Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 17 de 17
1.
Vaccine X ; 18: 100494, 2024 Jun.
Article En | MEDLINE | ID: mdl-38746060

Background: Previous phase 3 studies showed that the AS01E-adjuvanted respiratory syncytial virus (RSV) prefusion F protein-based vaccine for older adults (RSVPreF3 OA) is well tolerated and efficacious in preventing RSV-associated lower respiratory tract disease in adults ≥ 60 years of age. This study evaluated lot-to-lot immunogenicity consistency, reactogenicity, and safety of three RSVPreF3 OA lots. Methods: This phase 3, multicenter, double-blind study randomized (1:1:1) participants ≥ 60 years of age to receive one of three RSVPreF3 OA lots. Serum RSVPreF3-binding immunoglobulin G (IgG) concentration was assessed at baseline and 30 days post-vaccination. Lot-to-lot consistency was demonstrated if the two-sided 95 % confidence intervals (CIs) of the RSVPreF3-binding IgG geometric mean concentration (GMC) ratios between each lot pair at 30 days post-vaccination were within 0.67 and 1.50. Solicited adverse events (AEs) within four days, unsolicited AEs within 30 days, and serious AEs (SAEs) and potential immune-mediated diseases within six months post-vaccination were recorded. Results: A total of 757 participants received RSVPreF3 OA, of whom 708 were included in the per-protocol set (234, 237, and 237 participants for each lot). Lot-to-lot consistency was demonstrated: GMC ratios were 1.06 (95 % CI: 0.94-1.21), 0.92 (0.81-1.04), and 0.87 (0.77-0.99) between the lot pairs (lot 1/2; 1/3; 2/3). For the three lots, the RSVPreF3-binding IgG concentration increased 11.84-, 11.29-, and 12.46-fold post-vaccination compared to baseline. The reporting rates of solicited and unsolicited AEs, SAEs, and potential immune-mediated diseases were balanced between lots. Twenty-one participants reported SAEs; one of these-a case of atrial fibrillation-was considered by the investigator as vaccine-related. SAEs with a fatal outcome were reported for four participants, none of which were considered by the investigator as vaccine-related. Conclusion: This study demonstrated lot-to-lot immunogenicity consistency of three RSVPreF3 OA vaccine lots and indicated that the vaccine had an acceptable safety profile.ClinicalTrials.gov: NCT05059301.

2.
Clin Infect Dis ; 2024 Jan 08.
Article En | MEDLINE | ID: mdl-38189778

BACKGROUND: Co-administration of vaccines against respiratory syncytial virus (RSV) and influenza can be considered given their overlapping seasonality, and may increase vaccine uptake and compliance. In this phase 3, open-label, randomized study, we evaluated the immunogenicity, reactogenicity, and safety of the AS01E-adjuvanted RSV prefusion F protein-based candidate vaccine (RSVPreF3 OA) when co-administered with a seasonal quadrivalent influenza vaccine (FLU-QIV) in older adults. METHODS: Participants aged ≥60 years (randomized 1:1) received either RSVPreF3 OA and FLU-QIV simultaneously on day 1 (Co-Ad group) or FLU-QIV on day 1 followed by RSVPreF3 OA on day 31 (sequential administration [SA] group). The co-primary objectives were to demonstrate noninferiority of RSVPreF3 OA in terms of RSV-A neutralization geometric mean titer (GMT) ratio and FLU-QIV in terms of hemagglutination inhibition GMT ratio for each FLU-QIV strain, when co-administered versus when administered alone at 1-month post-vaccination. Noninferiority was demonstrated if the upper limit of the 95% confidence interview of the group GMT ratio (SA/Co-Ad) was ≤1.5. Secondary descriptive objectives comprised additional immunogenicity assessments, reactogenicity, and safety. RESULTS: Of the 885 participants who received one dose of the study vaccines, 837 were included in the per protocol set. Demographic and baseline characteristics were balanced between the groups. Both co-primary objectives were met for both vaccines. Reported adverse events in both groups were mild-to-moderate, with a low frequency of grade 3 events. CONCLUSIONS: Data from this study demonstrate that RSVPreF3 OA can be co-administered with FLU-QIV. Co-administration is well tolerated, with an acceptable safety profile. CLINICALTRIALS.GOV REGISTRATION: NCT04841577.

3.
Clin Infect Dis ; 2024 Jan 22.
Article En | MEDLINE | ID: mdl-38253338

BACKGROUND: The adjuvanted RSV prefusion F protein-based vaccine (RSVPreF3 OA) was efficacious against RSV-related lower respiratory tract disease (RSV-LRTD) in ≥60-year-olds over 1 RSV season. We evaluated efficacy and safety of 1 RSVPreF3 OA dose and of 2 RSVPreF3 OA doses given 1 year apart against RSV-LRTD over 2 RSV seasons post-dose 1. METHODS: In this phase 3, blinded trial, ≥60-year-olds were randomized (1:1) to receive RSVPreF3 OA or placebo pre-season 1. RSVPreF3 OA recipients were re-randomized (1:1) to receive a second RSVPreF3 OA dose (RSV_revaccination group) or placebo (RSV_1dose group) pre-season 2; participants who received placebo pre-season 1 received placebo pre-season 2 (placebo group). Efficacy of both vaccine regimens against RSV-LRTD was evaluated over 2 seasons combined (confirmatory secondary objective, success criterion: lower limits of 2-sided confidence intervals [CIs] around efficacy estimates >20%). RESULTS: The efficacy analysis comprised 24,967 participants (RSV_1dose: 6227, RSV_revaccination: 6242, placebo: 12,498). Median efficacy follow-up was 17.8 months. Efficacy over 2 seasons of 1 RSVPreF3 OA dose was 67.2% (97.5% CI: 48.2-80.0) against RSV-LRTD and 78.8% (95% CI: 52.6-92.0) against severe RSV-LRTD. Efficacy over 2 seasons of a first dose followed by revaccination was 67.1% (97.5% CI: 48.1-80.0) against RSV-LRTD and 78.8% (95% CI: 52.5-92.0) against severe RSV-LRTD. Reactogenicity/safety of the revaccination dose were similar to dose 1. CONCLUSION: One RSVPreF3 OA dose was efficacious against RSV-LRTD over 2 RSV seasons in ≥60-year-olds. Revaccination 1 year post-dose 1 was well tolerated but did not seem to provide additional efficacy benefit in the overall study population. ClinicalTrials.gov registration: NCT04886596.

4.
J Infect Dis ; 2023 Dec 14.
Article En | MEDLINE | ID: mdl-38099559

BACKGROUND: The recently approved AS01E-adjuvanted respiratory syncytial virus (RSV) prefusion F protein-based vaccine (RSVPreF3 OA) demonstrated high efficacy against RSV-related disease in ≥60-year-olds. METHODS: This ongoing phase 3 study in ≥60-year-olds evaluates the immune persistence until three years post-RSVPreF3 OA vaccination. Here, we describe interim results on humoral and cell-mediated immunogenicity, reactogenicity, and safety until one year post-dose 1. RESULTS: In total, 1653 participants were vaccinated. One month post-dose 1, neutralization titers increased 10.5-fold (RSV-A) and 7.8-fold (RSV-B) versus pre-dose 1. Titers then declined to levels 4.4-fold (RSV-A) and 3.5-fold (RSV-B) above pre-dose 1 at month 6, and remained 3.1-fold (RSV-A) and 2.3-fold (RSV-B) above pre-dose 1 levels after one year. RSVPreF3-binding immunoglobulin G levels and CD4+ T-cell frequencies showed similar kinetics. Solicited administration-site and systemic adverse events (mostly mild to moderate and transient) were reported by 62.2% and 49.5% of participants. Serious adverse events were reported by 3.9% of participants within 6 months post-dose 1; one case was considered vaccine-related. CONCLUSIONS: One RSVPreF3 OA dose elicited cell-mediated and RSV-A and RSV-B-specific humoral immune responses that declined over time but remained above pre-dose 1 levels for at least one year. The vaccine was well tolerated with an acceptable safety profile.Clinicaltrials.gov NCT04732871.


Respiratory syncytial virus (RSV) is a major cause of illness and hospitalization in older adults. An RSV vaccine for older adults developed by GSK was recently approved. The vaccine was well tolerated and provided protection against RSV disease in adults aged 60 years and older during at least one RSV season. In this ongoing study, we are evaluating the magnitude and durability of the immune response, as well as vaccine safety, until three years after vaccination of adults aged 60 years and older from five countries. Here, we report the results of an interim analysis until one year after vaccination with one dose. In total, 1653 participants were vaccinated. We found that the vaccine induced a strong immune response that was evident one month after vaccination, after which it declined, but persisted for at least one year. Study participants most often reported pain at the injection site, muscle pain, tiredness, and headache as adverse reactions, which were mostly mild to moderate and of short duration. One serious adverse reaction was considered related to the vaccine. The long-term immune response that was observed in this study is consistent with the vaccine providing protection during at least one RSV season.

5.
BMJ Open ; 11(7): e042921, 2021 07 07.
Article En | MEDLINE | ID: mdl-34233965

INTRODUCTION: Septic shock is the subgroup of patients with sepsis, which presents as vasopressor dependence, an elevated blood lactate concentration and is associated with a mortality of at least 30%. Expression of the triggering receptor expressed on myeloid cells 1 (TREM-1) pathway, measured using a serum biomarker of pathway activation (soluble TREM-1, sTREM-1) has been associated with outcome in septic shock. Preclinical and early phase patient data suggest that therapeutic modulation of this pathway may improve survival. METHODS AND ANALYSIS: Efficacy, Safety and Tolerability of Nangibotide in Patients with Septic Shock is a phase IIb randomised controlled trial that will take place in up to 50 centres in seven countries and recruit 450 patients with septic shock to receive either placebo or one of two doses of nangibotide, a novel regulator of the TREM-1 pathway. The primary outcome will be the impact of nangibotide therapy on the change in Sequential Organ Failure Assessment score from a baseline determined before initiation of study drug therapy. This will be assessed first in the patients with an elevated sTREM-1 level and then in the study population as a whole. In addition to safety, secondary outcomes of the study will include efficacy of nangibotide in relation to sTREM-1 levels in terms of organ function, mortality and long-term morbidity. This study will also facilitate the development of a novel platform for the measurement of sTREM-1 at the point of care. ETHICS AND DISSEMINATION: The study has been approved by the responsible ethics committees/institutional review boards in all study countries: Belgium: Universitair Ziekenhuis Antwerpen, France: CPP Ile de France II, Denmark: Region Hovedstaden, Spain: ethics committee from Valld'Hebron Hospital, Barcelona, Finland: Tukija, Ireland: St. James' Hospital (SJH) / Tallaght University Hospital (TUH) Joint Research Ethics Committee, USA: Lifespan, Providence TRIAL REGISTRATION NUMBERS: EudraCT Number: 2018-004827-36 and NCT04055909.


Shock, Septic , Belgium , Finland , France , Humans , Ireland , Randomized Controlled Trials as Topic , Shock, Septic/drug therapy , Spain , Treatment Outcome
6.
Intensive Care Med ; 46(7): 1425-1437, 2020 Jul.
Article En | MEDLINE | ID: mdl-32468087

PURPOSE: Nangibotide is a specific TREM-1 inhibitor that tempered deleterious host-pathogens interactions, restored vascular function, and improved survival, in animal septic shock models. This study evaluated the safety and pharmacokinetics of nangibotide and its effects on clinical and pharmacodynamic parameters in septic shock patients. METHODS: This was a multicenter randomized, double-blind, two-stage study. Patients received either continuous infusion of nangibotide (0.3, 1.0, or 3.0 mg/kg/h) or placebo. Treatment began < 24 h after shock onset and continued for up to 5 days. Safety primary outcomes were adverse events (AEs), whether serious or not, and death. Exploratory endpoints evaluated nangibotide effects on pharmacodynamics, organ function, and mortality, and were analyzed according to baseline sTREM-1 concentrations. RESULTS: Forty-nine patients were randomized. All treatment emergent AEs (TEAEs) were collected until Day 28. No significant differences were observed in TEAEs between treatment groups. No drug withdrawal linked to TEAE nor appearance of anti-drug antibodies were reported. Nangibotide pharmacokinetics appeared to be dose-proportional and clearance was dose-independent. Nangibotide did not significantly affect pharmacodynamic markers. Decrease in SOFA score LS mean change (± SE) from baseline to Day 5 in pooled nangibotide groups versus placebo was - 0.7 (± 0.85) in the randomized population and - 1.5 (± 1.12) in patients with high baseline plasma sTREM-1 concentrations (non-significant). This pattern was similar to organ support end points. CONCLUSION: No significant increases in TEAEs were detected in nangibotide-treated patients versus placebo. These results encourage further evaluation of nangibotide and further exploration of plasma sTREM-1 concentrations as a predictive efficacy biomarker.


Shock, Septic , Animals , Double-Blind Method , Humans , Immunologic Factors , Shock, Septic/drug therapy , Treatment Outcome
7.
Br J Clin Pharmacol ; 84(10): 2270-2279, 2018 10.
Article En | MEDLINE | ID: mdl-29885068

AIMS: The peptide nangibotide is the first clinical-stage agent targeting the immunoreceptor TREM-1 (triggering receptor expressed on myeloid cells-1) and is being investigated as a novel therapy for acute inflammatory disorders such as septic shock. This first-in-man, randomized, double-blind, ascending dose, placebo-controlled Phase I study evaluated the safety, tolerability and pharmacokinetics of nangibotide. METHODS: Twenty-seven healthy subjects (aged 18-45 years) were randomized into eight groups. Nangibotide was administered as a single continuous intravenous infusion. The first two groups received a single i.v. dose of 1 and 10 mg, respectively, over 15 min. Subsequent groups were randomized in a product : placebo ratio of 3:1 at doses ranging from 0.03 to 6 mg kg-1  h-1 over 7 h 45 min, preceded by a 15-minute loading dose of up to 5 mg kg-1 . RESULTS: Nangibotide was safe and well tolerated up to the highest dose tested. There were only few adverse events and they were mild in severity and considered unrelated to treatment. Nangibotide displayed dose-proportional PK properties, with a clearance of 6.6 l kg-1  h-1 for a subject of 70 kg and a 3 min effective half-life, which are compatible with extensive enzymatic metabolism in blood. Central and peripheral volumes of distribution were 16.7 l and 15.9 l respectively, indicating limited distribution of the drug mainly in blood and interstitial fluid. No circulating anti-drug antibodies were detectable up to 28 days after administration. CONCLUSIONS: The novel immunomodulator nangibotide displayed favourable safety and PK profiles at all doses, including expected pharmacologically active doses, and warrants further clinical development.


Drugs, Investigational/pharmacokinetics , Immunologic Factors/pharmacokinetics , Peptides/pharmacokinetics , Triggering Receptor Expressed on Myeloid Cells-1/antagonists & inhibitors , Adult , Area Under Curve , Dose-Response Relationship, Drug , Double-Blind Method , Drugs, Investigational/administration & dosage , Drugs, Investigational/adverse effects , Half-Life , Healthy Volunteers , Humans , Immunity, Innate/drug effects , Immunologic Factors/administration & dosage , Immunologic Factors/adverse effects , Infusions, Intravenous , Male , Metabolic Clearance Rate , Peptides/administration & dosage , Peptides/adverse effects , Young Adult
8.
Eur J Immunol ; 41(8): 2303-13, 2011 Aug.
Article En | MEDLINE | ID: mdl-21538349

TLR agonists have been suggested to directly impact Tregs, thereby enhancing or reversing their suppressive function. Here, in order to select TLR agonists leading to potent effector T-cell responses, while minimizing Treg inhibitory function, we used a model antigen, covalently linked to an inert delivery system, combined with a large panel of TLR agonists, for the immunization of mice with an attenuated/depleted or intact Treg subset. We observed that the negative modulation of effector CD4(+) T cells exerted by Tregs cannot be circumvented, whatever the TLR agonist used as adjuvant. To better understand the impact of TLR agonists on Tregs, we investigated (i) the TLR expression profile of highly purified CD4(+) Foxp3(+) Tregs, at steady state or subsequent to in vivo activation by TLR agonists and (ii) the Treg phenotype after in vivo and in vitro activation by TLR agonists. Our results demonstrate that TLR agonists, as single signal inducers, are not able to directly activate Tregs. The phenotypic Treg activation observed in vivo, following TLR administration, does not result from cross-talk with conventional T cells but is rather a consequence of the interaction with other immune cell type(s).


Adjuvants, Immunologic/pharmacology , CD4-Positive T-Lymphocytes/drug effects , T-Lymphocytes, Regulatory/drug effects , Toll-Like Receptors/agonists , Amino Acid Sequence , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Female , Flow Cytometry , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Profiling , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Imidazoles/pharmacology , Lipopolysaccharides/pharmacology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , Oligodeoxyribonucleotides/pharmacology , Ovalbumin/administration & dosage , Ovalbumin/immunology , Peptide Fragments/administration & dosage , Peptide Fragments/immunology , Poly I-C/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Toll-Like Receptors/genetics , Toll-Like Receptors/immunology , Zymosan/pharmacology
9.
PLoS One ; 6(4): e18891, 2011 Apr 27.
Article En | MEDLINE | ID: mdl-21556142

BACKGROUND: The Plasmodium falciparum pre-erythrocytic stage candidate vaccine RTS,S is being developed for protection of young children against malaria in sub-Saharan Africa. RTS,S formulated with the liposome based adjuvant AS01(E) or the oil-in-water based adjuvant AS02(D) induces P. falciparum circumsporozoite (CSP) antigen-specific antibody and T cell responses which have been associated with protection in the experimental malaria challenge model in adults. METHODS: This study was designed to evaluate the safety and immunogenicity induced over a 19 month period by three vaccination schedules (0,1-, 0,1,2- and 0,1,7-month) of RTS,S/AS01(E) and RTS,S/AS02(D) in children aged 5-17 months in two research centers in Ghana. Control Rabies vaccine using the 0,1,2-month schedule was used in one of two study sites. RESULTS: Whole blood antigen stimulation followed by intra-cellular cytokine staining showed RTS,S/AS01(E) induced CSP specific CD4 T cells producing IL-2, TNF-α, and IFN-γ. Higher T cell responses were induced by a 0,1,7-month immunization schedule as compared with a 0,1- or 0,1,2-month schedule. RTS,S/AS01(E) induced higher CD4 T cell responses as compared to RTS,S/AS02(D) when given on a 0,1,7-month schedule. CONCLUSIONS: These findings support further Phase III evaluation of RTS,S/AS01(E). The role of immune effectors and immunization schedules on vaccine protection are currently under evaluation. TRIAL REGISTRATION: ClinicalTrials.gov NCT00360230.


Malaria Vaccines/immunology , T-Lymphocytes/immunology , Antibodies, Protozoan/biosynthesis , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Child , Child, Preschool , Cytokines/metabolism , Flow Cytometry , Ghana , Humans , Infant , Malaria Vaccines/administration & dosage
10.
PLoS One ; 6(4): e18559, 2011 Apr 11.
Article En | MEDLINE | ID: mdl-21494604

UNLABELLED: The recombinant circumsporozoite protein (CS) based vaccine, RTS,S, confers protection against Plasmodium falciparum infection in controlled challenge trials and in field studies. The RTS,S recombinant antigen has been formulated with two adjuvant systems, AS01 and AS02, which have both been shown to induce strong specific antibody responses and CD4 T cell responses in adults. As infants and young children are particularly susceptible to malaria infection and constitute the main target population for a malaria vaccine, we have evaluated the induction of adaptive immune responses in young children living in malaria endemic regions following vaccination with RTS,S/AS01(E) and RTS,S/AS02(D). Our data show that a CS-specific memory B cell response is induced one month after the second and third vaccine dose and that CS-specific antibodies and memory B cells persist up to 12 months after the last vaccine injection. Both formulations also induced low but significant amounts of CS-specific IL-2(+) CD4(+) T cells one month after the second and third vaccine dose, upon short-term in vitro stimulation of whole blood cells with peptides covering the entire CS derived sequence in RTS,S. These results provide evidence that both RTS,S/AS01(E) and RTS,S/AS02(D) induced adaptive immune responses including antibodies, circulating memory B cells and CD4(+) T cells directed against P. falciparum CS protein. TRIAL REGISTRATION: ClinicalTrials.gov NCT00307021.


B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Malaria Vaccines/immunology , Plasmodium falciparum/immunology , Vaccination , Antibodies, Protozoan/immunology , Antibody Formation/immunology , Antibody Specificity/immunology , Child , Cytokines/blood , Gabon , Hepatitis B Vaccines/immunology , Humans , Immunoglobulin G/biosynthesis , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Protozoan Proteins/immunology , Species Specificity , Titrimetry
11.
PLoS One ; 5(11): e14090, 2010 Nov 29.
Article En | MEDLINE | ID: mdl-21124768

The malaria vaccine candidate, RTS,S/AS01(E), showed promising protective efficacy in a trial of Kenyan and Tanzanian children aged 5 to 17 months. Here we report on the vaccine's safety and tolerability. The experimental design was a Phase 2b, two-centre, double-blind (observer- and participant-blind), randomised (1∶1 ratio) controlled trial. Three doses of study or control (rabies) vaccines were administered intramuscularly at 1 month intervals. Solicited adverse events (AEs) were collected for 7 days after each vaccination. There was surveillance and reporting for unsolicited adverse events for 30 days after each vaccination. Serious adverse events (SAEs) were recorded throughout the study period which lasted for 14 months after dose 1 in Korogwe, Tanzania and an average of 18 months post-dose 1 in Kilifi, Kenya. Blood samples for safety monitoring of haematological, renal and hepatic functions were taken at baseline, 3, 10 and 14 months after dose 1. A total of 894 children received RTS,S/AS01(E) or rabies vaccine between March and August 2007. Overall, children vaccinated with RTS,S/AS01(E) had fewer SAEs (51/447) than children in the control group (88/447). One SAE episode in a RTS,S/AS01(E) recipient and nine episodes among eight rabies vaccine recipients met the criteria for severe malaria. Unsolicited AEs were reported in 78% of subjects in the RTS,S/AS01(E) group and 74% of subjects in the rabies vaccine group. In both vaccine groups, gastroenteritis and pneumonia were the most frequently reported unsolicited AE. Fever was the most frequently observed solicited AE and was recorded after 11% of RTS,S/AS01(E) doses compared to 31% of doses of rabies vaccine. The candidate vaccine RTS,S/AS01(E) showed an acceptable safety profile in children living in a malaria-endemic area in East Africa. More data on the safety of RTS,S/AS01(E) will become available from the Phase 3 programme.


Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Plasmodium falciparum/immunology , Vaccination/methods , Alanine Transaminase/blood , Creatinine/blood , Double-Blind Method , Fever/etiology , Gastroenteritis/etiology , Humans , Infant , Kenya , Malaria Vaccines/administration & dosage , Malaria Vaccines/adverse effects , Malaria, Falciparum/prevention & control , Pain/etiology , Pneumonia/etiology , Rabies Vaccines/administration & dosage , Rabies Vaccines/adverse effects , Rabies Vaccines/immunology , Sleep Stages/drug effects , Tanzania , Vaccination/adverse effects
12.
J Infect Dis ; 200(9): 1367-70, 2009 Nov 01.
Article En | MEDLINE | ID: mdl-19817587

In Staphylococcus aureus, rsbU down-regulates agr and stimulates production of staphyloxanthin (STX), an antioxidant that may contribute to intracellular survival after phagocytosis. Using isogenic rsbU(-) and rsbU(+) strains, we show that rsbU causes increased internalization and intracellular growth in both THP-1 macrophages and human umbilical vein endothelial cells (more so for the latter) without change in subcellular localization and that inhibition of STX biosynthesis markedly reduces intracellular growth of the rsbU(+) strain (and of clinical isolates, including USA300; tested with macrophages only) without affecting internalization. Thus, rsbU is important for uptake and for STX biosynthesis and is critical for intracellular multiplication of S. aureus.


Bacterial Proteins/metabolism , Endothelial Cells/microbiology , Macrophages/microbiology , Phagocytosis , Phosphoric Monoester Hydrolases/metabolism , Staphylococcus aureus/growth & development , Xanthophylls/biosynthesis , Cell Line , Humans , Staphylococcus aureus/metabolism , Umbilical Veins/cytology
13.
Antimicrob Agents Chemother ; 52(8): 2797-805, 2008 Aug.
Article En | MEDLINE | ID: mdl-18519727

Staphylococcus aureus invades eukaryotic cells. When methicillin-resistant S. aureus (MRSA) ATCC 33591 is phagocytized by human THP-1 macrophages, complete restoration of susceptibility to cloxacillin and meropenem is shown and the strain becomes indistinguishable from MSSA ATCC 25923 due to the acid pH prevailing in phagolysosomes (S. Lemaire et al., Antimicrob. Agents Chemother. 51:1627-1632, 2007). We examined whether this observation can be extended to (i) strains of current clinical and epidemiological interest (three hospital-acquired MRSA [HA-MRSA] strains, two community-acquired MRSA [CA-MRSA] strains, two HA-MRSA strains with the vancomycin-intermediate phenotype, one HA-MRSA strain with the vancomycin-resistant phenotype, and one animal [porcine] MRSA strain), (ii) activated THP-1 cells and nonprofessional phagocytes (keratinocytes, Calu-3 bronchial epithelial cells), and (iii) other beta-lactams (imipenem, oxacillin, cefuroxime, cefepime). All strains showed (i) a marked reduction in MICs in broth at pH 5.5 compared with the MIC at pH 7.4 and (ii) sigmoidal dose-response curves with cloxacillin (0.01x to 100x MIC, 24 h of incubation) after phagocytosis by THP-1 macrophages that were indistinguishable from each other and from the dose-response curve for methicillin-susceptible S. aureus (MSSA) ATCC 25923 (relative potency [50% effect], 6.09x MIC [95% confidence interval {CI}, 4.50 to 8.25]; relative efficacy [change in bacterial counts over the original inoculum for an infinitely large cloxacillin concentration, or maximal effect], -0.69 log CFU [95% CI, -0.79 to -0.58]). Similar dose-response curves for cloxacillin were also observed with MSSA ATCC 25923 and MRSA ATCC 33591 after phagocytosis by activated THP-1 macrophages, keratinocytes, and Calu-3 cells. By contrast, there was a lower level of restoration of susceptibility of MRSA ATCC 33591 to cefuroxime and cefepime after phagocytosis by THP-1 macrophages, even when the data were normalized for differences in MICs. We conclude that the restoration of MRSA susceptibility to beta-lactams after phagocytosis is independent of the strain and the types of cells but varies between beta-lactams.


Anti-Bacterial Agents/pharmacology , Methicillin Resistance/drug effects , Staphylococcus aureus/drug effects , beta-Lactams/pharmacology , Carbapenems/pharmacology , Cefepime , Cefuroxime/pharmacology , Cell Line , Cells, Cultured , Cephalosporins/pharmacology , Cloxacillin/pharmacology , Humans , Hydrogen-Ion Concentration , Imipenem/pharmacology , Meropenem , Methicillin/pharmacology , Microbial Sensitivity Tests , Oxacillin/pharmacology , Phagocytosis/drug effects , Thienamycins/pharmacology
14.
Blood ; 109(12): 5318-26, 2007 Jun 15.
Article En | MEDLINE | ID: mdl-17339421

We investigated whether Toll-like receptor ligands (TLR-Ls) can bypass the requirement for CD4(+) T-cell help in the induction of fully efficient memory CD8(+) T cells (cytotoxic T lymphocytes [CTLs]). "Helpless" CTLs were induced by a synthetic CD8(+) T-cell epitope administered with TLR3-L and TLR9-L, but not with TLR2/6-L, TLR4-L, or TLR7-L. The up-regulation of MHC-I and costimulatory molecules by dendritic cells following TLR stimulation was not sufficient for the priming of "helpless" CTLs, which depended essentially on the induction of a strong IFN-alpha/beta response. The "helpless" CTLs induced by TLR-Ls differentiated into fully functional memory CTLs able to proliferate as well as their "helped" counterparts upon challenge, in the absence of CD4(+) T-cell help.


CD8-Positive T-Lymphocytes/physiology , Immunologic Memory , Toll-Like Receptor 3/physiology , Animals , CD4-Positive T-Lymphocytes , Cell Proliferation , Cross-Priming , Epitopes, T-Lymphocyte/administration & dosage , Epitopes, T-Lymphocyte/pharmacology , Ligands , Mice , Mice, Inbred C57BL , T-Lymphocytes, Cytotoxic , Toll-Like Receptor 3/immunology , Up-Regulation/drug effects
15.
Mol Ther ; 13(4): 729-37, 2006 Apr.
Article En | MEDLINE | ID: mdl-16360341

The Wiskott Aldrich syndrome protein (WASP) is a hematopoietic-specific cytoskeletal regulator that is necessary for induction of normal immunity. In the context of effective gene therapy for WAS, cellular models of human WASP deficiency are important for definition of the threshold of protein expression required for optimal activity. Using lentiviral vector-mediated RNA interference (RNAi), we were able to down-regulate the levels of human WASP in cell lines and primary cells. In dendritic cells (DC), RNAi-induced WASP deficiency did not impair phenotypic maturation but perturbed cytoskeletal organization. As a result, podosomes, which are actin-rich structures present in immature adherent DC, were formed less efficiently and motility was disturbed. Overall, treatment of cells with RNAi recapitulated the phenotype of cells derived from patients or animals with inactivating mutations of the WAS gene. Interestingly, reduction of the levels of WASP to about 60% of normal was sufficient to inhibit the formation of podosomes in DC, implying that this cell type requires near-normal levels of WASP to sustain physiological cytoskeleton-dependent activities.


Cell Movement/physiology , Dendritic Cells/metabolism , Down-Regulation , RNA, Small Interfering/pharmacology , Wiskott-Aldrich Syndrome Protein/physiology , Cell Movement/drug effects , Cells, Cultured , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Dose-Response Relationship, Drug , Gene Expression , Genetic Therapy , Genetic Vectors/therapeutic use , Humans , Jurkat Cells , Lentivirus/genetics , Mutation , RNA, Small Interfering/metabolism , Transduction, Genetic , Wiskott-Aldrich Syndrome/genetics , Wiskott-Aldrich Syndrome Protein/deficiency , Wiskott-Aldrich Syndrome Protein/genetics , Wiskott-Aldrich Syndrome Protein/metabolism
16.
Blood ; 107(7): 2694-701, 2006 Apr 01.
Article En | MEDLINE | ID: mdl-16357328

Plasmacytoid dendritic cells (pDCs) play an important role in innate and adaptive immunity, prompting interest in mechanisms controlling the production of this lineage of cells. Notch signaling via one of the Notch ligands, delta-like 1 (delta-1), influences the hematopoietic development of several lymphoid and myeloid lineages, but whether or not delta-1 affects the formation of pDCs is unknown and was tested here. Human CD34+ progenitor cells were cultured onto delta-1-expressing OP9 stroma in the presence of flt-3 ligand and IL-7, and this efficiently generated BDCA-2+ CD123+ CD4+ CD11c- cells with the characteristic morphology of pDCs, expressing toll-like receptor-9 (TLR9), pre-Talpha mRNAs, and secreting CpG-induced IFN-alpha. Delta-1 augmented the numbers of BDCA-2+ cells produced without affecting their proliferation, and the effect was blocked by gamma-secretase inhibition. The development of pDCs was stroma-, delta-1-, and cytokine-dependent and could be induced from committed lymphoid progenitor cells, which responded to delta-1 by opposite changes in pDC- and B-cell production. Our results identify delta-1 as a novel factor enhancing pDC hematopoiesis and delineate a new role for Notch signaling in lymphopoiesis by showing its opposite effect on pDC and B lineage determination.


Dendritic Cells/physiology , Hematopoiesis/physiology , Receptors, Notch/physiology , Antigens, CD/immunology , Antigens, CD34/immunology , Cell Line , DNA Primers , Dendritic Cells/cytology , Dendritic Cells/immunology , Fetal Blood/cytology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Humans , Interferon-alpha/genetics , Interleukin-12/genetics , Interleukin-8/genetics , Lectins, C-Type/genetics , Membrane Glycoproteins/genetics , Polymerase Chain Reaction , Receptors, Immunologic/genetics , Reverse Transcriptase Polymerase Chain Reaction , Stromal Cells/cytology , Stromal Cells/physiology , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/immunology
17.
J Exp Biol ; 208(Pt 3): 461-8, 2005 Feb.
Article En | MEDLINE | ID: mdl-15671334

Even though variation in morphology is known to translate into variation in performance, studies looking at structural variability of a sensor to predict its consequences on the performance of animals are exceedingly rare. We investigated the morphological variability of air-flow-sensing receptors in wild populations of wood crickets (Nemobius sylvestris) sampled in a wide variety of habitats differing in latitude, litter structure, vegetation and predator communities. These hair receptors act as predator sensors. The observed levels of hair morphological variation were then incorporated into a biomechanical model of the hair canopy response to air flow to predict their influence on cricket predator perception. Cricket populations differ from each other, often strongly so, in the total number of hairs and in the number of hairs longer than 1 mm, which are the hairs most sensitive for the perception of approaching predators. The hair canopy response, the output of the biomechanical model, sums up over the entire canopy the angles of deflection at which a neurophysiological response is triggered and represents the sensitivity of the cercal system. It is 35% higher in the most sensitive population, compared with the least sensitive population. These large differences in perception sensitivity for a given predator signal translate into larger distances at which predators could be perceived. Thus, differences in morphology at the sensor level seem to be translated both at the perception level and subsequently at the performance level of crickets.


Behavior, Animal/physiology , Gryllidae/anatomy & histology , Air Movements , Animals , Biomechanical Phenomena , Gryllidae/physiology , Models, Biological , Sense Organs/anatomy & histology , Sense Organs/physiology
...