24 research outputs found

    Resistance mutations outside the integrase coding region have an effect on human immunodeficiency virus replicative fitness but do not affect its susceptibility to integrase strand transfer inhibitors.

    Get PDF
    Most studies describing phenotypic resistance to integrase strand transfer inhibitors have analyzed viruses carrying only patient-derived HIV-1 integrase genes (INT-recombinant viruses). However, to date, many of the patients on INSTI-based treatment regimes, such as raltegravir (RAL), elvitegravir (EVG), and dolutegravir (DTG) are infected with multidrug-resistant HIV-1 strains. Here we analyzed the effect of drug resistance mutations in Gag (p2/NCp7/p1/p6), protease (PR), reverse transcriptase (RT), and integrase (IN) coding regions on susceptibility to INSTIs and viral replicative fitness using a novel HIV-1 phenotyping assay. Initial characterization based on site-directed mutant INSTI-resistant viruses confirmed the effect of a series of INSTI mutations on reduced susceptibility to EVG and RAL and viral replicative fitness (0.6% to 99% relative to the HIV-1NL4-3 control). Two sets of recombinant viruses containing a 3,428-bp gag-p2/NCp7/p1/p6/pol-PR/RT/IN (p2-INT) or a 1,088 bp integrase (INT) patient-derived fragment were constructed from plasma samples obtained from 27 virologic failure patients participating in a 48-week dose-ranging study of elvitegravir, GS-US-183-0105. A strong correlation was observed when susceptibility to EVG and RAL was assayed using p2-INT- vs. INT-recombinant viruses (Pearson coefficient correlation 0.869 and 0.918, P<0.0001 for EVG and RAL, respectively), demonstrating that mutations in the protease and RT have limited effect on susceptibility to these INSTIs. On the other hand, the replicative fitness of viruses harboring drug resistance mutations in PR, RT, and IN was generally impaired compared to viruses carrying only INSTI-resistance mutations. Thus, in the absence of drug pressure, drug resistance mutations in the PR and RT contribute to decrease the replicative fitness of the virus already impaired by mutations in the integrase. The use of recombinant viruses containing most or all HIV-1 regions targeted by antiretroviral drugs might be essential to understand the collective effect of epistatic interactions in multidrug-resistant viruses

    Clinical and virological parameters of 27 HIV-infected individuals participating in the GS-US-183-0105 study of elvitegravir.

    No full text
    a<p>Major mutations associated with resistance to INSTI as described <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0065631#pone.0065631-McColl1" target="_blank">[4]</a>, <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0065631#pone.0065631-Blanco1" target="_blank">[11]</a>.</p>b<p>Plasma HIV viral load (log10 copies/ml).</p><p>INSTI-R, mutations associated with resistance to INSTI; PI (1), number of primary mutations associated with resistance to PI; PI (2), number of secondary mutations associated with resistance to PI; #TAMs, number of thymidine analogue-associated mutations; #NAMs, number of nucleoside analogue-associated mutations; NNRTI (1), number of primary mutations associated with resistance to NNRTI. EC<sub>50</sub> FC, based on VIRALARTS™HIV three independent EC<sub>50</sub> replicates for each drug were used to calculate the fold changes (FC) of the query viruses relative to the HIV-1<sub>NL4-3</sub> control and the mean EC<sub>50</sub> FC is indicated. MAX, complete virus inhibition was not achieved using the maximum drug concentration, i.e., virus was completely resistant to the respective antiretroviral drug.</p

    Replicative fitness of p2-INT-recombinant viruses carrying mutations associated with resistance to INSTIs in the absence and presence of EVG or RAL.

    No full text
    <p>(A) Fifteen p2-INT-recombinant viruses (i.e., 14 INSTI-resistance and the HIV-1<sub>NL4-3</sub> wild-type virus) were evaluated for their ability to replicate in MT-4 cells in the absence of drug pressure. Virus replication was quantified by measuring reverse transcriptase (RT) activity in the cell-free supernatant. Error bars indicate the range of values obtained from three independent experiments. (B) Viral replication slopes were calculated using the slopes between RLU values at days 0 & 3, 0 & 4, 0 & 5, and 0 & 6. All four slope values for each virus were used to calculate the mean, standard deviation, and 10<sup>th</sup> & 90<sup>th</sup> percentiles. Differences in the mean values were calculated using a One Way Analysis of Variance test and the significance difference from HIV-1<sub>NL4-3</sub> calculated using the Bonferroni’s Multiple Comparison Test. The replication kinetics of viruses marked with an asterisk (*) were significantly different from the HIV-1<sub>NL4-3</sub> control (p<0.05, 95% CI). Each p2-INT-recombinant virus was competed against the HIV-1<sub>NL4-3</sub> control in the absence (C) or presence (D) of EVG (0.01 nM) or RAL (1 nM) and their replicative fitness calculated and expressed as a percentage of the replicative fitness of the reference virus (HIV-1<sub>NL4-3</sub>) as described <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0065631#pone.0065631-Weber4" target="_blank">[55]</a>, <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0065631#pone.0065631-Weber5" target="_blank">[69]</a>. The Ratio of Replicative Fitness was calculated dividing the % Replicative Fitness in the presence of drug (EVG or RAL) by the % Replicative Fitness in the absence of drug pressure; e.g., 105.5% ÷ 0.6% = 176x, for Q148R. Values represent results obtained from single growth competition experiments.</p
    corecore