12 research outputs found

    Immunological non-inferiority of a new fully liquid presentation of the MenACWY-CRM vaccine to the licensed vaccine : results from a randomized, controlled, observer-blind study in adolescents and young adult

    Get PDF
    A fully liquid MenACWY-CRM vaccine presentation has been developed, modifying the meningococcal serogroup A (MenA) component from lyophilized to liquid. The safety and immunogenicity of the liquid presentation at the end of the intended shelf-life (aged for 24 or 30 months) were compared to the licensed lyophilized/liquid presentation. This multicenter, randomized (1:1), observer-blind, phase 2b study (NCT03433482) enrolled adolescents and young adults (age 10-40 years). In part 1, 844 participants received one dose of liquid presentation stored for approximately 24 months or licensed presentation. In part 2, 846 participants received one dose of liquid presentation stored for approximately 30 months or licensed presentation. After storage, the MenA free saccharide (FS) level was approximately 25% and O-acetylation was approximately 45%. The primary objective was to demonstrate non-inferiority of the liquid presentation to licensed presentation, as measured by human serum bactericidal assay (hSBA) geometric mean titers (GMTs) against MenA, 1-month post-vaccination. Immune responses against each vaccine serogroup were similar between groups. Between-group ratios of hSBA GMTs for MenA were 1.21 (part 1) and 1.11 (part 2), with two-sided 95% confidence interval lower limits (0.94 and 0.87, respectively) greater than the prespecified non-inferiority margin (0.5), thus meeting the primary study objective. No safety concerns were identified. Despite reduced O-acetylation of MenA and increased FS content, serogroup-specific immune responses induced by the fully liquid presentation were similar to those induced by the licensed MenACWY-CRM vaccine, with non-inferior anti-MenA responses. The safety profiles of the vaccine presentations were similar.GlaxoSmithKline Biologicals SAhttps://www.tandfonline.com/journals/KHVIMedical Microbiolog

    A Heterologous MF59-Adjuvanted H5N1 Prepandemic Influenza Booster Vaccine Induces a Robust, Cross-Reactive Immune Response in Adults and the Elderlyâ–¿

    No full text
    Immunogenicity and safety of a booster dose of an MF59-adjuvanted H5N1 vaccine containing 7.5 μg A/turkey/Turkey/1/2005-like (clade 2.2) H5N1 hemagglutinin, given approximately 18 months after primary vaccination with a heterologous strain, were evaluated. The booster vaccine was well tolerated and induced a robust, cross-reactive immune response

    One-year immunogenicity kinetics and safety of a purified chick embryo cell rabies vaccine and an inactivated Vero cell-derived Japanese encephalitis vaccine administered concomitantly according to a new, 1-week, accelerated primary series

    Full text link
    BACKGROUND Conventional rabies pre-exposure prophylaxis (PrEP) and Japanese encephalitis (JE) primary series vaccination regimens each require up to 4 weeks to complete and, thus, may not be feasible for individuals who need these immunizations on short notice. This Phase 3b, randomized, controlled, observer-blind study evaluated the immunogenicity and safety of concomitant administration of a purified chick embryo cell culture rabies vaccine and an inactivated, adsorbed JE vaccine according to an accelerated (1 week) regimen when compared with the conventional regimens (4 weeks). This report describes the kinetics of immune responses up to 1 year after vaccination. METHODS A total of 661 healthy adults (18 to ≤65 years) were randomized into the following accelerated or conventional vaccine regimens: Rabies + JE-Conventional, Rabies + JE-Accelerated, Rabies-Conventional and JE-Conventional. Immunogenicity was assessed by virus neutralization tests. Safety and tolerability were also evaluated. RESULTS Irrespective of rabies vaccination regimen, ≥97% of subjects had adequate levels of rabies virus neutralizing antibody (RVNA) concentrations (≥0.5 IU/ml) up to Day 57, with percentages of subjects with RVNA concentrations ≥0.5 IU/ml at Day 366 ranging between 68% in the Rabies + JE-Accelerated group and 80% of subjects in the Rabies-Conventional group. The Rabies + JE-Accelerated group revealed high JE neutralizing antibody titers at all-time points. At Day 366, the percentage of subjects with antibody titers indicative of seroprotection (PRNT50 titers ≥1:10) remained high across JE vaccine groups (86-94%). CONCLUSIONS The accelerated PrEP rabies and JE vaccination regimens, once licensed, could represent a valid alternative in the short-term to currently recommended conventional regimens. The concomitant administration of these two vaccines does not compromise immune responses to any of the vaccine antigens particularly when aiming for short-term protection. Further evidence will clarify the need for and timing to administration of rabies vaccine booster doses in subjects primed with an accelerated PrEP regimen. (NCT01662440)

    Simultaneous identification of B lymphocytes specific for HA from different influenza strains in <i>ex vivo</i> PBMCs samples.

    No full text
    <p><b>A.</b> PBMCs from an anonymous blood donor were pre-incubated with subunits from either B/Brisbane/60/2008 or A/Panama/2007/1999 (H3N2), and then stained with anti-CD20 mAb, HSA conjugated with A488 and A647, with A647-rH3 (from A/Brisbane/10/2007) and A488-rH1 (from A/California/07/09), or with A647-rB/HA (from B/Brisbane/60/2008) and A488-rH1 (from A/California/07/09). The staining patterns observed in the CD20<sup>+</sup> B-cell gate are shown. <b>B.</b> PBMCs from 16 anonymous blood donors were pre-saturated with B/Brisbane/60/2008 and then stained with anti-CD20 mAb, A647-rH3 (from A/Brisbane/10/2007) and A488-rH1 (from A/California/07/09). The scatter plot depicts paired values of H1<sup>+</sup> (y-axis) and H3<sup>+</sup> (x-axis) B-cells. The insert box plot depicts the distribution of H1<sup>+</sup>, H3<sup>+</sup> and H1<sup>+</sup>H3<sup>+</sup> B-cells in the same 16 donors. Mean values are indicated by dotted lines.</p

    Identification of B lymphocytes specific for HA from A and B influenza strains in <i>ex vivo</i> PBMCs samples.

    No full text
    <p>PBMCs from different anonymous blood donors were pre-incubated with vaccine mono-bulk subunits from the B/Brisbane/60/2008 (B/HA pretreatment), or the H3N2 A/Panama/2007/1999 strain (A/HA pretreatment) and then stained HSA, rH3 (from A/Brisbane/10/2007), rH1 (from A/California/07/2009), or B/HA (from B/Brisbane/60/2008), as indicated. <b>A.</b> Staining pattern observed on CD20<sup>+</sup> cells in PBMCs stained with the different rHA bait. The rectangular gates identify brilliant HA+ B-cells; the dotted vertical lines mark the gates used to sort HA<sup>+</sup> B-cells for the ELISPOT assays. <b>B.</b> Expression of the CD27 memory marker on HA<sup>+</sup> and HA<sup>neg</sup> B cells identified based on the sorting gates. <b>C.</b> H3<sup>+</sup> (n = 15,234), H1<sup>+</sup> (n = 6482) and B/HA<sup>+</sup> (n = 26,803) B-cells identified in A were sorted, mixed with autologous CD20<sup>neg</sup> cells (in the ratio of 1∶20, 1∶100 and 1∶33) and activated with CpG and IL-2 for 5 days <i>in vitro</i>. Unsorted PBMCs and CD20<sup>neg</sup> cells mixed with HA<sup>neg</sup> B cells were also cultured in the same manner, as controls. After 5 days cultured cells were harvested and assayed by ELISPOT for the number of cells secreting IgG and IgG specific for mono-bulk subunits from the vaccine strain homologous to the sorting bait. <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0070620#s2" target="_blank">Results</a> are expressed as numbers of antibody secreting cells (ASC) normalized to 10<sup>6</sup> cultured cells assayed by ELISPOT. Nd indicates undetectable ASC.</p

    Molecular cloning of HA<sup>+</sup> B lymphocytes.

    No full text
    <p>PBMCs from 4 anonymous blood bank donors were stained as in <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0070620#pone-0070620-g004" target="_blank">Figure 4B</a>. Single H1<sup>+</sup>, H3<sup>+</sup>, or H1<sup>neg</sup>H3<sup>neg</sup> CD20<sup>+</sup> B-cells were sorted to perform molecular cloning and analysis of their paired V<sub>H</sub>V<sub>L</sub> Ig regions as described in Material and Methods section. <b>A–E.</b> Distribution of V<sub>H</sub> (A), D<sub>H</sub> (B), J<sub>H</sub> (C), V<sub>k</sub> (D) and J<sub>k</sub> (E) gene use across arrays of B-cells sorted from each donor (16 and 18 HA<sup>+</sup> clones from donors #1 and #2; 35 HA<sup>+</sup> and 20 HA<sup>neg</sup> clones from donor 3; 16 HA<sup>+</sup> and 16 HA<sup>neg</sup> clones from donor #4. <b>F–I.</b> Number of mutations in H1<sup>+</sup> and H1<sup>neg</sup> CD20<sup>+</sup> B-cells from donors #3 and #4, which cause dissimilar (F, H) or similar (G, I) amino acid substitutions in V<sub>H</sub> (G,I) and V<sub>L</sub> (F,H). NS and ** indicate not significant, or significant (p<0.036) difference between mean numbers of mutations by one-way Wilcoxon non-parametric test.</p

    H1<sup>+</sup> IgG<sup>+</sup> MBCs frequencies measured by flow-cytometry and by ELISPOT correlated linearly.

    No full text
    <p><b>A–B.</b> Specificity of the staining with the rH1 bait from A/Solomon Island/3/06. PBMCs (1.6×10<sup>8</sup>) from anonymous blood donors were stained with Live/Dead, incubated with an H3N2 mono-bulk vaccine subunit (from A/Panama/2007/1999), and then stained with Alexa647-conjugated HSA (6×10<sup>7</sup>), or Alexa647-conjugated rH1 (1×10<sup>8</sup>), and with an antiCD20 mAb. <b>A.</b> Binding pattern of HSA (A647-HSA; left panel) and of rH1 (A647-rH1; right panel) in the CD20<sup>+</sup> B-cell gates. <b>B.</b> H1<sup>+</sup> B-cells identified in A were sorted (n = 8215), mixed with autologous CD20<sup>neg</sup> cells in the ratio of 1∶50 and activated with CpG and IL-2 for 5 days <i>in vitro</i>. Unsorted PBMC and CD20<sup>neg</sup> cells were also cultured in the same manner as controls. After 5 days, equal numbers of cultured cells were harvested and assayed by ELISPOT for numbers of cells secreting IgG and IgG specific for H1N1 (from A/Solomon Island/3/06). <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0070620#s2" target="_blank">Results</a> are expressed as number of antibody secreting cells (ASC) normalized to 10<sup>6</sup> cultured cells assayed by ELISPOT. Nd indicates undetectable ASC. <b>C.</b> Distribution of IgG<sup>+</sup> B-cells among H1<sup>neg</sup> and H1<sup>+</sup> B cells expressing or not the CD27 B cell memory marker; shown is one representative subject. <b>D.</b> Replicates of frozen PBMCs from 4 anonymous blood donors were assayed by conventional ELISPOT, or incubated with an H3N2 mono-bulk vaccine subunit and stained with rH1, and anti-CD20 plus anti-human IgG antibodies. The scatter plot depicts paired values of H1<sup>+</sup> IgG<sup>+</sup> B-cell frequencies measured by flow-cytometry (y-axis) and by ELISPOT (x-axis) across three different experimental sessions. Shown are: the regression line with the related 95% confidence interval (gray areas), slope, intercepts, R<sup>2</sup> and p-value. <b>E.</b> Variability plot showing mean standard deviations of the measurements done by ELISPOT and flow-cytometry. The three dotted lines mark the grand mean and the upper and lower control limits.</p

    Vaccination induced changes in the pool of H1+ B-cells.

    No full text
    <p>PBMCs samples collected before (day 0) and at 3 and 6 weeks after vaccination from four seasonal influenza vaccinees, were pre-incubated with H3N2 subunit (from A/Panama/2007/1999) and then stained with rH1 A/Solomon Island/3/06) and mAbs anti-CD20, anti-CD27 and anti-human IgG. <b>A.</b> Dot plots gated on CD20<sup>+</sup> B-cells showing the distribution of H1<sup>+</sup> (middle panels) and H1<sup>neg</sup> (bottom panels) B-cells from donor #a across: the mature memory (CD27<sup>+</sup>) and putatively naive (CD27<sup>neg</sup>) CD20+ B-cell subsets (upper panels); un-switched mature memory (CD27<sup>+</sup>IgG<sup>neg</sup>), IgG-switched mature (CD27<sup>+</sup>IgG<sup>+</sup>) and immature (CD27<sup>neg</sup>IgG<sup>+</sup>) memory B-cells. <b>B.</b> Numbers of circulating H1<sup>+</sup> CD20<sup>+</sup> B-cells in 4 vaccinees before and at 3 and 6 weeks after seasonal vaccination are overlaid with paired titers of antibodies inhibiting virus-induced hemmaglutination measured in their blood. The frequencies of H1<sup>+</sup> B-cells are normalized according to the frequencies of CD20<sup>+</sup> B-cells in 10<sup>6</sup> PBMCs. <b>C.</b> Distribution of circulating H1<sup>+</sup> and H1<sup>neg</sup> B-cells across same subsets identified in <b>B</b> in all vaccinees.</p
    corecore