Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 82
Filter
1.
Front Immunol ; 14: 1221108, 2023.
Article in English | MEDLINE | ID: mdl-37828992

ABSTRACT

Staphylococcus aureus pathology is caused by a plethora of virulence factors able to combat multiple host defence mechanisms. Fibrinogen (Fg), a critical component in the host coagulation cascade, plays an important role in the pathogenesis of this bacterium, as it is the target of numerous staphylococcal virulence proteins. Amongst its secreted virulence factors, coagulase (Coa) and Extracellular fibrinogen-binding protein (Efb) share common Fg binding motives and have been described to form a Fg shield around staphylococcal cells, thereby allowing efficient bacterial spreading, phagocytosis escape and evasion of host immune system responses. Targeting these proteins with monoclonal antibodies thus represents a new therapeutic option against S. aureus. To this end, here we report the selection and characterization of fully human, sequence-defined, monoclonal antibodies selected against the C-terminal of coagulase. Given the functional homology between Coa and Efb, we also investigated if the generated antibodies bound the two virulence factors. Thirteen unique antibodies were isolated from naïve antibodies gene libraries by antibody phage display. As anticipated, most of the selected antibodies showed cross-recognition of these two proteins and among them, four were able to block the interaction between Coa/Efb and Fg. Furthermore, our monoclonal antibodies could interact with the two main Fg binding repeats present at the C-terminal of Coa and distinguish them, suggesting the presence of two functionally different Fg-binding epitopes.


Subject(s)
Coagulase , Staphylococcal Infections , Humans , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/metabolism , Bacterial Proteins , Coagulase/immunology , Fibrinogen/chemistry , Fibrinogen/metabolism , Phagocytosis , Staphylococcus aureus , Virulence Factors/metabolism , Binding Sites, Antibody
2.
PLoS Pathog ; 18(1): e1010227, 2022 01.
Article in English | MEDLINE | ID: mdl-35041705

ABSTRACT

The blood-clotting protein fibrin(ogen) plays a critical role in host defense against invading pathogens, particularly against peritoneal infection by the Gram-positive microbe Staphylococcus aureus. Here, we tested the hypothesis that direct binding between fibrin(ogen) and S. aureus is a component of the primary host antimicrobial response mechanism and prevention of secondary microbe dissemination from the peritoneal cavity. To establish a model system, we showed that fibrinogen isolated from FibγΔ5 mice, which express a mutant form lacking the final 5 amino acids of the fibrinogen γ chain (termed fibrinogenγΔ5), did not support S. aureus adherence when immobilized and clumping when in suspension. In contrast, purified wildtype fibrinogen supported robust adhesion and clumping that was largely dependent on S. aureus expression of the receptor clumping factor A (ClfA). Following peritoneal infection with S. aureus USA300, FibγΔ5 mice displayed worse survival compared to WT mice coupled to reduced bacterial killing within the peritoneal cavity and increased dissemination of the microbes into circulation and distant organs. The failure of acute bacterial killing, but not enhanced dissemination, was partially recapitulated by mice infected with S. aureus USA300 lacking ClfA. Fibrin polymer formation and coagulation transglutaminase Factor XIII each contributed to killing of the microbes within the peritoneal cavity, but only elimination of polymer formation enhanced systemic dissemination. Host macrophage depletion or selective elimination of the fibrin(ogen) ß2-integrin binding motif both compromised local bacterial killing and enhanced S. aureus systemic dissemination, suggesting fibrin polymer formation in and of itself was not sufficient to retain S. aureus within the peritoneal cavity. Collectively, these findings suggest that following peritoneal infection, the binding of S. aureus to stabilized fibrin matrices promotes a local, macrophage-mediated antimicrobial response essential for prevention of microbe dissemination and downstream host mortality.


Subject(s)
Fibrinogen/immunology , Peritonitis/immunology , Staphylococcal Infections/immunology , Animals , Coagulase/immunology , Coagulase/metabolism , Fibrin/metabolism , Mice , Peritonitis/metabolism , Staphylococcal Infections/metabolism , Staphylococcus aureus/immunology , Staphylococcus aureus/metabolism
3.
Microb Pathog ; 160: 105171, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34481860

ABSTRACT

Staphylococcus aureus infection is emerging as a global threat because of the highly debilitating nature of the associated disease's unprecedented magnitude of its spread and growing global resistance to antimicrobial medicines. Recently WHO has categorized these bacteria under the high global priority pathogen list and is one of the six nosocomial pathogens termed as ESKAPE pathogens which have emerged as a serious threat to public health worldwide. The development of a specific vaccine can stimulate an optimal antibody response, thus providing immunity against it. Therefore, in the present study efforts have been made to identify potential vaccine candidates from the Clumping factor surface proteins (ClfA and ClfB) of S. aureus. Employing the immunoinformatics approach, fourteen antigenic peptides including T-cell, B-cell epitopes were identified which were non-toxic, non-allergenic, high antigenicity, strong binding efficiency with commonly occurring MHC alleles. Consequently, a multi-epitope vaccine chimera was designed by connecting these epitopes with suitable linkers an adjuvant to enhance immunogenicity. Further, homology modeling and molecular docking were performed to construct the three-dimensional structure of the vaccine and study the interaction between the modeled structure and immune receptor (TLR-2) present on lymphocyte cells. Consequently, molecular dynamics simulation for 100 ns period confirmed the stability of the interaction and reliability of the structure for further analysis. Finally, codon optimization and in silico cloning were employed to ensure the successful expression of the vaccine candidate. As the targeted protein is highly antigenic and conserved, hence the designed novel vaccine construct holds potential against emerging multi-drug-resistant organisms.


Subject(s)
Adhesins, Bacterial/immunology , Coagulase/immunology , Epitopes, B-Lymphocyte , Epitopes, T-Lymphocyte , Staphylococcal Infections , Computational Biology , Humans , Molecular Docking Simulation , Molecular Dynamics Simulation , Peptides , Reproducibility of Results , Staphylococcal Infections/prevention & control , Staphylococcus aureus , Vaccines, Subunit
4.
PLoS Pathog ; 16(8): e1008733, 2020 08.
Article in English | MEDLINE | ID: mdl-32817694

ABSTRACT

Staphylococcus aureus (S. aureus) is one of the most common bacterial infections worldwide, and antibiotic resistant strains such as Methicillin-Resistant S. aureus (MRSA) are a major threat and burden to public health. MRSA not only infects immunocompromised patients but also healthy individuals and has rapidly spread from the healthcare setting to the outside community. However, all vaccines tested in clinical trials to date have failed. Immunocompromised individuals such as patients with HIV or decreased levels of CD4+ T cells are highly susceptible to S. aureus infections, and they are also at increased risk of developing fungal infections. We therefore wondered whether stimulation of antifungal immunity might promote the type of immune responses needed for effective host defense against S. aureus. Here we show that vaccination of mice with a fungal ß-glucan particle (GP) loaded with S. aureus antigens provides protective immunity to S. aureus. We generated glucan particles loaded with the four S. aureus proteins ClfA, IsdA, MntC, and SdrE, creating the 4X-SA-GP vaccine. Vaccination of mice with three doses of 4X-SA-GP promoted protection in a systemic model of S. aureus infection with a significant reduction in the bacterial burden in the spleen and kidneys. 4X-SA-GP vaccination induced antigen-specific Th1 and Th17 CD4+ T cell and antibody responses and provided long-term protection. This work suggests that the GP vaccine system has potential as a novel approach to developing vaccines for S. aureus.


Subject(s)
Saccharomyces cerevisiae/immunology , Staphylococcal Infections/immunology , Staphylococcal Vaccines/immunology , Staphylococcus aureus/immunology , Animals , Antibodies, Bacterial/immunology , Coagulase/administration & dosage , Coagulase/genetics , Coagulase/immunology , Female , Humans , Mice , Mice, Inbred C57BL , Saccharomyces cerevisiae/chemistry , Staphylococcal Infections/microbiology , Staphylococcal Vaccines/administration & dosage , Staphylococcal Vaccines/genetics , Staphylococcus aureus/genetics , Th1 Cells/immunology , Th17 Cells/immunology , Vaccination , beta-Glucans/administration & dosage , beta-Glucans/immunology
5.
Biosci Biotechnol Biochem ; 84(9): 1846-1855, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32501144

ABSTRACT

Here, we prepared the novel combined adjuvants, CTB as intra-molecular adjuvant, CpG and aluminum hydroxide (Alum) to strengthen the immunogenicity of clumping factor A221-550 of Staphylococcus aureus (S. aureus). The protein-immunoactive results showed CTB-ClfA221-550 elicited the strong immune responses to serum from mice immunized with CTB and ClfA221-550, respectively. The mice immunized with CTB-ClfA221-550 plus CpG and Alum adjuvant exhibited significantly stronger CD4+ T cell responses for IFN-γ, IL-2, IL-4, and IL-17 and displayed the higher proliferation response of splenic lymphocytes than the control groups, in addition, these mice generated the strongest humoral immune response against ClfA221-550 among all groups. Our results also showed CTB-ClfA221-550 plus CpG and Alum adjuvant obviously increased the survival percentage of the mice challenged by S. aureus. These data suggested that the novel combined adjuvants, CTB, CpG, and Alum, significantly enhance the immune responses triggered with ClfA221-550, and could provide a new approach against infection of S. aureus. ABBREVIATIONS: CTB: Cholera Toxin B; CpG: Cytosine preceding Guanosine; ODN: Oligodeoxynucleotides; Alum: Aluminum hydroxide; TRAP: Target of RNAIII-activating Protein; TLR9: Toll-like Receptor 9; TMB: 3, 3', 5, 5'-tetramethylbenzidine; mAbs: Monoclonal Antibodies; OD: Optical Densities; S. aureus: Staphylococcus aureus; ClfA: Clumping factor A; FnBPA: Fibronection-binding protein A; IsdB: Iron-regulated surface determinant B; SasA: Staphylococcus aureus Surface Protein A; GapC: Glycer-aldehyde-3-phosphate dehydrogenase-C.


Subject(s)
Adjuvants, Immunologic/pharmacology , Aluminum Hydroxide/pharmacology , Cholera Toxin/pharmacology , Coagulase/immunology , Animals , Cell Proliferation/drug effects , Drug Interactions , Immunization , Lymphocytes/cytology , Lymphocytes/drug effects , Mice , Oligodeoxyribonucleotides/pharmacology
6.
Article in English | MEDLINE | ID: mdl-31138566

ABSTRACT

Surgical site infections (SSIs) are commonly caused by Staphylococcus aureus We report that a combination of three monoclonal antibodies (MEDI6389) that neutralize S. aureus alpha-toxin, clumping factor A, and four leukocidins (LukSF, LukED, HlgAB, and HlgCB) plus vancomycin had enhanced efficacy compared with control antibody plus vancomycin in two mouse models of S. aureus SSI. Therefore, monoclonal antibody-based neutralization of multiple S. aureus virulence factors may provide an adjunctive perioperative approach to combat S. aureus SSIs.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antibodies, Monoclonal/pharmacology , Staphylococcal Infections/drug therapy , Surgical Wound Infection/drug therapy , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Bacterial Proteins/immunology , Broadly Neutralizing Antibodies/pharmacology , Coagulase/immunology , Leukocidins/immunology , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Mice, Inbred C57BL , Mice, Inbred Strains , Staphylococcal Infections/microbiology , Surgical Wound Infection/microbiology , Vancomycin/pharmacology
7.
Microbes Infect ; 21(3-4): 163-169, 2019.
Article in English | MEDLINE | ID: mdl-30447395

ABSTRACT

Coagulase (Coa) secreted by Staphylococcus aureus is associated with the establishment of staphylococcal disease, which activates host prothrombin and generates fibrin shields. The R domain of Coa, consisting of several conserved repeats, is important in immune evasion during S. aureus infection. However, previous research showed that the Coa R domain induced very weak specific antibody responses. In this study, we constructed a new R domain, CoaR6, consisting of 6 repeats that occur most frequently in clinical isolates. By fusing CoaR6 with Hc, the C-terminal fragment of the heavy chain of tetanus neurotoxin, we successfully increased anti-CoaR6 IgG levels in immunized mice which were hardly detected in mice immunized with CoaR6 plus alum. To further improve anti-CoaR6 responses, the combination adjuvants alum plus CpG were formulated with the antigen and exhibited a significantly higher specific antibody response. Moreover, active Th1/Th17 immune responses were observed in Hc-CoaR6 immunized group rather than CoaR6. Active immunization of Hc-CoaR6 with alum plus CpG showed protective effects in a peritonitis model induced by two S. aureus strains with different coagulase types. Our results provided strategies to improve the immunogenicity of R domain and supporting evidences for R domain to be an S. aureus vaccine candidate.


Subject(s)
Bacterial Proteins/immunology , Coagulase/immunology , Staphylococcal Infections/prevention & control , Staphylococcal Vaccines/immunology , Staphylococcus aureus/enzymology , Staphylococcus aureus/immunology , Amino Acid Sequence , Animals , Antibodies, Bacterial/blood , Antigens, Bacterial/immunology , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Coagulase/chemistry , Coagulase/genetics , Coagulase/metabolism , Cytokines/immunology , Immunity, Cellular , Metalloendopeptidases/genetics , Mice , Protein Interaction Domains and Motifs , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , Staphylococcal Infections/immunology , Staphylococcal Vaccines/administration & dosage , Survival Analysis , Tetanus Toxin/genetics , Vaccination
8.
Vaccine ; 36(24): 3513-3521, 2018 06 07.
Article in English | MEDLINE | ID: mdl-29739718

ABSTRACT

Staphylococcus aureus causes a chronic, contagious disease of the udder, or mastitis, in dairy cows. This infection is often refractory to antibiotic treatment, and has a significant economic impact on milk production worldwide. An effective vaccine to prevent S. aureus mastitis would improve animal health, reduce antibiotic dependence and inform human vaccine approaches. The iron-regulated surface determinant A (IsdA) and clumping factor A (ClfA) are conserved S. aureus extracellular-matrix adhesins and target vaccine antigens. Here we report the results of two bovine immunogenicity trials using purified IsdA and ClfA-cholera toxin A2/B chimeras (IsdA-CTA2/B and ClfA-CTA2/B). Cows were intranasally inoculated with IsdA-CTA2/B + ClfA-CTA2/B at dry off and followed for 70 days. Trial 1 utilized three groups with one or two booster doses at a total concentration of 600 or 900 µg. Trial 2 utilized two groups with one booster at a total concentration of 1200 µg. Humoral immune responses in serum and milk were examined by ELISA. Responses in serum were significant between groups and provide evidence of antigen-specific IgG induction after vaccination in both trials. Cellular proliferation was detected by flow cytometry using antigen-stimulated PBMCs from day 60 of Trial 2 and revealed an increase in CD4+ T cells from vaccinated cows. IsdA and ClfA stimulation induced IL-4 expression, but not IFN-γ or IL-17, in PBMCs from day 60 as determined by cytokine expression analysis. Opsonophagocytosis of S. aureus confirmed the functional in vitro activity of anti-IsdA antibodies from Trial 2 serum and milk. The vaccine was well tolerated and safe, and results support the potential of mucosally-delivered CTA2/B chimeras to protect cows from mastitis caused by S. aureus.


Subject(s)
Antibodies, Bacterial/biosynthesis , Mastitis, Bovine/prevention & control , Recombinant Fusion Proteins/immunology , Staphylococcal Infections/prevention & control , Staphylococcal Infections/veterinary , Staphylococcal Vaccines/biosynthesis , Administration, Intranasal , Animals , Antibodies, Bacterial/blood , Antigens, Bacterial/administration & dosage , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/microbiology , Cattle , Cell Proliferation/drug effects , Cholera Toxin/administration & dosage , Cholera Toxin/genetics , Cholera Toxin/immunology , Coagulase/administration & dosage , Coagulase/genetics , Coagulase/immunology , Female , Gene Expression , Immunity, Humoral/drug effects , Immunization, Secondary/methods , Immunogenicity, Vaccine , Immunoglobulin G/biosynthesis , Immunoglobulin G/blood , Interleukin-4/biosynthesis , Interleukin-4/metabolism , Mammary Glands, Animal/immunology , Mammary Glands, Animal/microbiology , Mastitis, Bovine/immunology , Mastitis, Bovine/microbiology , Milk/chemistry , Milk/immunology , Milk/microbiology , Protein Isoforms/administration & dosage , Protein Isoforms/genetics , Protein Isoforms/immunology , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Staphylococcal Vaccines/administration & dosage , Staphylococcal Vaccines/genetics , Staphylococcus aureus/drug effects , Staphylococcus aureus/immunology , Staphylococcus aureus/pathogenicity
9.
Vet J ; 235: 47-53, 2018 05.
Article in English | MEDLINE | ID: mdl-29704938

ABSTRACT

Staphylococcus aureus is a worldwide pathogen that causes mastitis in dairy herds. Shortcomings in control programs have encouraged the development of vaccines against this pathogen. This study evaluated the vaccine candidate VacR, which included recombinant S. aureus protein clumping factor A (rClf), fibronectin binding protein A (rFnBP) and hemolysin beta (rBt), formulated with a novel immune-stimulating complex. Comparisons were made between healthy pregnant heifers that received either VacR (n=8; VacR group) or phosphate buffered saline (PBS) plus adjuvant (control group) SC in the supramammary lymph node area on days 45 and 15 before the expected calving date. Blood and foremilk samples were collected from 7 to 60days post-calving. After calving, heifers in the VacR group produced higher total IgG (IgGtotal) titers against each component, in both serum (rBt, 3.4×105; rClf, 3.1×105; rFnBP, 2.3×105) and milk (rBt, 2.6×104; rClf, 1.3×104; rFnBP, 1.1×104), than control heifers (P<0.0001). There were increased concentrations of IgG1 and IgG2 in VacR group (P<0.05), in both serum and milk. Humoral responses remained high throughout the period most susceptible to intramammary infections (P<0.01). Antibodies produced against S. aureus rClf and rFnBP reduced bacterial adherence to fibronectin and fibrinogen by 73% and 67%, respectively (P<0.001). Milk antibodies against these adhesins inhibited S. aureus invasion of a mammary epithelial cell line (MAC-T), resulting in 15.7% of bacteria internalized (P<0.0001). There was an approximately 6-fold reduction in the hemolysis titer for the native hemolysin in the VacR group compared to the control group (P<0.0001) and a significantly increase in the proportion of positive neutrophils (VacR, 29.7%; PBS, 13.1%) and the mean fluorescent index (VacR, 217.4; PBS, 152.6; P<0.01) in the VacR group. The results suggest that VacR is a valuable vaccine candidate against S. aureus infections, and merits further field trials and experimental challenges.


Subject(s)
Staphylococcal Infections/veterinary , Staphylococcus aureus/immunology , Vaccines, Synthetic/immunology , ATP-Binding Cassette Transporters/immunology , Adhesins, Bacterial/immunology , Animals , Antibodies, Bacterial/analysis , Antibodies, Bacterial/blood , Cattle , Coagulase/immunology , Female , Hemolysin Proteins/immunology , Immunity, Humoral , Mastitis, Bovine/microbiology , Milk/immunology , Pregnancy , Staphylococcal Infections/prevention & control
10.
Can J Vet Res ; 82(1): 48-54, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29382968

ABSTRACT

To obtain immunogenic conjugate antigens, adipic acid dihydrazide (ADH), as a bridge, and 1-ethyl-3-(3-dimethylaminopropyl) carbodiimidehydrochloride (EDAC), as a coupling agent, were used to conjugate the purified fusion protein, clumping factor A-fibronectin binding protein ClfA A-FnBPA, and type 5 capsular polysaccharide (CP5). The conjugates were mixed with an adjuvant, and mice were immunized 3 times and challenged with Staphylococcus aureus 1 week later. Antibody titers were determined by indirect enzyme-linked immunosorbent assay (ELISA). At 14 days after the first immunization, antibodies against the purified protein and conjugate were detected; after 28 days, antibody levels increased; and a week after the third immunization, antibody levels continued to increase. However, the conjugate antibody titers were higher than those of the purified protein during the study, and no IgG antibodies against purified CP5 were detected during the entire experiment. The protection rate increased to 90% in the conjugate group, indicating that the conjugate imparts a relatively higher protective efficacy than the purified protein and purified CP5.


Afin d'obtenir des antigènes conjugués immunogéniques, du dihydrazide adipique acide (DHA), comme pont, et le 1-éthyl-3-(3-diméthylaminopropyl) carbodiimidehydhrochlorure (EDAC), comme agent liguant, ont été utilisés pour conjuguer la protéine de fusion purifiée ClfAA-FnBPA (facteur de coagulation A-protéine liant la fibronectine) et le polysaccharide capsulaire de type 5 (PC5). Les conjugués ont été mélangés avec un adjuvant, et des souris immunisées trois fois et soumis à une infection défi par Staphylococcus aureus 1 semaine plus tard. Les titres d'anticorps ont été déterminés par épreuve immuno-enzymatique indirecte (ELISA). Au jour 14 suivant la première immunisation, des anticorps dirigés contre la protéine purifiée et le conjugué ont été détectés; après 28 jours, les taux d'anticorps ont augmenté, et 1 semaine après la troisième immunisation, les taux d'anticorps ont continué d'augmenter. Toutefois, durant l'étude les titres d'anticorps envers le conjugué étaient supérieurs à ceux de la protéine purifiée, et aucun anticorps de type IgG dirigé contre le PC5 purifié ne fut détecté durant l'expérience entière. Le taux de protection augmenta jusqu'à 90 % dans le groupe du conjugué, indiquant que le conjugué entraîne une protection efficace supérieure à la protéine purifiée et au PC5 purifié.(Traduit par Docteur Serge Messier).


Subject(s)
Adhesins, Bacterial/immunology , Bacterial Capsules/immunology , Bacterial Vaccines/immunology , Coagulase/immunology , Staphylococcal Infections/prevention & control , Staphylococcus aureus/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Antibodies, Bacterial/blood , Antigens, Bacterial/immunology , Cloning, Molecular , Female , Mice , Mice, Inbred BALB C , Random Allocation , Staphylococcal Infections/microbiology
11.
Infect Immun ; 85(12)2017 12.
Article in English | MEDLINE | ID: mdl-28947645

ABSTRACT

Staphylococcus aureus has become increasingly resistant to antibiotics, and vaccines offer a potential solution to this epidemic of antimicrobial resistance. Targeting of specific T cell subsets is now considered crucial for next-generation anti-S. aureus vaccines; however, there is a paucity of information regarding T cell antigens of S. aureus This study highlights the importance of cell wall-anchored proteins as human CD4+ T cell activators capable of driving antigen-specific Th1 and Th17 cell activation. Clumping factor A (ClfA), which contains N1, N2, and N3 binding domains, was found to be a potent human T cell activator. We further investigated which subdomains of ClfA were involved in T cell activation and found that the full-length ClfA N123 and N23 were potent Th1 and Th17 activators. Interestingly, the N1 subdomain was capable of exclusively activating Th1 cells. Furthermore, when these subdomains were used in a model vaccine, N23 and N1 offered Th1- and Th17-mediated systemic protection in mice upon intraperitoneal challenge. Overall, however, full-length ClfA N123 is required for maximal protection both locally and systemically.


Subject(s)
Antigens, Bacterial/immunology , Coagulase/immunology , Staphylococcus aureus/immunology , T-Lymphocytes/immunology , Animals , Cells, Cultured , Disease Models, Animal , Humans , Mice, Inbred C57BL , Staphylococcal Infections/prevention & control , Staphylococcal Vaccines/administration & dosage , Staphylococcal Vaccines/immunology , Survival Analysis
12.
Article in English | MEDLINE | ID: mdl-28584141

ABSTRACT

Secreted alpha-toxin and surface-localized clumping factor A (ClfA) are key virulence determinants in Staphylococcus aureus bloodstream infections. We previously demonstrated that prophylaxis with a multimechanistic monoclonal antibody (MAb) combination against alpha-toxin (MEDI4893*) and ClfA (11H10) provided greater strain coverage and improved efficacy in an S. aureus lethal bacteremia model. Subsequently, 11H10 was found to exhibit reduced affinity and impaired inhibition of fibrinogen binding to ClfA002 expressed by members of a predominant hospital-associated methicillin-resistant S. aureus (MRSA) clone, ST5. Consequently, we identified another anti-ClfA MAb (SAR114) from human tonsillar B cells with >100-fold increased affinity for three prominent ClfA variants, including ClfA002, and potent inhibition of bacterial agglutination by 112 diverse clinical isolates. We next constructed bispecific Abs (BiSAbs) comprised of 11H10 or SAR114 as IgG scaffolds and grafted anti-alpha-toxin (MEDI4893*) single-chain variable fragment to the amino or carboxy terminus of the anti-ClfA heavy chains. Although the BiSAbs exhibited in vitro potencies similar to those of the parental MAbs, only 11H10-BiSAb, but not SAR114-BiSAb, showed protective activity in murine infection models comparable to the respective MAb combination. In vivo activity with SAR114-BiSAb was observed in infection models with S. aureus lacking ClfA. Our data suggest that high-affinity binding to ClfA sequesters the SAR114-BiSAb to the bacterial surface, thereby reducing both alpha-toxin neutralization and protection in vivo These results indicate that a MAb combination targeting ClfA and alpha-toxin is more promising for future development than the corresponding BiSAb.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/therapeutic use , Bacteremia/drug therapy , Bacterial Toxins/immunology , Coagulase/immunology , Hemolysin Proteins/immunology , Methicillin-Resistant Staphylococcus aureus/drug effects , Staphylococcal Infections/drug therapy , Animals , Antibodies, Bacterial/immunology , Antibodies, Bacterial/therapeutic use , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Humanized , Antibodies, Neutralizing/immunology , Bacteremia/microbiology , Broadly Neutralizing Antibodies , Female , Methicillin-Resistant Staphylococcus aureus/immunology , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Staphylococcal Infections/immunology , Virulence Factors
13.
EBioMedicine ; 13: 328-338, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27789272

ABSTRACT

The Staphylococcus aureus fibrinogen binding MSCRAMM (Microbial Surface Components Recognizing Adhesive Matrix Molecules), ClfA (clumping factor A) is an important virulence factor in staphylococcal infections and a component of several vaccines currently under clinical evaluation. The mouse monoclonal antibody aurexis (also called 12-9), and the humanized version tefibazumab are therapeutic monoclonal antibodies targeting ClfA that in combination with conventional antibiotics were effective in animal models but showed less impressive efficacy in a limited Phase II clinical trial. We here report the crystal structure and a biochemical characterization of the ClfA/tefibazumab (Fab) complex. The epitope for tefibazumab is located to the "top" of the N3 subdomain of ClfA and partially overlaps with a previously unidentified second binding site for fibrinogen. A high-affinity binding of ClfA to fibrinogen involves both an interaction at the N3 site and the previously identified docking of the C-terminal segment of the fibrinogen γ-chain in the N2N3 trench. Although tefibazumab binds ClfA with high affinity we observe a modest IC50 value for the inhibition of fibrinogen binding to the MSCRAMM. This observation, paired with a common natural occurring variant of ClfA that is not effectively recognized by the mAb, may partly explain the modest effect tefibazumab showed in the initial clinic trail. This information will provide guidance for the design of the next generation of therapeutic anti-staphylococcal mAbs targeting ClfA.


Subject(s)
Antibodies, Monoclonal/chemistry , Coagulase/chemistry , Models, Molecular , Protein Conformation , Amino Acid Substitution , Antibodies, Monoclonal/metabolism , Binding Sites , Coagulase/immunology , Coagulase/metabolism , Crystallography, X-Ray , Epitopes/chemistry , Epitopes/immunology , Epitopes/metabolism , Fibrinogen/chemistry , Fibrinogen/metabolism , Humans , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry
14.
J Infect ; 73(5): 437-454, 2016 11.
Article in English | MEDLINE | ID: mdl-27519620

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.


Subject(s)
Bacterial Capsules/immunology , Bacterial Vaccines/pharmacology , Staphylococcal Infections/immunology , Staphylococcal Infections/prevention & control , Staphylococcus aureus/immunology , Adolescent , Age Distribution , Aged , Aged, 80 and over , Antigens, Viral , Coagulase/immunology , Diphtheria Toxin/immunology , Double-Blind Method , Female , Humans , Immunization, Secondary , Male , Middle Aged , Placebos , Vaccination/methods , Young Adult
15.
mBio ; 7(3)2016 06 28.
Article in English | MEDLINE | ID: mdl-27353753

ABSTRACT

UNLABELLED: Staphylococcus aureus produces numerous virulence factors, each contributing different mechanisms to bacterial pathogenesis in a spectrum of diseases. Alpha toxin (AT), a cytolytic pore-forming toxin, plays a key role in skin and soft tissue infections and pneumonia, and a human anti-AT monoclonal antibody (MAb), MEDI4893*, has been shown to reduce disease severity in dermonecrosis and pneumonia infection models. However, interstrain diversity and the complex pathogenesis of S. aureus bloodstream infections suggests that MEDI4893* alone may not provide adequate protection against S. aureus sepsis. Clumping factor A (ClfA), a fibrinogen binding protein, is an important virulence factor facilitating S. aureus bloodstream infections. Herein, we report on the identification of a high-affinity anti-ClfA MAb, 11H10, that inhibits ClfA binding to fibrinogen, prevents bacterial agglutination in human plasma, and promotes opsonophagocytic bacterial killing (OPK). 11H10 prophylaxis reduced disease severity in a mouse bacteremia model and was dependent on Fc effector function and OPK. Additionally, prophylaxis with 11H10 in combination with MEDI4893* provided enhanced strain coverage in this model and increased survival compared to that obtained with the individual MAbs. The MAb combination also reduced disease severity in murine dermonecrosis and pneumonia models, with activity similar to that of MEDI4893* alone. These results indicate that an MAb combination targeting multiple virulence factors provides benefit over a single MAb neutralizing one virulence mechanism by providing improved efficacy, broader strain coverage, and protection against multiple infection pathologies. IMPORTANCE: Alternative strategies to broad-spectrum antibiotics are required to combat the antibiotic resistance epidemic. Previous attempts at active or passive immunization against Staphylococcus aureus targeting single antigens have failed in clinical trials despite positive preclinical data. To provide broad disease and isolate coverage, an effective immunization strategy likely must target multiple virulence mechanisms of the pathogen. Herein, we tested a multimechanistic MAb combination targeting alpha toxin (AT) and clumping factor A (ClfA) that neutralizes AT-mediated cytotoxicity, blocks fibrinogen binding by ClfA, prevents bacterial agglutination, targets the bacteria for opsonophagocytic killing, and provides broad isolate coverage in a lethal-bacteremia model. Although each MAb alone was effective in bacteremia against some individual isolates, the MAb combination provided improved protection against other isolates. These results illustrate the importance of targeting multiple virulence mechanisms and highlight the potential for an MAb combination targeting AT and ClfA to effectively prevent S. aureus disease.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , Bacterial Toxins/immunology , Coagulase/immunology , Staphylococcal Infections/immunology , Staphylococcal Infections/prevention & control , Staphylococcus aureus/immunology , Virulence Factors/immunology , Animals , Antibodies, Bacterial/blood , Antibodies, Bacterial/therapeutic use , Antibodies, Monoclonal/isolation & purification , Antibodies, Neutralizing/therapeutic use , Bacterial Load , Disease Models, Animal , HL-60 Cells , Humans , Immunization, Passive/methods , Mice , Phagocytosis , Staphylococcal Infections/blood , Staphylococcal Infections/microbiology
16.
mBio ; 7(2): e00225, 2016 Mar 08.
Article in English | MEDLINE | ID: mdl-26956591

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.


Subject(s)
Coagulase/immunology , Staphylococcal Infections/prevention & control , Staphylococcal Vaccines/immunology , Staphylococcus aureus/immunology , Animals , Disease Models, Animal , Drug Evaluation, Preclinical , Mice , Staphylococcal Vaccines/isolation & purification , Treatment Outcome , Vaccines, Subunit/immunology , Vaccines, Subunit/isolation & purification
18.
Sci Rep ; 6: 20929, 2016 Feb 11.
Article in English | MEDLINE | ID: mdl-26865417

ABSTRACT

Staphylococcus aureus causes serious sepsis and necrotic pneumonia worldwide. Due to the spread of multidrug-resistant strains, developing an effective vaccine is the most promising method for combating S. aureus infection. In this study, based on the immune-dominant areas of the iron surface determinant B (IsdB) and clumping factor A (ClfA), we designed the novel chimeric vaccine IsdB151-277ClfA33-213 (IC). IC formulated with the AlPO4 adjuvant induced higher protection in an S. aureus sepsis model compared with the single components alone and showed broad immune protection against several clinical S. aureus isolates. Immunisation with IC induced strong antibody responses. The protective effect of antibodies was demonstrated through the opsonophagocytic assay (OPA) and passive immunisation experiment. Moreover, this new chimeric vaccine induced Th1/Th17-skewed cellular immune responses based on cytokine profiles and CD4(+) T cell stimulation tests. Neutralisation of IL-17A alone (but not IFN-γ) resulted in a significant decrease in vaccine immune protection. Finally, we found that IC showed protective efficacy in a pneumonia model. Taken together, these data provide evidence that IC is a potentially promising vaccine candidate for combating S. aureus sepsis and pneumonia.


Subject(s)
Antibodies, Bacterial/biosynthesis , Pneumonia/prevention & control , Recombinant Fusion Proteins/administration & dosage , Sepsis/prevention & control , Staphylococcal Infections/prevention & control , Staphylococcal Vaccines/administration & dosage , Adjuvants, Immunologic/administration & dosage , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Cation Transport Proteins/genetics , Cation Transport Proteins/immunology , Cloning, Molecular , Coagulase/genetics , Coagulase/immunology , Disease Models, Animal , Escherichia coli/genetics , Escherichia coli/metabolism , Female , Gene Expression , Humans , Immunization , Interleukin-17/antagonists & inhibitors , Interleukin-17/genetics , Interleukin-17/immunology , Mice, Inbred BALB C , Mice, Inbred C57BL , Pneumonia/immunology , Pneumonia/microbiology , Pneumonia/mortality , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Sepsis/immunology , Sepsis/microbiology , Sepsis/mortality , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Staphylococcal Infections/mortality , Staphylococcal Vaccines/biosynthesis , Staphylococcal Vaccines/genetics , Staphylococcus aureus/drug effects , Staphylococcus aureus/growth & development , Staphylococcus aureus/pathogenicity , Survival Analysis
19.
J Exp Med ; 213(3): 293-301, 2016 Mar 07.
Article in English | MEDLINE | ID: mdl-26880578

ABSTRACT

Host immunity against bacteria typically involves antibodies that recognize the microbial surface and promote phagocytic killing. Methicillin-resistant Staphylococcus aureus (MRSA) is a frequent cause of lethal bloodstream infection; however, vaccines and antibody therapeutics targeting staphylococcal surface molecules have thus far failed to achieve clinical efficacy. S. aureus secretes coagulase (Coa), which activates host prothrombin and generates fibrin fibrils that protect the pathogen against phagocytosis by immune cells. Because of negative selection, the coding sequence for the prothrombin-binding D1-D2 domain is highly variable and does not elicit cross-protective immune responses. The R domain, tandem repeats of a 27-residue peptide that bind fibrinogen, is conserved at the C terminus of all Coa molecules, but its functional significance is not known. We show here that the R domain enables bloodstream infections by directing fibrinogen to the staphylococcal surface, generating a protective fibrin shield that inhibits phagocytosis. The fibrin shield can be marked with R-specific antibodies, which trigger phagocytic killing of staphylococci and protect mice against lethal bloodstream infections caused by a broad spectrum of MRSA isolates. These findings emphasize the critical role of coagulase in staphylococcal escape from opsonophagocytic killing and as a protective antigen for S. aureus vaccines.


Subject(s)
Antibodies, Bacterial/immunology , Coagulase/immunology , Phagocytosis , Staphylococcus aureus/immunology , Agglutination , Animals , Antibodies, Monoclonal/immunology , Coagulase/chemistry , Female , Fibrin/metabolism , Humans , Mice, Inbred BALB C , Opsonin Proteins/metabolism , Protein Structure, Tertiary , Repetitive Sequences, Amino Acid , Staphylococcal Infections/blood , Staphylococcal Infections/immunology , Staphylococcal Infections/prevention & control
20.
mBio ; 7(1): e02232-15, 2016 Feb 02.
Article in English | MEDLINE | ID: mdl-26838725

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.


Subject(s)
Antigens, Bacterial/immunology , Coagulase/immunology , Staphylococcal Infections/prevention & control , Staphylococcal Vaccines/immunology , Abscess/microbiology , Adjuvants, Immunologic/administration & dosage , Animals , Antibodies, Bacterial/blood , Antibodies, Bacterial/therapeutic use , Bacterial Load , Drug Evaluation, Preclinical , Mice , Mice, Inbred C57BL , Placebos/administration & dosage , Rats, Sprague-Dawley , Sepsis/mortality , Staphylococcal Vaccines/administration & dosage , Survival Analysis , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...