Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 8 de 8
1.
EClinicalMedicine ; 67: 102404, 2024 Jan.
Article En | MEDLINE | ID: mdl-38274114

Background: Toxic shock syndrome toxin-1 (TSST-1) is a superantigen produced by Staphylococcus aureus that causes the life-threatening toxic shock syndrome. The development of a safe and immunogenic vaccine against TSST-1 remains an unmet medical need. We investigated the safety, tolerability and immunogenicity of a recombinant TSST-1 variant vaccine (rTSST-1v) after 1-3 injections in healthy volunteers. Methods: In this randomised, double-blind, adjuvant-controlled, parallel-group, phase 2 trial, healthy adults aged 18-64 were randomly allocated to undergo 1-3 injections of either 10 or 100 µg rTSST-1v or Al(OH)3. The primary endpoint was safety and tolerability of rTSST-1v in the intention-to-treat population. The per-protocol population was used for the immunogenicity analysis. The trial is registered with EudraCT#: 2015-003714-24; ClinicalTrials.gov#: NCT02814708. Findings: Between April and November 2017,140 subjects were enrolled and 126 completed the trial. rTSST-1v showed a good safety and tolerability profile. A total of 855 systemic adverse events occurred, 280 of which were suspected related adverse events, without dose dependency. Two participants were discontinued early because of allergic reactions. Seroconversion occurred in >81% of subjects within 3 months of the first immunisation which was sustained until 18 months after the third immunisation in over 70% of subjects in the pooled low-dose group and in over 85% in the pooled high-dose group. Interpretation: rTSST-1v in cumulative doses of up to 300 µg was safe, well-tolerated and highly immunogenic. Two immunisations with 100 µg rTSST-1v provided the most persistent immune response and may be evaluated in future trials. Funding: Biomedizinische Forschung & Bio-Produkte AG funded this study.

2.
Toxins (Basel) ; 14(9)2022 08 29.
Article En | MEDLINE | ID: mdl-36136533

Staphylococcus aureus is a human and animal pathogen as well as a commensal bacterium. It can be a causative agent of severe, life-threatening infections with high mortality, e.g., toxic shock syndrome, septic shock, and multi-organ failure. S. aureus strains secrete a number of toxins. Exotoxins/enterotoxins are considered important in the pathogenesis of the above-mentioned conditions. Exotoxins, e.g., superantigen toxins, cause uncontrolled and polyclonal T cell activation and unregulated activation of inflammatory cytokines. Here we show the importance of genomic analysis of infectious strains in order to identify disease-causing exotoxins. Further, we show through functional analysis of superantigenic properties of staphylococcal exotoxins that even very small amounts of a putative superantigenic contaminant can have a significant mitogenic effect. The results show expression and production of two distinct staphylococcal exotoxins, SEC and SEL, in several strains from clinical isolates. Antibodies against both toxins are required to neutralise the superantigenic activity of staphylococcal supernatants and purified staphylococcal toxins.


Shock, Septic , Staphylococcal Infections , Animals , Cytokines/metabolism , Enterotoxins/genetics , Enterotoxins/toxicity , Exotoxins/genetics , Humans , Staphylococcal Infections/microbiology , Staphylococcus aureus/metabolism , Superantigens/genetics , Superantigens/toxicity
3.
Toxins (Basel) ; 12(10)2020 10 02.
Article En | MEDLINE | ID: mdl-33023185

Staphylococcal superantigen toxins lead to a devastating cytokine storm resulting in shock and multi-organ failure. We have previously assessed the safety and immunogenicity of a recombinant toxic shock syndrome toxin 1 variant vaccine (rTSST-1v) in clinical trials (NCT02971670 and NCT02340338). The current study assessed neutralizing antibody titers after repeated vaccination with escalating doses of rTSST-1v. At study entry, 23 out of 34 subjects (67.6%) had neutralizing antibody titers inhibiting T cell activation as determined by 3H-thymidine incorporation at a serum dilution of ≤1:100 with similar figures for inhibition of IL-2 activation (19 of 34 subjects, 55.9%) as assessed by quantitative PCR. After the first vaccination, numbers of subjects with neutralization titers inhibiting T cell activation (61.7% ≥ 1:1000) and inhibiting IL-2 gene induction (88.2% ≥ 1:1000) increased. The immune response was augmented after the second vaccination (inhibiting T cell activation: 78.8% ≥ 1:1000; inhibiting IL-2 induction: 93.9% ≥ 1:1000) corroborated with a third immunization months later in a small subgroup of subjects. Assessment of IFNγ, TNFα and IL-6 inhibition revealed similar results, whereas neutralization titers did not change in placebo participants. Antibody titer studies show that vaccination with rTSST-1v in subjects with no/low neutralizing antibodies can rapidly induce high titer neutralizing antibodies persisting over months.


Antibodies, Bacterial/blood , Antibodies, Neutralizing/blood , Bacterial Toxins/administration & dosage , Cytokine Release Syndrome/prevention & control , Enterotoxins/administration & dosage , Immunogenicity, Vaccine , Shock, Septic/prevention & control , Staphylococcal Infections/prevention & control , Staphylococcal Vaccines/administration & dosage , Staphylococcus aureus/drug effects , Superantigens/administration & dosage , Bacterial Toxins/genetics , Bacterial Toxins/immunology , Cells, Cultured , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/microbiology , Cytokines/genetics , Cytokines/metabolism , Double-Blind Method , Enterotoxins/genetics , Enterotoxins/immunology , Humans , Lymphocyte Activation/drug effects , Prospective Studies , Shock, Septic/immunology , Shock, Septic/microbiology , Single-Blind Method , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Staphylococcal Vaccines/genetics , Staphylococcal Vaccines/immunology , Staphylococcus aureus/immunology , Staphylococcus aureus/pathogenicity , Superantigens/genetics , Superantigens/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Time Factors , Treatment Outcome , Vaccination , Vaccines, Synthetic/administration & dosage
4.
Toxins (Basel) ; 8(11)2016 10 28.
Article En | MEDLINE | ID: mdl-27801832

Among the toxin family of bacterial superantigens, the six members of the enterotoxin gene cluster (egc) seem to have unusual characteristics. They are present in the majority of Staphylococcus aureus strains, but their role in disease remains uncertain. We assessed secretion levels, immunogenicity, and toxicity of native and recombinant egc proteins. After having developed enzyme-linked immunosorbent assays, we found different quantities of egc proteins secreted by bacterial isolates. Supernatants induced proliferation of human peripheral blood mononuclear cells. However, purified recombinant egc proteins were shown to have differing superantigenicity potentials. Immunization with identical amounts of all members of egc, and the prominent toxic agent SEB, resulted in neutralizing antisera. Two egc proteins, SEI and SElN, were found to play a predominant role within the cluster. Both displayed the highest potential to activate blood cells, and were essential to be neutralized in supernatants. The application of a supernatant of a strain bearing only egc was sufficient for a lethal outcome in a rabbit model. Again, neutralization of SEI and SElN led to the survival of all tested animals. Finally, nanogram amounts of purified rSEI and rSElN led to lethality in vivo, pointing out the importance of both as virulence determinants among egc superantigens.


Bacterial Proteins , Bacterial Toxins , Enterotoxins , Staphylococcus aureus , Superantigens , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/pharmacology , Bacterial Toxins/genetics , Bacterial Toxins/metabolism , Bacterial Toxins/pharmacology , Cell Proliferation/drug effects , Cells, Cultured , Enterotoxins/genetics , Enterotoxins/metabolism , Enterotoxins/pharmacology , Humans , Leukocytes, Mononuclear/drug effects , Rabbits , Recombinant Proteins/pharmacology , Staphylococcus aureus/genetics , Staphylococcus aureus/isolation & purification , Staphylococcus aureus/metabolism , Superantigens/genetics , Superantigens/metabolism , Superantigens/pharmacology
5.
Lancet Infect Dis ; 16(9): 1036-1044, 2016 09.
Article En | MEDLINE | ID: mdl-27296693

BACKGROUND: Staphylococcal toxic shock syndrome is a superantigen-driven potentially life-threatening disease affecting mainly young and otherwise healthy individuals. Currently, no specific treatment or preventive measure is available. We aimed to assess the safety, tolerability, and immunogenicity of a recombinant detoxified toxic shock syndrome toxin-1 variant (rTSST-1v) vaccine in adult volunteers. METHODS: In this randomised, double-blind, adjuvant-controlled, dose-escalation first-in-human trial, healthy adults aged 18-64 years were enrolled from the Medical University of Vienna, Austria. Participants were randomly assigned (2:1 and 3:1) by block randomisation (block sizes of three and 12) to receive increasing doses of rTSST-1v (100 ng to 30 µg) or the adjuvant comparator aluminium hydroxide (Al(OH)3) (200 µg, 600 µg, or 1 mg). Investigators and participants were masked to group allocation. The per-protocol population received a booster immunisation 42 days after the first vaccination. The primary endpoint was safety and tolerability of rTSST-1v. The per-protocol population included all participants who had adhered to the study protocol without any major protocol deviations. The per-protocol population was the primary analysis population for immunogenicity. The trial is registered with EudraCT, number 2013-003716-50, and ClinicalTrials.gov, number NCT02340338. FINDINGS: Between Aug 19, 2014, and April 14, 2015, 46 participants were enrolled (safety population), of whom three were assigned to cohort 1 (two to receive 100 ng rTSST-1v and one to receive 200 µg Al(OH)3), three to cohort 2 (two to receive 300 ng rTSST-1v and one to receive 600 µg Al(OH)3), four to cohort 3 (three to receive 1 µg rTSST-1v and one to receive 1 mg Al(OH)3), 12 to cohort 4 (nine to receive 3 µg rTSST-1v and three to receive 1 mg Al(OH)3), 12 to cohort 5 (nine to receive 10 µg rTSST-1v and three to receive 1 mg Al(OH)3), and 12 to cohort 6 (nine to receive 300 µg rTSST-1v and three to receive 1 mg Al(OH)3). 45 participants (98%) were included in the per-protocol population. rTSST-1v had a good safety profile, and no vaccination-related severe or serious adverse events occurred. Adverse event rates were similar between participants who received rTSST-1v and those who received placebo (26 [76%] vs 10 [83%]; p=0·62) independent of pre-existing TSST-1 immunity. INTERPRETATION: rTSST-1v was safe, well-tolerated, and immunogenic. This study represents an important step in vaccine development to prevent or treat a potentially lethal disease. FUNDING: Biomedizinische Forschungs GmbH.


Adjuvants, Immunologic/administration & dosage , Immunization, Secondary/methods , Shock, Septic/prevention & control , Vaccines, Synthetic/administration & dosage , Adolescent , Adult , Bacterial Toxins/isolation & purification , Double-Blind Method , Enterotoxins/isolation & purification , Female , Healthy Volunteers , Humans , Immunization Schedule , Male , Middle Aged , Superantigens/isolation & purification , Vaccination , Vaccines, Synthetic/adverse effects , Vaccines, Synthetic/immunology , Young Adult
6.
BMC Microbiol ; 16: 13, 2016 Feb 01.
Article En | MEDLINE | ID: mdl-26830934

BACKGROUND: Nosocomial infections caused by the bacterial pathogen Staphylococcus aureus can lead to serious complications due to the varying presence of secreted toxins. Comparative studies of genomic information and production rates are needed to assess the pathogenic potential of isolated strains. Genotypic and phenotypic profiling of clinical and colonising isolates of S. aureus was used to characterise the release of exotoxins. Blood isolates were compared with colonisation strains to determine similarities and differences of single strains and clusters. RESULTS: Fifty-one fresh isolates obtained from colonised individuals (n = 29) and S. aureus bacteremia (SAB) patients (n = 22) were investigated. The prevalence of genes encoding for three cytolysins (alpha/beta/gamma toxin) and twenty-four superantigens (SEA-SElX) was determined. Isolates exhibited eighteen distinct combinations of superantigens. Sequence analysis identified mutated open reading frames in hla in 13.7% of all strains, in selw (92.2%) and in selx (15.7%). All corrupted genes were associated with specific clonal complexes. Functional assessment of alpha toxin activity by a rabbit erythrocyte lysis assay revealed that supernatants lacking alpha toxin still displayed hemolysis. This was due to the presence of gamma toxin, as proven by inhibition experiments using antisera raised against the respective recombinant proteins. Alpha toxin, SEC, and TSST1 production was quantified by enzyme-linked immunosorbent assays on supernatants of all hla, sec, and tst positive isolates. Blood isolates and colonising strains showed comparable amounts of secreted proteins within a wide range. Agr types I to IV were identified, but did not allow a prediction of high or low production rates. In contrast, alpha toxin production rates between distinct clonal complexes clearly differed. Spa typing was performed and revealed thirty-two unique spa gene patterns and eight small clusters comprising nineteen isolates. Recognised spa-typing clusters displayed highly similar production rates. CONCLUSION: Production rates of the three most prevalent exotoxins varied within both groups of blood isolates and colonising strains. By comparing genotypes and secretion, we found that identical complex gene patterns did not allow predictions of toxin production and function. However, identification of spa typing clusters was suitable to predict similar quantities of released exotoxins.


Bacterial Proteins/genetics , Bacterial Toxins/metabolism , Hemolytic Agents/metabolism , Staphylococcal Infections/microbiology , Staphylococcus aureus/genetics , Animals , Bacterial Proteins/metabolism , Bacterial Toxins/toxicity , Erythrocytes/cytology , Erythrocytes/drug effects , Genotype , Hemolytic Agents/toxicity , Humans , Phenotype , Rabbits , Staphylococcus aureus/classification , Staphylococcus aureus/isolation & purification , Staphylococcus aureus/metabolism
7.
Toxins (Basel) ; 6(6): 1724-41, 2014 May 30.
Article En | MEDLINE | ID: mdl-24887085

Toxic shock syndrome (TSS) results from the host's overwhelming inflammatory response and cytokine storm mainly due to superantigens (SAgs). There is no effective specific therapy. Application of immunoglobulins has been shown to improve the outcome of the disease and to neutralize SAgs both in vivo and in vitro. However, in most experiments that have been performed, antiserum was either pre-incubated with SAg, or both were applied simultaneously. To mirror more closely the clinical situation, we applied a multiple dose (over five days) lethal challenge in a rabbit model. Treatment with toxic shock syndrome toxin 1 (TSST-1) neutralizing antibody was fully protective, even when administered late in the course of the challenge. Kinetic studies on the effect of superantigen toxins are scarce. We performed in vitro kinetic studies by neutralizing the toxin with antibodies at well-defined time points. T-cell activation was determined by assessing T-cell proliferation (3H-thymidine incorporation), determination of IL-2 release in the cell supernatant (ELISA), and IL-2 gene activation (real-time PCR (RT-PCR)). Here we show that T-cell activation occurs continuously. The application of TSST-1 neutralizing antiserum reduced IL-2 and TNFα release into the cell supernatant, even if added at later time points. Interference with the prolonged stimulation of proinflammatory cytokines is likely to be in vivo relevant, as postexposure treatment protected rabbits against the multiple dose lethal SAg challenge. Our results shed new light on the treatment of TSS by specific antibodies even at late stages of exposure.


Antibodies, Neutralizing/therapeutic use , Antitoxins/therapeutic use , Bacterial Toxins/antagonists & inhibitors , Disease Models, Animal , Enterotoxins/antagonists & inhibitors , Shock, Septic/drug therapy , Animals , Antibodies, Neutralizing/pharmacology , Antitoxins/pharmacology , Bacterial Toxins/genetics , Bacterial Toxins/metabolism , Bacterial Toxins/toxicity , Cell Proliferation/drug effects , Cells, Cultured , Enterotoxins/genetics , Enterotoxins/metabolism , Enterotoxins/toxicity , Female , Gene Expression Regulation/drug effects , Humans , Interleukin-2/genetics , Interleukin-2/metabolism , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Lymphocyte Activation/drug effects , Male , Mutant Proteins/antagonists & inhibitors , Mutant Proteins/metabolism , Mutant Proteins/toxicity , Rabbits , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Recombinant Proteins/toxicity , Shock, Septic/etiology , Shock, Septic/immunology , Shock, Septic/metabolism , Superantigens/genetics , Superantigens/metabolism , Superantigens/toxicity , Survival Analysis , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Toxicokinetics , Tumor Necrosis Factor-alpha/metabolism
8.
Toxins (Basel) ; 2(9): 2272-88, 2010 09.
Article En | MEDLINE | ID: mdl-22069685

Staphylococcal superantigens (sAgs), such as toxic shock syndrome toxin 1 (TSST-1), induce massive cytokine production, which may result in toxic shock syndrome (TSS) and sepsis. Recently, we reported that in vitro studies in human peripheral blood mononuclear cells (PBMC) do not reflect the immunological situation of the host, because after exposure to superantigens (sAgs) in vivo, mononuclear cells (MNC) leave the circulation and migrate to organs, e.g., the spleen, liver and lung. Our experimental model of choice is the rabbit because it is comparable to humans in its sensitivity to sAg. T cell activation has been assessed by lymphocyte proliferation and IL-2 gene expression after in vivo challenge with TSST-1 and the mutant antigens; expression of the genes of proinflammatory cytokines were taken as indicators for the inflammatory reaction after the combined treatment with TSST-1 and LPS. The question as to whether the biological activities of TSST-1, e.g., lymphocyte extravasation, toxicity and increased sensitivity to LPS, are mediated by T cell activation or activation by MHC II-only, are unresolved and results are contradictory. We have addressed this question by studying these reactions in vivo, with two TSST-1 mutants: one mutated at the MHC binding site (G31R) with reduced MHC binding with residual activity still present, and the other at the T cell binding site (H135A) with no residual function detectable. Here, we report that the mutant G31R induced all the biological effects of the wild type sAg, while the mutant with non-functional TCR binding did not retain any of the toxic effects, proving the pivotal role of T cells in this system.


Bacterial Toxins/pharmacology , Cytokines/metabolism , Enterotoxins/pharmacology , Lymphocyte Activation/drug effects , Mutant Proteins/pharmacology , Mutation , Superantigens/pharmacology , T-Lymphocytes/drug effects , Animals , Bacterial Toxins/genetics , Bacterial Toxins/immunology , Binding Sites , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Proliferation/drug effects , Disease Models, Animal , Enterotoxins/genetics , Enterotoxins/immunology , Female , Gene Expression/drug effects , Host-Pathogen Interactions , Lymphocyte Activation/immunology , Mutant Proteins/genetics , Mutant Proteins/immunology , Protein Binding , Rabbits , Superantigens/genetics , Superantigens/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
...