The effect of raltegravir intensification on low-level residual viremia in HIV-infected patients on antiretroviral therapy: a randomized controlled trial

Rajesh T Gandhi, Lu Zheng, Ronald J Bosch, Ellen S Chan, David M Margolis, Sarah Read, Beatrice Kallungal, Sarah Palmer, Kathy Medvik, Michael M Lederman, Nadia Alatrakchi, Jeffrey M Jacobson, Ann Wiegand, Mary Kearney, John M Coffin, John W Mellors, Joseph J Eron, AIDS Clinical Trials Group A5244 team, Randi Leavitt, Barbara Philpotts, Betty A Donoval, Robert Levaro, Ana Martinez, Lisa M Demeter, Richard D'Aquila, Carla Pettinelli, Jennifer Janik, Heather Springer, Amy Jennings, Rajesh T Gandhi, Lu Zheng, Ronald J Bosch, Ellen S Chan, David M Margolis, Sarah Read, Beatrice Kallungal, Sarah Palmer, Kathy Medvik, Michael M Lederman, Nadia Alatrakchi, Jeffrey M Jacobson, Ann Wiegand, Mary Kearney, John M Coffin, John W Mellors, Joseph J Eron, AIDS Clinical Trials Group A5244 team, Randi Leavitt, Barbara Philpotts, Betty A Donoval, Robert Levaro, Ana Martinez, Lisa M Demeter, Richard D'Aquila, Carla Pettinelli, Jennifer Janik, Heather Springer, Amy Jennings

Abstract

Background: Most HIV-1-infected patients on effective antiretroviral therapy (ART) with plasma HIV-1 RNA levels below the detection limits of commercial assays have residual viremia measurable by more sensitive methods. We assessed whether adding raltegravir lowered the level of residual viremia in such patients.

Methods and findings: Patients receiving ART who had plasma HIV-1 RNA levels below 50 copies/mL but detectable viremia by single copy assay (SCA) were randomized to add either raltegravir or placebo to their ART regimen for 12 weeks; patients then crossed-over to the other therapy for an additional 12 weeks while continuing pre-study ART. The primary endpoint was the plasma HIV-1 RNA by SCA averaged between weeks 10 and 12 (10/12) compared between treatment groups. Fifty-three patients were enrolled. The median screening HIV-1 RNA was 1.7 copies/mL. The HIV-1 RNA level at weeks 10/12 did not differ significantly between the raltegravir-intensified (n = 25) and the placebo (n = 24) groups (median 1.2 versus 1.7 copies/mL, p = 0.55, Wilcoxon rank sum test), nor did the change in HIV-1 RNA level from baseline to week 10/12 (median -0.2 and -0.1 copies/mL, p = 0.71, Wilcoxon rank sum test). There was also no significant change in HIV-1 RNA level from weeks 10/12 to weeks 22/24 after patients crossed-over. There was a greater CD4 cell count increase from baseline to week 12 in the raltegravir-intensified group compared with the placebo group (+42 versus -44 cells/mm(3), p = 0.082, Wilcoxon rank sum test), which reversed after the cross-over. This CD4 cell count change was not associated with an effect of raltegravir intensification on markers of CD4 or CD8 cell activation in blood.

Conclusion: In this randomized, double-blind cross-over study, 12 weeks of raltegravir intensification did not demonstrably reduce low-level plasma viremia in patients on currently recommended ART. This finding suggests that residual viremia does not arise from ongoing cycles of HIV-1 replication and infection of new cells. New therapeutic strategies to eliminate reservoirs that produce residual viremia will be required to eradicate HIV-1 infection.

Trial registration: ClinicalTrials.gov NCT00515827

Conflict of interest statement

JWM: Gilead Sciences (consultant); Merck (consultant and grant support); Chimerix (consultant); RFS Pharmaceuticals (owns stock options). JJE: Merck, GlaxoSmithKline (consultant and grant support), Bristol-Myers Squibb, Tibotec, Chimerix, Avexa, and Tobira (consultant). RTG: Tibotec (research grant support) and Gilead (grant support). DM: Merck, Bristol-Myers Squibb, and GlaxoSmithKline (consultant and research support); Gilead Sciences (research support and common stock); Chimerix (consultant); Roche, Trimeris, and Tibotec (research support).

Figures

Figure 1. Participant disposition.
Figure 1. Participant disposition.
Figure 2. Study schema.
Figure 2. Study schema.
Figure 3. No reduction in low-level residual…
Figure 3. No reduction in low-level residual viremia after raltegravir intensification.
(A) HIV-1 RNA values in patients in the raltegravir-first group (red diamonds) compared with participants in the placebo-first group (blue circles). Shaded areas designate time periods during which participants were receiving raltegravir intensification. Open symbols represent results for which one or both single copy assay (SCA) determinations were below the lower limit of quantification. Median SCA values at each time point are reported (horizontal bars). The average HIV-1 RNA level at week 10/12—the primary endpoint of the study—did not differ significantly between the raltegravir-first and the placebo-first groups: median 1.2 vs. 1.7 copies/mL (p = 0.55, Wilcoxon rank sum test). (B) Median HIV-1 RNA levels in participants adding raltegravir-first at week 0 and then crossing over to placebo at week 12 (red solid line) compared with participants adding placebo-first at week 0 and then crossing over to raltegravir at week 12 (blue dashed line). The lines connect medians. The bars represent interquartile ranges (25th and 75th percentiles).

References

    1. Dornadula G, Zhang H, VanUitert B, Stern J, Livornese L, Jr, et al. Residual HIV-1 RNA in blood plasma of patients taking suppressive highly active antiretroviral therapy. JAMA. 1999;282:1627–1632.
    1. Maldarelli F, Palmer S, King MS, Wiegand A, Polis MA, et al. ART suppresses plasma HIV-1 RNA to a stable set point predicted by pretherapy viremia. PLoS Pathog. 2007;3:e46.
    1. Palmer S, Maldarelli F, Wiegand A, Bernstein B, Hanna GJ, et al. Low-level viremia persists for at least 7 years in patients on suppressive antiretroviral therapy. Proc Natl Acad Sci U S A. 2008;105:3879–3884.
    1. Coiras M, Lopez-Huertas MR, Perez-Olmeda M, Alcami J. Understanding HIV-1 latency provides clues for the eradication of long-term reservoirs. Nat Rev Microbiol. 2009;7:798–812.
    1. Dinoso JB, Kim SY, Wiegand AM, Palmer SE, Gange SJ, et al. Treatment intensification does not reduce residual HIV-1 viremia in patients on highly active antiretroviral therapy. Proc Natl Acad Sci U S A. 2009;106:9403–9408.
    1. McMahon D, Jones J, Wiegand A, Gange S, Kearney M, et al. Short Course Raltegravir Intensification Does Not Reduce Persistent Low Level Viremia in Patients Suppressed on Combination Antiretroviral Therapy. Clin Infect Dis. 2010;50:912–919.
    1. Steigbigel RT, Cooper DA, Kumar PN, Eron JE, Schechter M, et al. Raltegravir with optimized background therapy for resistant HIV-1 infection. N Engl J Med. 2008;359:339–354.
    1. Lennox JL, DeJesus E, Lazzarin A, Pollard RB, Madruga JV, et al. Safety and efficacy of raltegravir-based versus efavirenz-based combination therapy in treatment-naive patients with HIV-1 infection: a multicentre, double-blind randomised controlled trial. Lancet. 2009;374:796–806.
    1. Palmer S, Wiegand AP, Maldarelli F, Bazmi H, Mican JM, et al. New real-time reverse transcriptase-initiated PCR assay with single-copy sensitivity for human immunodeficiency virus type 1 RNA in plasma. J Clin Microbiol. 2003;41:4531–4536.
    1. Clement LT, Vink PE, Bradley GE. Novel immunoregulatory functions of phenotypically distinct subpopulations of CD4+ cells in the human neonate. J Immunol. 1990;145:102–108.
    1. Rabian-Herzog C, Lesage S, Gluckman E. Characterization of lymphocyte subpopulations in cord blood. Bone Marrow Transplant. 1992;9(Suppl 1):64–67.
    1. Dianzani U, Funaro A, DiFranco D, Garbarino G, Bragardo M, et al. Interaction between endothelium and CD4+CD45RA+ lymphocytes. Role of the human CD38 molecule. J Immunol. 1994;153:952–959.
    1. Havlir DV, Strain MC, Clerici M, Ignacio C, Trabattoni D, et al. Productive infection maintains a dynamic steady state of residual viremia in human immunodeficiency virus type 1-infected persons treated with suppressive antiretroviral therapy for five years. J Virol. 2003;77:11212–11219.
    1. Sedaghat AR, Siliciano JD, Brennan TP, Wilke CO, Siliciano RF. Limits on replenishment of the resting CD4+ T cell reservoir for HIV in patients on HAART. PLoS Pathog. 2007;3:e122.
    1. Kieffer TL, Finucane MM, Nettles RE, Quinn TC, Broman KW, et al. Genotypic analysis of HIV-1 drug resistance at the limit of detection: virus production without evolution in treated adults with undetectable HIV loads. J Infect Dis. 2004;189:1452–1465.
    1. Buzon MJ, Massanella M, Llibre JM, Esteve A, Dahl V, et al. HIV-1 replication and immune dynamics are affected by raltegravir intensification of HAART-suppressed subjects. Nat Med. 2010;16:460–465.
    1. Hunt PW, Martin JN, Sinclair E, Bredt B, Hagos E, et al. T cell activation is associated with lower CD4+ T cell gains in human immunodeficiency virus-infected patients with sustained viral suppression during antiretroviral therapy. J Infect Dis. 2003;187:1534–1543.
    1. Goicoechea M, Smith DM, Liu L, May S, Tenorio AR, et al. Determinants of CD4+ T cell recovery during suppressive antiretroviral therapy: association of immune activation, T cell maturation markers, and cellular HIV-1 DNA. J Infect Dis. 2006;194:29–37.
    1. Gandhi RT, Spritzler J, Chan E, Asmuth DM, Rodriguez B, et al. Effect of baseline- and treatment-related factors on immunologic recovery after initiation of antiretroviral therapy in HIV-1-positive subjects: results from ACTG 384. J Acquir Immune Defic Syndr. 2006;42:426–434.
    1. Hsue PY, Hunt PW, Schnell A, Kalapus SC, Hoh R, et al. Role of viral replication, antiretroviral therapy, and immunodeficiency in HIV-associated atherosclerosis. AIDS. 2009;23:1059–1067.
    1. Richman DD, Margolis DM, Delaney M, Greene WC, Hazuda D, et al. The challenge of finding a cure for HIV infection. Science. 2009;323:1304–1307.

Source: PubMed

3
Abonnere