study. Diabetol Metab Syndr 2017; 9:76. [PMID: 29026443 PMCID: PMC5627489 DOI: 10.1186/s13098-017-0276-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 09/25/2017] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Obesity, metabolic syndrome (MetS), and diabetes are frequent in surgical populations and can enhance susceptibility to postoperative surgical site infections. Reduced neutrophil function has been linked with diabetes and risk of Staphylococcus aureus infection. Therefore, neutrophil function in diabetic and obese subjects (± MetS) was assessed in this prospective serological and cellular surveillance study to determine whether vaccines administered to protect against infections after surgery could be effective in these populations. METHODS Neutrophil function (chemotaxis, phagocytosis, and opsonophagocytic killing of S. aureus) was assessed in subjects classified according to diabetes status, body mass index, and presence/absence of MetS. Neutrophils were characterized within functional subsets by flow cytometry. A serologic assay was used to measure baseline antibody presence to each antigen in SA4Ag: capsular polysaccharide (CP) type 5, CP8, recombinant mutant Clumping factor A (rmClfA), and recombinant Manganese transport protein C (rMntC). RESULTS Neutrophil function was similar for comorbid and healthy cohorts, with no significant between-group differences in cell counts, migration, phagocytosis ability, neutrophil subset proportions, and S. aureus killing ability when neutrophils were isolated 3-6 months apart (Visit 1 [n = 90] and Visit 2 [n = 70]) and assessed. Median pre-existing antibody titers to CP5, CP8, and rmClfA were comparable for all cohorts (insufficient subjects with rMntC titers for determination). CONCLUSIONS MetS, diabetes, and obesity do not impact in vitro neutrophil function with regard to S. aureus killing, suggesting that if an effective S. aureus vaccine is developed it may be effective in individuals with these comorbidities.
Collapse
Affiliation(s)
- Ingrid Lea Scully
- Pfizer Vaccine Research and Development, 401 North Middletown Rd, Pearl River, NY 10965 USA
| | - Lisa Kristin McNeil
- Pfizer Vaccine Research and Development, 401 North Middletown Rd, Pearl River, NY 10965 USA
| | - Sudam Pathirana
- Pfizer Vaccine Research and Development, 401 North Middletown Rd, Pearl River, NY 10965 USA
| | - Christine Lee Singer
- Pfizer Vaccine Research and Development, 401 North Middletown Rd, Pearl River, NY 10965 USA
| | - Yongdong Liu
- Pfizer Vaccine Research and Development, 401 North Middletown Rd, Pearl River, NY 10965 USA
| | - Stanley Mullen
- Pfizer Vaccine Research and Development, 401 North Middletown Rd, Pearl River, NY 10965 USA
| | - Douglas Girgenti
- Pfizer Vaccine Research and Development, 401 North Middletown Rd, Pearl River, NY 10965 USA
| | - Alejandra Gurtman
- Pfizer Vaccine Research and Development, 401 North Middletown Rd, Pearl River, NY 10965 USA
| | - Michael W. Pride
- Pfizer Vaccine Research and Development, 401 North Middletown Rd, Pearl River, NY 10965 USA
| | - Kathrin Ute Jansen
- Pfizer Vaccine Research and Development, 401 North Middletown Rd, Pearl River, NY 10965 USA
| | - Paul L. Huang
- Cardiovascular Research Center and Cardiology Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - Annaliesa S. Anderson
- Pfizer Vaccine Research and Development, 401 North Middletown Rd, Pearl River, NY 10965 USA
| |
Collapse
|
29
|
Landrum ML, Lalani T, Niknian M, Maguire JD, Hospenthal DR, Fattom A, Taylor K, Fraser J, Wilkins K, Ellis MW, Kessler PD, Fahim REF, Tribble DR. Safety and immunogenicity of a recombinant Staphylococcus aureus α-toxoid and a recombinant Panton-Valentine leukocidin subunit, in healthy adults. Hum Vaccin Immunother 2017; 13:791-801. [PMID: 28010246 PMCID: PMC5404372 DOI: 10.1080/21645515.2016.1248326] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 09/29/2016] [Accepted: 10/11/2016] [Indexed: 01/01/2023] Open
Abstract
We conducted a randomized, double-blind, placebo-controlled dose-escalation study in healthy adults to evaluate the safety and immunogenicity of recombinant Staphylococcus aureus candidate vaccine antigens, recombinant α-toxoid (rAT) and a sub-unit of Panton-Valentine leukocidin (rLukS-PV). 176 subjects were enrolled and randomized within 1 of 11 treatment cohorts: monovalent rAT or rLukS-PV dosages of 10, 25, 50, and 100 μg; bivalent rAT:rLukS dosages of 10:10, 25:25, and 50:50 μg; and alum or saline placebo. All subjects were assessed at Days 0, 7, 14, 28, and 84. Subjects in the 50:50 μg bivalent cohort received a second injection on Day 84 and were assessed on Days 98 and 112. Incidence and severity of reactogenicity and adverse events (AEs) were compared. Geometric mean serum concentrations (GMC) and neutralizing activity of anti-rAT and anti-rLukS-PV IgG were assessed. Reactogenicity incidence was significantly higher in vaccine than placebo recipients (77% versus 55%, respectively; p = 0.006). However, 77% of reactogenicity events were mild and 19% were moderate in severity. The AE incidence and severity were similar between the cohorts. All monovalent and bivalent rAT dosages resulted in a significant increase in the anti-rAT IgG and anti- rLukS-PV GMCs between day 0 and 28 compared with placebo, and persisted through Day 84. Exploratory subgroup analyses suggested a higher GMC and neutralizing antibody titers for the 50 μg monovalent or bivalent rAT and rLukS-PV dose as compared to the other doses. No booster effect was observed after administration of the second dose. We conclude that the rAT and rLukS-PV vaccine formulations were well-tolerated and had a favorable immunogenicity profile, producing antibody with neutralizing activity through day 84. There was no benefit observed with a booster dose of the vaccine.
Collapse
Affiliation(s)
- Michael L. Landrum
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Rockville, MD, USA
- Division of Infectious Diseases, San Antonio Military Medical Center, Fort Sam Houston, TX, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Tahaniyat Lalani
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Rockville, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
- Division of Infectious Diseases, Naval Medical Center Portsmouth, Portsmouth, VA, USA
| | | | - Jason D. Maguire
- Division of Infectious Diseases, Naval Medical Center Portsmouth, Portsmouth, VA, USA
| | - Duane R. Hospenthal
- Division of Infectious Diseases, San Antonio Military Medical Center, Fort Sam Houston, TX, USA
| | - Ali Fattom
- Nabi Biopharmaceuticals, Rockville, MD, USA
| | | | - Jamie Fraser
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Rockville, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Kenneth Wilkins
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Rockville, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Michael W. Ellis
- Infectious Diseases Division, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | | | | | - David R. Tribble
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Rockville, MD, USA
| |
Collapse
|
30
|
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
|
31
|
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
|
32
|
Khatun F, Stephenson RJ, Toth I. An Overview of Structural Features of Antibacterial Glycoconjugate Vaccines That Influence Their Immunogenicity. Chemistry 2017; 23:4233-4254. [PMID: 28097690 DOI: 10.1002/chem.201603599] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Indexed: 12/13/2022]
Abstract
Bacterial cell-surface-derived or mimicked carbohydrate moieties that act as protective antigens are used in the development of antibacterial glycoconjugate vaccines. The carbohydrate antigen must have a minimum length or size to maintain the conformational structure of the antigenic epitope(s). The presence or absence of O-acetate, phosphate, glycerol phosphate and pyruvate ketal plays a vital role in defining the immunogenicity of the carbohydrate antigen. The nature of the carrier protein, spacer and conjugation pattern used to develop the glycoconjugate vaccine also defines its overall spatial orientation which in turn affects its avidity and selectivity of interaction with the desired target(s). In addition, the ratio of carbohydrate to protein in glycoconjugate vaccines also makes an important contribution in determining the optimum immunological response. This Review article presents the importance of these variables in the development of antibacterial glycoconjugate vaccines and their effects on immune efficacy.
Collapse
Affiliation(s)
- Farjana Khatun
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD, Australia
| | - Rachel J Stephenson
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD, Australia
| | - Istvan Toth
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD, Australia.,School of Pharmacy, Woolloongabba, The University of Queensland, QLD, Australia.,Institute for Molecular Bioscience, St. Lucia, The University of Queensland, QLD, Australia
| |
Collapse
|
33
|
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
|
34
|
Safety, tolerability, and immunogenicity of a single dose 4-antigen or 3-antigen Staphylococcus aureus vaccine in healthy older adults: Results of a randomised trial. Vaccine 2016; 35:385-394. [PMID: 27866765 DOI: 10.1016/j.vaccine.2016.11.032] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 11/01/2016] [Accepted: 11/07/2016] [Indexed: 12/23/2022]
Abstract
BACKGROUND The decline in immune function with age is a challenge to vaccine development. Following an initial study in adults aged 18-64years, this study evaluated the safety and immunogenicity of Staphylococcus aureus (S. aureus) 4-antigen (SA4Ag) and 3-antigen (SA3Ag) vaccine in older adults. SA3Ag included capsular polysaccharide serotypes 5 and 8 (CP5 and CP8) conjugated to the nontoxic mutant form of diphtheria toxin (CRM197) and a recombinant version of clumping factor A (ClfA). SA4Ag included these antigens, with the addition of a recombinant manganese transporter C (rP305A or MntC). Both vaccines were unadjuvanted. METHODS In this double-blind, sponsor-unblinded, placebo-controlled, phase 1/2 study, 284 healthy adults (aged 65-85years) were randomised to receive a single dose of one of three formulations of SA4Ag with escalating dose levels of rP305A, SA3Ag, or placebo. Functional immune responses were measured using opsonophagocytic activity (OPA) killing and fibrinogen-binding inhibition (FBI) assays; immunogenicity was also assessed using a competitive Luminex® immunoassay (cLIA). T-cell responses were measured in a small subgroup of subjects using intracellular cytokine staining (ICS) assays. RESULTS The results demonstrated rapid and robust functional immune responses to all antigens in healthy older adults. A high proportion of active vaccine recipients met the pre-defined antibody thresholds for each antigen at Day 29. SA4Ag elicited a dose-level response to rP305A with up to a 13-fold rise in cLIA titres at Day 29. Opsonophagocytic activity (OPA) assays showed >50- and >20-fold rises in functional titres using S. aureus strains expressing CP5 and CP8, respectively, at Day 29. T-cell cytokine responses were not substantially above background levels. There were no safety concerns in this study population and no increases in adverse events with higher rP305A dose levels. CONCLUSIONS Single-dose vaccination of SA4Ag and SA3Ag in healthy adults aged 65-85years safely induced rapid and robust functional immune responses, supporting further development of SA4Ag for the prevention of S. aureus disease in adults up to age 85years. TRIAL REGISTRATION NUMBER NCT01643941.
Collapse
|
35
|
Abstract
INTRODUCTION The success of the vaccines available on the market has significantly increased interest in vaccine development. Areas covered: The main aim of this paper is to discuss the most important vaccines of pediatric interest that are currently being developed. New pneumococcal vaccines and vaccines against group B Streptococcus, Staphylococcus aureus and respiratory syncytial virus are analyzed in detail. Expert commentary: Advances in understanding human immunology, including human monoclonal antibody identification, sequencing technology, and the ability to solve atomic level structures of vaccine targets have provided tools to guide the rational design of future vaccines. It is likely that some of these vaccines will reach the market in the future and will thus partially contribute to the prevention of very severe diseases that significantly affect the morbidity and mortality of children. However, further studies in animals and several clinical trials in children must be performed before new vaccines become licensed.
Collapse
Affiliation(s)
- Susanna Esposito
- a Pediatric Highly Intensive Care Unit, Department of Pathophysiology and Transplantation , Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico , Milan , Italy
| | - Nicola Principi
- a Pediatric Highly Intensive Care Unit, Department of Pathophysiology and Transplantation , Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico , Milan , Italy
| |
Collapse
|
36
|
Safety and immunogenicity of a booster dose of a 3-antigen Staphylococcus aureus vaccine (SA3Ag) in healthy adults: A randomized phase 1 study. J Infect 2016; 73:437-454. [PMID: 27519620 DOI: 10.1016/j.jinf.2016.08.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 07/29/2016] [Accepted: 08/05/2016] [Indexed: 12/28/2022]
Abstract
OBJECTIVE A 2-stage, phase 1, randomized, placebo-controlled study in healthy adults to assess immunogenicity and safety of a booster dose at three dose levels of a 3-antigen Staphylococcus aureus vaccine (SA3Ag) containing recombinant clumping factor A (ClfA) and capsular polysaccharides 5 and 8 (CP5 and CP8) conjugated to a diphtheria toxoid. METHODS Six months after initial single vaccination, in Stage 2, SA3Ag recipients were randomized (1:1) to booster vaccination or placebo, while Stage 1 placebo recipients received placebo again. Pre- and post-vaccination blood samples were analyzed. RESULTS In Stage 2 (n = 345), pre-booster CP5 and CP8 titers remained high with no increase post-booster. ClfA titers remained high after initial vaccination and increased post-booster, approaching the peak response to the initial dose. Post-booster local reactions were more frequent and of greater severity than reported after the initial vaccination, particularly for the high-dose level recipients. Post hoc analysis showed no dose-response pattern and no obvious association between diphtheria toxoid titers and local reactions after initial or booster vaccination. CONCLUSION Immune responses after the initial vaccination persisted for the 12 months studied, with little additional response after the booster dose at 6 months. Post-booster injection site reactions were more frequent and more severe but self-limiting. CLINICALTRIALS. GOV IDENTIFIER NCT01018641.
Collapse
|
37
|
Skurnik D, Cywes-Bentley C, Pier GB. The exceptionally broad-based potential of active and passive vaccination targeting the conserved microbial surface polysaccharide PNAG. Expert Rev Vaccines 2016; 15:1041-53. [PMID: 26918288 PMCID: PMC4985264 DOI: 10.1586/14760584.2016.1159135] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 02/24/2016] [Indexed: 11/08/2022]
Abstract
A challenging component of vaccine development is the large serologic diversity of protective antigens. Remarkably, there is a conserved surface/capsular polysaccharide, one of the most effective vaccine targets, expressed by a large number of bacterial, fungal and eukaryotic pathogens: poly-N-acetyl glucosamine (PNAG). Natural antibodies to PNAG are poorly effective at mediating in vitro microbial killing or in vivo protection. Removing most of the acetate substituents to produce a deacetylated glycoform, or using synthetic oligosaccharides of poly-β-1-6-linked glucosamine conjugated to carrier proteins, results in vaccines that elicit high levels of broad-based immunity. A fully human monoclonal antibody is highly active in laboratory and preclinical studies and has been successfully tested in a phase-I setting. Both the synthetic oligosaccharide conjugate vaccine and MAb will be further tested in humans starting in 2016; but, even if effective against only a fraction of the PNAG-producing pathogens, a major advance in vaccine-preventable diseases will occur.
Collapse
Affiliation(s)
- David Skurnik
- Division of Infectious Diseases, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 181 Longwood Ave., Boston, MA 02115, Phone: 617-525-2269; FAX: 617-525-2510
| | - Colette Cywes-Bentley
- Division of Infectious Diseases, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 181 Longwood Ave., Boston, MA 02115, Phone: 617-525-2269; FAX: 617-525-2510
| | - Gerald B. Pier
- Division of Infectious Diseases, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 181 Longwood Ave., Boston, MA 02115, Phone: 617-525-2269; FAX: 617-525-2510
| |
Collapse
|
38
|
Yasomanee JP, Visansirikul S, Pornsuriyasak P, Thompson M, Kolodziej SA, Demchenko AV. Synthesis of the Repeating Unit of Capsular Polysaccharide Staphylococcus aureus Type 5 To Study Chemical Activation and Conjugation of Native CP5. J Org Chem 2016; 81:5981-7. [DOI: 10.1021/acs.joc.6b00910] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- Jagodige P. Yasomanee
- Department
of Chemistry and Biochemistry, University of Missouri—St. Louis, One University Boulevard, St. Louis, Missouri 63121, United States
| | - Satsawat Visansirikul
- Department
of Chemistry and Biochemistry, University of Missouri—St. Louis, One University Boulevard, St. Louis, Missouri 63121, United States
| | - Papapida Pornsuriyasak
- Department
of Chemistry and Biochemistry, University of Missouri—St. Louis, One University Boulevard, St. Louis, Missouri 63121, United States
| | - Melissa Thompson
- Analytical R&D, Biotherapeutics Pharmaceutical Sciences, Pfizer, Inc., 700 Chesterfield Parkway W, Chesterfield, Missouri 63017, United States
| | - Stephen A. Kolodziej
- Bioprocess R&D, Biotherapeutics Pharmaceutical Sciences, Pfizer, Inc., 700 Chesterfield Parkway W, Chesterfield, Missouri 63017, United States
| | - Alexei V. Demchenko
- Department
of Chemistry and Biochemistry, University of Missouri—St. Louis, One University Boulevard, St. Louis, Missouri 63121, United States
| |
Collapse
|
39
|
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
|
40
|
Kuipers A, Stapels DAC, Weerwind LT, Ko YP, Ruyken M, Lee JC, van Kessel KPM, Rooijakkers SHM. The Staphylococcus aureus polysaccharide capsule and Efb-dependent fibrinogen shield act in concert to protect against phagocytosis. MICROBIOLOGY-SGM 2016; 162:1185-1194. [PMID: 27112346 DOI: 10.1099/mic.0.000293] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Staphylococcus aureus has developed many mechanisms to escape from human immune responses. To resist phagocytic clearance, S. aureus expresses a polysaccharide capsule, which effectively masks the bacterial surface and surface-associated proteins, such as opsonins, from recognition by phagocytic cells. Additionally, secretion of the extracellular fibrinogen binding protein (Efb) potently blocks phagocytic uptake of the pathogen. Efb creates a fibrinogen shield surrounding the bacteria by simultaneously binding complement C3b and fibrinogen at the bacterial surface. By means of neutrophil phagocytosis assays with fluorescently labelled encapsulated serotype 5 (CP5) and serotype 8 (CP8) strains we compare the immune-modulating function of these shielding mechanisms. The data indicate that, in highly encapsulated S. aureus strains, the polysaccharide capsule is able to prevent phagocytic uptake at plasma concentrations <10 %, but loses its protective ability at higher concentrations of plasma. Interestingly, Efb shows a strong inhibitory effect on both capsule-negative and encapsulated strains at all tested plasma concentrations. Furthermore, the results suggest that both shielding mechanisms can exist simultaneously and collaborate to provide optimal protection against phagocytosis at a broad range of plasma concentrations. As opsonizing antibodies will be shielded from recognition by either mechanism, incorporating both capsular polysaccharides and Efb in future vaccines could be of great importance.
Collapse
Affiliation(s)
- Annemarie Kuipers
- Medical Microbiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Daphne A C Stapels
- Medical Microbiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Lleroy T Weerwind
- Medical Microbiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Ya-Ping Ko
- Center for Infectious and Inflammatory Disease, Institute of Bioscience and Technology, Texas A&M University Health Science Center, Houston, TX 77030, USA
| | - Maartje Ruyken
- Medical Microbiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Jean C Lee
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Kok P M van Kessel
- Medical Microbiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Suzan H M Rooijakkers
- Medical Microbiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| |
Collapse
|
41
|
Bröker BM, Mrochen D, Péton V. The T Cell Response to Staphylococcus aureus. Pathogens 2016; 5:pathogens5010031. [PMID: 26999219 PMCID: PMC4810152 DOI: 10.3390/pathogens5010031] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 03/01/2016] [Accepted: 03/08/2016] [Indexed: 01/04/2023] Open
Abstract
Staphylococcus aureus (S. aureus) is a dangerous pathogen and a leading cause of both nosocomial and community acquired bacterial infection worldwide. However, on the other hand, we are all exposed to this bacterium, often within the first hours of life, and usually manage to establish equilibrium and coexist with it. What does the adaptive immune system contribute toward lifelong control of S. aureus? Will it become possible to raise or enhance protective immune memory by vaccination? While in the past the S. aureus-specific antibody response has dominated this discussion, the research community is now coming to appreciate the role that the cellular arm of adaptive immunity, the T cells, plays. There are numerous T cell subsets, each with differing functions, which together have the ability to orchestrate the immune response to S. aureus and hence to tip the balance between protection and pathology. This review summarizes the state of the art in this dynamic field of research.
Collapse
Affiliation(s)
- Barbara M Bröker
- Department of Immunology, University Medicine Greifswald, Sauerbruchstraße DZ7, 17475 Greifswald, Germany.
| | - Daniel Mrochen
- Department of Immunology, University Medicine Greifswald, Sauerbruchstraße DZ7, 17475 Greifswald, Germany.
| | - Vincent Péton
- Department of Immunology, University Medicine Greifswald, Sauerbruchstraße DZ7, 17475 Greifswald, Germany.
| |
Collapse
|
42
|
Staphylococcus aureus Clumping Factor A Remains a Viable Vaccine Target for Prevention of S. aureus Infection. mBio 2016; 7:e00225. [PMID: 26956591 PMCID: PMC4810491 DOI: 10.1128/mbio.00225-16] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
In a recent article, X. Li et al. [mBio 7(1):e02232-15, 2016, http://dx.doi.org/10.1128/mBio.02232-15] investigate the utility of a vaccine composed of the Staphylococcus aureus protein clumping factor A (ClfA) in protecting mice from S. aureus infection. ClfA, one of the first proteins to be identified as a potential vaccine antigen for S. aureus prophylaxis, is currently a component of several investigational vaccines. The authors conclude that ClfA may not be effective for S. aureus prophylaxis. In contrast, previously published papers reporting positive data suggested that ClfA was potentially an important vaccine target to prevent invasive S. aureus disease. This commentary addresses the observed differences between the findings of Li et al. and those from other publications, highlighting the importance for preclinical vaccine antigen assessments to reflect the biological role of said antigen in virulence and, consequently, the importance of choosing appropriate preclinical disease models to test such antigens.
Collapse
|
43
|
Whitehouse J, Flaxman A, Rollier C, O'Shea MK, Fallowfield J, Lindsay M, Gunner F, Knox K, Wyllie DH, Yamaguchi Y. Population variation in anti-S. aureus IgG isotypes influences surface protein A mediated immune subversion. Vaccine 2016; 34:1792-9. [PMID: 26921780 DOI: 10.1016/j.vaccine.2016.02.034] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 02/03/2016] [Accepted: 02/15/2016] [Indexed: 01/10/2023]
Abstract
BACKGROUND Staphylococcus aureus is a pathogen which causes life-threatening infection, the incidence of which rises during adult life. This, together with the emergence of drug-resistant strains and the expansion of more susceptible elderly populations, represents the rationale for the ongoing development of S. aureus vaccines targeting adult populations. Humoral responses to S. aureus naturally develop early in life, influence susceptibility to infection, and potentially influence the effect of vaccination. Despite this, the nature of pre-existing anti-S. aureus antibodies in healthy adult populations is not fully characterised. METHODS Immunoglobulin levels against S. aureus surface antigens were measured by a filter membrane enzyme-linked immunosorbent assay using fixed ΔSpA S. aureus as an antigen in serum samples obtained from three clinical cohorts comprising 133 healthy adult volunteers from 19 to 65 years of age. Functional capacity of antibody was also assessed, using antibody-mediated attachment of FITC-stained S. aureus to differentiated HL-60 cells. RESULTS Wide variation in the concentrations of immunoglobulins recognising S. aureus surface antigens was observed among individuals in all three cohorts. There was a decline of anti-S. aureus IgG1 with age, and a similar trend was observed in IgM, but not in IgA or other IgG sub-classes. Antibody mediated bacterial attachment to cells was associated with IgG1 and IgG3 concentrations in serum. The presence of SpA on the bacterial cell surface reduced antibody-mediated binding of bacteria to phagocytes in serum with low, but not high, levels of naturally occurring anti-S. aureus IgG3 antibodies. CONCLUSIONS Naturally acquired immunoglobulin responses to S. aureus are heterogeneous in populations and their concentrations alter during adulthood. Elevated IgG1 or IgG3 titres against S. aureus enhance S. aureus recognition by phagocytosis and may be correlates of natural protection and/or vaccine efficacy in adult populations.
Collapse
Affiliation(s)
- Julia Whitehouse
- Jenner Institute, Centre for Cellular & Molecular Physiology, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, United Kingdom
| | - Amy Flaxman
- Jenner Institute, Centre for Cellular & Molecular Physiology, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, United Kingdom
| | - Christine Rollier
- Oxford Vaccine Group, Department of Paediatrics, Centre for Clinical Vaccinology and Tropical Medicine, University of Oxford, Churchill Drive, Oxford OX3 7LE, United Kingdom
| | - Matthew K O'Shea
- Jenner Institute, Centre for Cellular & Molecular Physiology, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, United Kingdom; Department of Academic Medicine, Royal Centre for Defence Medicine, Birmingham B15 2SQ, United Kingdom
| | - Joanne Fallowfield
- Environmental Medicine and Science Division, Institute of Naval Medicine, Hampshire PO12 2DL, United Kingdom
| | - Michael Lindsay
- Environmental Medicine and Science Division, Institute of Naval Medicine, Hampshire PO12 2DL, United Kingdom
| | - Frances Gunner
- Environmental Medicine and Science Division, Institute of Naval Medicine, Hampshire PO12 2DL, United Kingdom
| | - Kyle Knox
- Nuffield Department of Primary Care Health Sciences, New Radcliffe House, 2nd floor, Walton Street, Jericho OX2 6NW, United Kingdom
| | - David H Wyllie
- Jenner Institute, Centre for Cellular & Molecular Physiology, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, United Kingdom; Nuffield Department of Medicine, Department of Microbiology, John Radcliffe Hospital, Oxford OX3 9DU, United Kingdom.
| | - Yuko Yamaguchi
- Jenner Institute, Centre for Cellular & Molecular Physiology, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, United Kingdom
| |
Collapse
|
44
|
Lacey KA, Geoghegan JA, McLoughlin RM. The Role of Staphylococcus aureus Virulence Factors in Skin Infection and Their Potential as Vaccine Antigens. Pathogens 2016; 5:pathogens5010022. [PMID: 26901227 PMCID: PMC4810143 DOI: 10.3390/pathogens5010022] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 01/27/2016] [Accepted: 02/03/2016] [Indexed: 01/13/2023] Open
Abstract
Staphylococcus aureus (S. aureus) causes the vast majority of skin and soft tissue infections (SSTIs) in humans. S. aureus has become increasingly resistant to antibiotics and there is an urgent need for new strategies to tackle S. aureus infections. Vaccines offer a potential solution to this epidemic of antimicrobial resistance. However, the development of next generation efficacious anti-S. aureus vaccines necessitates a greater understanding of the protective immune response against S. aureus infection. In particular, it will be important to ascertain if distinct immune mechanisms are required to confer protection at distinct anatomical sites. Recent discoveries have highlighted that interleukin-17-producing T cells play a particularly important role in the immune response to S. aureus skin infection and suggest that vaccine strategies to specifically target these types of T cells may be beneficial in the treatment of S. aureus SSTIs. S. aureus expresses a large number of cell wall-anchored (CWA) proteins, which are covalently attached to the cell wall peptidoglycan. The virulence potential of many CWA proteins has been demonstrated in infection models; however, there is a paucity of information regarding their roles during SSTIs. In this review, we highlight potential candidate antigens for vaccines targeted at protection against SSTIs.
Collapse
Affiliation(s)
- Keenan A Lacey
- Host Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland.
- Microbiology Department, Moyne Institute of Preventive Medicine, Trinity College Dublin, Dublin 2, Ireland.
| | - Joan A Geoghegan
- Microbiology Department, Moyne Institute of Preventive Medicine, Trinity College Dublin, Dublin 2, Ireland.
| | - Rachel M McLoughlin
- Host Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland.
| |
Collapse
|
45
|
Abstract
UNLABELLED Treatment of Staphylococcus aureus infections has become increasingly difficult because of the emergence of multidrug-resistant isolates. Development of a vaccine to prevent staphylococcal infections remains a priority. To determine whether clumping factor A (ClfA) is a good target protein for inclusion in a multivalent vaccine, we evaluated its efficacy in a variety of relevant staphylococcal infection models, challenging with different S. aureus strains. ClfA adsorbed to Alhydrogel and mixed with Sigma Adjuvant System was more immunogenic and stimulated a more robust Th17 response than ClfA administered with alum alone. ClfA immunization induced the production of functional antibodies in rabbits and mice that blocked S. aureus binding to fibrinogen and were opsonic for S. aureus strains that produced little or no capsular polysaccharide. Mice immunized with ClfA showed a modest reduction in the bacterial burden recovered from subcutaneous abscesses provoked by S. aureus USA300 strain LAC. In addition, the ClfA vaccine reduced lethality in a sepsis model following challenge with strain Newman, but not ST80. Vaccination with ClfA did not protect against surgical wound infection, renal abscess formation, or bacteremia. Passive immunization with antibodies to ClfA did not protect against staphylococcal bacteremia in mice or catheter-induced endocarditis in rats. Some enhancement of bacteremia was observed by ClfA immunization or passive administration of ClfA antibodies when mice were challenged by the intraperitoneal route. Although rodent models of staphylococcal infection have their limitations, our data do not support the inclusion of ClfA in an S. aureus multivalent vaccine. IMPORTANCE Antibiotics are often ineffective in eradicating Staphylococcus aureus infections, and thus, a preventative vaccine is sorely needed. Two single-component vaccines and two immunoglobulin preparations failed to meet their designated endpoints in phase III clinical trials. Importantly, recipients of an S. aureus surface protein (iron surface determinant B) vaccine who developed a staphylococcal infection experienced a higher rate of multiorgan failure and mortality than placebo controls, raising safety concerns. Multicomponent S. aureus vaccines have now been generated, and several include surface protein clumping factor A (ClfA). We immunized mice with ClfA and generated a robust T cell response and serum antibodies that were functional in vitro. Nonetheless, ClfA was not protective in a number of relevant animal models of S. aureus infection, and high levels of ClfA antibodies enhanced bacteremia when mice were challenged with community-acquired methicillin-resistant S. aureus strains. Evidence supporting ClfA as a vaccine component is lacking.
Collapse
|
46
|
Bhattacharya M, Wozniak DJ, Stoodley P, Hall-Stoodley L. Prevention and treatment of Staphylococcus aureus biofilms. Expert Rev Anti Infect Ther 2015; 13:1499-516. [PMID: 26646248 DOI: 10.1586/14787210.2015.1100533] [Citation(s) in RCA: 196] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
S. aureus colonizes both artificial and tissue surfaces in humans causing chronic persistent infections that are difficult to cure. It is a notorious pathogen due to its antibiotic recalcitrance and phenotypic adaptability, both of which are facilitated by its ability to develop biofilms. S. aureus biofilms challenge conventional anti-infective approaches, most notably antibiotic therapy. Therefore there is an unmet need to develop and include parallel approaches that target S. aureus biofilm infections. This review discusses two broad anti-infective strategies: (1) preventative approaches (anti-biofilm surface coatings, the inclusion of biofilm-specific vaccine antigens); and (2) approaches aimed at eradicating established S. aureus biofilms, particularly those associated with implant infections. Advances in understanding the distinct nature of S. aureus biofilm development and pathogenesis have led to growing optimism in S. aureus biofilm targeted anti-infective strategies. Further research is needed however, to see the successful administration and validation of these approaches to the diverse types of infections caused by S. aureus biofilms from multiple clinical strains.
Collapse
Affiliation(s)
- Mohini Bhattacharya
- a Department of Microbiology , The Ohio State University , Columbus , OH , USA
| | - Daniel J Wozniak
- a Department of Microbiology , The Ohio State University , Columbus , OH , USA.,b Department of Microbial Infection and Immunity , The Ohio State University College of Medicine , Columbus , OH , USA.,c The Center for Microbial Interface Biology, The Ohio State University , Columbus , OH , USA
| | - Paul Stoodley
- b Department of Microbial Infection and Immunity , The Ohio State University College of Medicine , Columbus , OH , USA.,c The Center for Microbial Interface Biology, The Ohio State University , Columbus , OH , USA.,d Department of Orthopedics , The Ohio State University College of Medicine , Columbus , OH , USA.,e Department of Engineering Sciences, National Centre for Advanced Tribology at Southampton (nCATS) , University of Southampton , Southampton , UK
| | - Luanne Hall-Stoodley
- b Department of Microbial Infection and Immunity , The Ohio State University College of Medicine , Columbus , OH , USA.,c The Center for Microbial Interface Biology, The Ohio State University , Columbus , OH , USA
| |
Collapse
|
47
|
Vuong C, Yeh AJ, Cheung GYC, Otto M. Investigational drugs to treat methicillin-resistant Staphylococcus aureus. Expert Opin Investig Drugs 2015; 25:73-93. [PMID: 26536498 DOI: 10.1517/13543784.2016.1109077] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
INTRODUCTION Staphylococcus aureus remains one of the leading causes of morbidity and mortality worldwide. This is to a large extent due to antibiotic-resistant strains, in particular methicillin-resistant S. aureus (MRSA). While the toll of invasive MRSA infections appears to decrease in U.S. hospitals, the rate of community-associated MRSA infections remains constant and there is a surge of MRSA in many other countries, a situation that calls for continuing if not increased efforts to find novel strategies to combat MRSA infections. AREAS COVERED This review provides an overview of current investigational drugs and therapeutic antibodies against S. aureus in early clinical development (up to phase II clinical development). It includes a short description of the mechanism of action and a presentation of microbiological and clinical data. EXPERT OPINION Increased recent antibiotic development efforts and results from pathogenesis research have led to several new antibiotics and therapies, such as anti-virulence drugs, as well as a more informed selection of targets for vaccination efforts against MRSA. This developing portfolio of novel anti-staphylococcal drugs will hopefully provide us with additional and more efficient ways to combat MRSA infections in the near future and prevent us from running out of treatment options, even if new resistances arise.
Collapse
Affiliation(s)
- Cuong Vuong
- a Principal Scientist/Laboratory Head, Bacteriology , AiCuris GmbH & Co. KG, Friedrich-Ebert-Str. 475/Geb. 302, 42117 Wuppertal , Germany
| | - Anthony J Yeh
- b Post-baccalaureate IRTA, Laboratory of Bacteriology , National Institute of Allergy and Infectious Diseases, National Institutes of Health , Bldg. 33, 1W10, 9000 Rockville Pike, Bethesda , MD 20892 , USA
| | - Gordon Y C Cheung
- c Staff Scientist, National Institute of Allergy and Infectious Diseases , National Institutes of Health, Laboratory of Bacteriology , Bldg. 33, 1W10, 9000 Rockville Pike, Bethesda , MD 20892 , USA
| | - Michael Otto
- d Senior Investigator, National Institute of Allergy and Infectious Diseases , National Institutes of Health, Laboratory of Bacteriology , Bldg. 33, 1W10, 9000 Rockville Pike, Bethesda , MD 20892 , USA
| |
Collapse
|
48
|
Scully IL, Swanson K, Green L, Jansen KU, Anderson AS. Anti-infective vaccination in the 21st century—new horizons for personal and public health. Curr Opin Microbiol 2015; 27:96-102. [DOI: 10.1016/j.mib.2015.07.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 07/10/2015] [Indexed: 12/17/2022]
|
49
|
Weidenmaier C, Lee JC. Structure and Function of Surface Polysaccharides of Staphylococcus aureus. Curr Top Microbiol Immunol 2015; 409:57-93. [PMID: 26728067 DOI: 10.1007/82_2015_5018] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The major surface polysaccharides of Staphylococcus aureus include the capsular polysaccharide (CP), cell wall teichoic acid (WTA), and polysaccharide intercellular adhesin/poly-β(1-6)-N-acetylglucosamine (PIA/PNAG). These glycopolymers are important components of the staphylococcal cell envelope, but none of them is essential to S. aureus viability and growth in vitro. The overall biosynthetic pathways of CP, WTA, and PIA/PNAG have been elucidated, and the functions of most of the biosynthetic enzymes have been demonstrated. Because S. aureus CP and WTA (but not PIA/PNAG) utilize a common cell membrane lipid carrier (undecaprenyl-phosphate) that is shared by the peptidoglycan biosynthesis pathway, there is evidence that these processes are highly integrated and temporally regulated. Regulatory elements that control glycopolymer biosynthesis have been described, but the cross talk that orchestrates the biosynthetic pathways of these three polysaccharides remains largely elusive. CP, WTA, and PIA/PNAG each play distinct roles in S. aureus colonization and the pathogenesis of staphylococcal infection. However, they each promote bacterial evasion of the host immune defences, and WTA is being explored as a target for antimicrobial therapeutics. All the three glycopolymers are viable targets for immunotherapy, and each (conjugated to a carrier protein) is under evaluation for inclusion in a multivalent S. aureus vaccine. Future research findings that increase our understanding of these surface polysaccharides, how the bacterial cell regulates their expression, and their biological functions will likely reveal new approaches to controlling this important bacterial pathogen.
Collapse
Affiliation(s)
- Christopher Weidenmaier
- Interfaculty Institute for Microbiology and Infection Medicine Tübingen, University of Tübingen and German Center for Infection Research, Tübingen, Germany
| | - Jean C Lee
- Division of Infectious Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|