15 research outputs found

    Potential of conventional & bispecific broadly neutralizing antibodies for prevention of HIV-1 subtype A, C & D infections

    No full text
    <div><p>There is great interest in passive transfer of broadly neutralizing antibodies (bnAbs) and engineered bispecific antibodies (Abs) for prevention of HIV-1 infections due to their <i>in vitro</i> neutralization breadth and potency against global isolates and long <i>in vivo</i> half-lives. We compared the potential of eight bnAbs and two bispecific Abs currently under clinical development, and their 2 Ab combinations, to prevent infection by dominant HIV-1 subtypes in sub-Saharan Africa. Using <i>in vitro</i> neutralization data for Abs against 25 subtype A, 100 C, and 20 D pseudoviruses, we modeled neutralization by single Abs and 2 Ab combinations assuming realistic target concentrations of 10ÎŒg/ml total for bnAbs and combinations, and 5ÎŒg/ml for bispecifics. We used IC<sub>80</sub> breadth-potency, completeness of neutralization, and simultaneous coverage by both Abs in the combination as metrics to characterize prevention potential. Additionally, we predicted <i>in vivo</i> protection by Abs and combinations by modeling protection as a function of <i>in vitro</i> neutralization based on data from a macaque simian-human immunodeficiency virus (SHIV) challenge study. Our model suggests that nearly complete neutralization of a given virus is needed for <i>in vivo</i> protection (~98% neutralization for 50% relative protection). Using the above metrics, we found that bnAb combinations should outperform single bnAbs, as expected; however, different combinations are optimal for different subtypes. Remarkably, a single bispecific 10E8-iMAb, which targets HIV Env and host-cell CD4, outperformed all combinations of two conventional bnAbs, with 95–97% predicted relative protection across subtypes. Combinations that included 10E8-iMAb substantially improved protection over use of 10E8-iMAb alone. Our results highlight the promise of 10E8-iMAb and its combinations to prevent HIV-1 infections in sub-Saharan Africa.</p></div

    Modeling <i>in vivo</i> protection as a function of <i>in vitro</i> neutralization.

    No full text
    <p>(A) The maximum likelihood model for the <i>in vivo</i> probability of infection as a function of IIP using data from the low-dose, repeated SHIV challenge macaque study [<a href="http://www.plospathogens.org/article/info:doi/10.1371/journal.ppat.1006860#ppat.1006860.ref021" target="_blank">21</a>] is shown in black line. This model had the same parameters across all Abs. The IIP values at the time of challenge when animals were infected are shown on top with ‘+’ and when animals were protected are shown below with ‘x’. Data for animals from different Ab groups are shown separately with different colors, and data for control animals (9 out of 33 challenges resulting in infection) are not shown. (B) The relative probability of infection as a function of concentration of the bispecific 10E8-iMAb against each clade C panel pseudovirus (grey lines) and the average across the panel viruses (red line).</p

    Potential of conventional & bispecific broadly neutralizing antibodies for prevention of HIV-1 subtype A, C & D infections - Fig 1

    No full text
    <p><b><i>In vitro</i> neutralization data for individual antibodies against A, C & D subtype pseudovirus panels.</b> (A) Distribution of HIV-1 subtypes in sub-Saharan Africa using the “Geography Search Interface” on the Los Alamos HIV Database. (B-D) Experimental IC<sub>80</sub> titers for antibodies against subtype C, A and D panels, respectively. Viruses are represented on rows and antibodies on columns. Red-yellow shades indicate more-less potent neutralization, and blue cells indicate IC<sub>80</sub> titers above experimental threshold or 10ÎŒg/ml. (E) Comparison of IC<sub>80</sub> distributions for each antibody across subtypes. The percent of viruses in each subtype with IC<sub>80</sub> above experimental threshold for each antibody are indicated in the figure. The IC<sub>80</sub> distributions between subtypes were compared for each antibody using Wilcoxon rank sum test and comparisons with p < 0.01 are indicated.</p

    Performance of bispecific Abs and 2 conventional bnAb combinations.

    No full text
    <p>(A-C) show the results for subtypes A, C and D, respectively. The left panels show the IC<sub>80</sub> breadth-potency curves using experimental titers for bispecifics and predicted titers for 2 bnAb combinations. The central panels show fraction maximum inhibition values for bispecific Abs and 2 bnAb combinations against viruses from a given subtype virus panel. Maximum inhibition values are calculated at 5ÎŒg/ml for bispecific Abs and at 5ÎŒg/ml of each bnAb for 2 bnAb combinations. The right panels show breadth-potency curves for 2 bnAb combination IC<sub>80</sub> titers by considering only those viruses that were neutralized by both bnAbs with single bnAb IC<sub>80</sub> < 5ÎŒg/ml.</p

    Predicted <i>in vivo</i> protection for individual Abs and combinations.

    No full text
    <p>The average relative <i>in vivo</i> probability of infection predicted using above modeling is shown for single Abs (A), combinations of two conventional Abs (B) and combinations with one or two bispecific Abs (C) for the subtype A (left), C (middle) and D (right) panels. The curves show the relative probability of infection at a given concentration averaged over pseudoviruses from each panel. For Ab combinations, the total concentration of both Abs is shown.</p
    corecore