Impact of intensification with raltegravir on HIV-1-infected individuals receiving monotherapy with boosted PIs

Maria C Puertas, Elisabet Gómez-Mora, José R Santos, José Moltó, Víctor Urrea, Sara Morón-López, Agueda Hernández-Rodríguez, Silvia Marfil, Marta Martínez-Bonet, Lurdes Matas, Mª Angeles Muñoz-Fernández, Bonaventura Clotet, Julià Blanco, Javier Martinez-Picado, Maria C Puertas, Elisabet Gómez-Mora, José R Santos, José Moltó, Víctor Urrea, Sara Morón-López, Agueda Hernández-Rodríguez, Silvia Marfil, Marta Martínez-Bonet, Lurdes Matas, Mª Angeles Muñoz-Fernández, Bonaventura Clotet, Julià Blanco, Javier Martinez-Picado

Abstract

Background: Monotherapy with ritonavir-boosted PIs (PI/r) has been used to simplify treatment of HIV-1-infected patients. In previous studies raltegravir intensification evidenced ongoing viral replication and reduced T cell activation, preferentially in subjects receiving PI-based triple ART. However, data about low-level viral replication and its consequences in patients receiving PI/r monotherapy are scarce.

Methods: We evaluated the impact of 24 weeks of intensification with raltegravir on markers of viral persistence, cellular immune activation and inflammation biomarkers in 33 patients receiving maintenance PI/r monotherapy with darunavir or lopinavir boosted with ritonavir. ClinicalTrials.gov identifier: NCT01480713.

Results: The addition of raltegravir to PI/r monotherapy resulted in a transient increase in 2-LTR (long-terminal repeat) circles in a significant proportion of participants, along with decreases in CD8+ T cell activation levels and a temporary increase in the expression of the exhaustion marker CTLA-4 in peripheral T lymphocytes. Intensification with raltegravir also reduced the number of samples with intermediate levels of residual viraemia (10-60 HIV-1 RNA copies/mL) compared with samples taken during PI/r monotherapy. However, there were no changes in cell-associated HIV-1 DNA in peripheral CD4+ T cells or soluble inflammatory biomarkers (CD14, IP-10, IL-6, C-reactive protein and D-dimer).

Conclusions: Intensification of PI/r monotherapy with raltegravir revealed persistent low-level viral replication and reduced residual viraemia in some patients during long-term PI/r monotherapy. The concomitant change in T cell phenotype suggests an association between active viral production and T cell activation. These results contribute to understanding the lower efficacy rates of PI/r monotherapies compared with triple therapies in clinical trials.

Figures

Figure 1.
Figure 1.
Effect of raltegravir intensification on residual viraemia. (a) Blood samples were obtained before intensification (week −8, week −4 and baseline), during intensification (from baseline to 24 weeks) and for up to 24 weeks after withdrawal of raltegravir (week 48). (b) Raw data from ultrasensitive viral load determination (left y-axis) and the percentages of samples with >10 HIV-1 RNA copies/mL of plasma (right y-axis) are shown for each timepoint. The grey zone represents the intensification period (week 0 to 24). (c) Difference in the probability of detecting intermediate residual viraemia levels (10–60 HIV-1 RNA copies/mL) during raltegravir intensification and after drug withdrawal, compared with the pre-intensification period. A mixed-effects logistic model was fitted for the binary outcome indicative of intermediate residual viraemia, assuming constant probabilities of exceeding 10 copies of residual viraemia for each patient in each period. BL, baseline; RAL, raltegravir; vRNA, HIV-1 RNA.
Figure 2.
Figure 2.
Viral reservoir dynamics upon intensification of PI monotherapy with raltegravir. (a) Increases in 2-LTR circles compared with individual baseline levels are shown as the median and IQR. The frequency diagram in (b) summarizes the timepoint at which each individual reached the maximum level of 2-LTR. (c) The dynamics of the proviral reservoir is shown as a fold change in total HIV-1 DNA in CD4+ T cells compared with individual baseline levels. The median and IQR are shown. The grey zone represents the intensification period (week 0 to 24). BL, baseline; vDNA, HIV-1 DNA.
Figure 3.
Figure 3.
Effect of intensification with raltegravir on T cell activation. The graph shows the immune activation levels (determined as a percentage of CD38+ cells) of all HIV-infected individuals analysed both in CD4+ (open symbols) (a) and CD8+ (filled symbols) (b) T cell populations. The median and IQR are represented. Asterisks denote significant differences between timepoints detected using the Wilcoxon signed rank test (*P <0.05, **P <0.01, ***P <0.001). T cell activation was compared between 2-LTR+ (black continuous line) and 2-LTR− (grey broken line) subgroups within CD4+ (c) and CD8+ (d) T cell subsets. The median and IQR are shown. Asterisks denote significant differences between 2-LTR subgroups detected using a linear mixed-effects model (*P <0.05, **P <0.01, ***P <0.001). The grey zone represents the intensification period (week 0 to 24).
Figure 4.
Figure 4.
Effect of intensification with raltegravir on T cell exhaustion. The graph shows immune exhaustion levels (determined as a percentage of CTLA-4+ cells) of all HIV-infected individuals analysed both in CD4+ (open symbols) (a) and CD8+ (filled symbols) (b) T cell populations. The median and IQR are represented. Asterisks denote significant differences between the timepoints assessed using the Wilcoxon signed rank test (*P <0.05, **P <0.01, ***P <0.001). T cell exhaustion was compared between 2-LTR+ (black continuous line) and 2-LTR− (grey broken line) subgroups within CD4+ (c) and CD8+ (d) T cell subsets. The median and IQR are shown. Asterisks denote significant differences between 2-LTR subgroups assessed using a linear mixed-effects model (*P <0.05, **P <0.01, ***P <0.001). The grey zone represents the intensification period (week 0 to 24).

References

    1. Friis-Møller N, Sabin CA, Weber R. et al. Combination antiretroviral therapy and the risk of myocardial infarction. N Engl J Med 2003; 349: 1993–2003.
    1. Gallant J, Parish M, Keruly J. et al. Changes in renal function associated with tenofovir disoproxil fumarate treatment, compared with nucleoside reverse-transcriptase inhibitor treatment. Clin Infect Dis 2005; 40: 1194–8.
    1. Brinkman K, ter Hofstede HJ, Burger DM. et al. Adverse effects of reverse transcriptase inhibitors: mitochondrial toxicity as common pathway. AIDS 1998; 12: 1735–44.
    1. Woodward C, Hall A, Williams I. et al. Tenofovir-associated renal and bone toxicity. HIV Med 2009; 10: 482–7.
    1. Oddershede L, Walker S, Paton N. et al. Cost-effectiveness analysis of protease inhibitor monotherapy vs. ongoing triple-therapy in the long-term management of HIV patients. J Int AIDS Soc 2014; 17: 19498..
    1. Palmer S, Maldarelli F, Wiegand A. et al. Low-level viremia persists for at least 7 years in patients on suppressive antiretroviral therapy. Proc Natl Acad Sci USA 2008; 105: 3879–84.
    1. Buzón MJ, Massanella M, Llibre JM. et al. HIV-1 replication and immune dynamics are affected by raltegravir intensification of HAART-suppressed subjects. Nat Med 2010; 16: 460–5.
    1. Hatano H, Strain MC, Scherzer R. et al. Increase in 2-long terminal repeat circles and decrease in D-dimer after raltegravir intensification in patients with treated HIV Infection: a randomized, placebo-controlled trial. J Infect Dis 2013; 208: 1436–42.
    1. Fletcher CV, Staskus K, Wietgrefe SW. et al. Persistent HIV-1 replication is associated with lower antiretroviral drug concentrations in lymphatic tissues. Proc Natl Acad Sci USA 2014; 111: 2307–12.
    1. Llibre JM, Buzón MJ, Massanella M. et al. Treatment intensification with raltegravir in subjects with sustained HIV-1 viraemia suppression: a randomized 48-week study. Antivir Ther 2012; 17: 355–64.
    1. Massanella M, Esteve A, Buzón MJ. et al. Dynamics of CD8 T-cell activation after discontinuation of HIV treatment intensification. J Acquir Immune Defic Syndr 2013; 63: 152–60.
    1. Muñoz-Moreno JA, Fumaz CR, Ferrer MJ. et al. Assessing self-reported adherence to HIV therapy by questionnaire: the SERAD (Self-Reported Adherence) study. AIDS Res Hum Retroviruses 2007; 23: 1166–75.
    1. Martínez-Bonet M, Puertas MC, Fortuny C. et al. Establishment and replenishment of the viral reservoir in perinatally HIV-1-infected children initiating very early antiretroviral therapy. Clin Infect Dis 2015; 61: 1169–78.
    1. Puertas MC, Noguera-Julian M, Massanella M. et al. Lack of concordance between residual viremia and viral variants driving de novo infection of CD4+ T cells on ART. Retrovirology 2016; 13: 51.
    1. Massanella M, Tural C, Papagno L. et al. Changes in T-cell subsets in HIV-HCV-coinfected patients during pegylated interferon-α2a plus ribavirin treatment. Antivir Ther 2010; 15: 333–42.
    1. Arribas JR, Girard PM, Paton N. et al. Efficacy of protease inhibitor monotherapy vs. triple therapy: meta-analysis of data from 2303 patients in 13 randomized trials. HIV Med 2016; 17: 358–67.
    1. EACS Guidelines 2017. .
    1. Guidelines for the Use of Antiretroviral Agents in Adults and Adolescents Living with HIV 2017. .
    1. Arenas-Pinto A, Milinkovic A, Peppa D. et al. Systemic inflammation and residual viraemia in HIV-positive adults on protease inhibitor monotherapy: a cross-sectional study. BMC Infect Dis 2015; 15: 138..
    1. Estébanez M, Stella-Ascariz N, Mingorance J. et al. Inflammatory, procoagulant markers and HIV residual viremia in patients receiving protease inhibitor monotherapy or triple drug therapy: a cross-sectional study. BMC Infect Dis 2014; 14: 379..
    1. Gandhi RT, Zheng L, Bosch RJ. et al. The effect of raltegravir intensification on low-level residual viremia in HIV-infected patients on antiretroviral therapy: a randomized controlled trial. PLoS Med 2010; 7: e1000321..
    1. McMahon D, Jones J, Wiegand A. et al. Short-course raltegravir intensification does not reduce persistent low-level viremia in patients with HIV-1 suppression during receipt of combination antiretroviral therapy. Clin Infect Dis 2010; 50: 912–9.
    1. Sharkey M, Triques K, Kuritzkes DR. et al. In vivo evidence for instability of episomal human immunodeficiency virus type 1 cDNA. J Virol 2005; 79: 5203–10.
    1. Lambert-Niclot S, Flandre P, Valantin MA. et al. Similar evolution of cellular HIV-1 DNA level in darunavir/ritonavir monotherapy versus triple therapy in MONOI -ANRS136 trial over 96 weeks. PLoS One 2012; 7: e41390.
    1. Geretti AM, Arribas JR, Lathouwers E. et al. Dynamics of cellular HIV-1 DNA levels over 144 weeks of darunavir/ritonavir monotherapy versus triple therapy in the MONET trial. HIV Clin Trials 2013; 14: 45–50.
    1. Massanella M, Negredo E, Puig J. et al. Raltegravir intensification shows differing effects on CD8 and CD4 T cells in HIV-infected HAART-suppressed individuals with poor CD4 T-cell recovery. AIDS 2012; 26: 2285–93.
    1. Kaufmann DE, Kavanagh DG, Pereyra F. et al. Upregulation of CTLA-4 by HIV-specific CD4+ T cells correlates with disease progression and defines a reversible immune dysfunction. Nat Immunol 2007; 8: 1246–54.
    1. Leng Q, Bentwich Z, Magen E. et al. CTLA-4 upregulation during HIV infection: association with anergy and possible target for therapeutic intervention. AIDS 2002; 16: 519–29.
    1. Kaufmann DE, Walker BD.. PD-1 and CTLA-4 inhibitory cosignaling pathways in HIV infection and the potential for therapeutic intervention. J Immunol 2009; 182: 5891–7.
    1. Day CL, Kaufmann DE, Kiepiela P. et al. PD-1 expression on HIV-specific T cells is associated with T-cell exhaustion and disease progression. Nature 2006; 443: 350–4.
    1. Larsson M, Shankar EM, Che KF. et al. Molecular signatures of T-cell inhibition in HIV-1 infection. Retrovirology 2013; 10: 31..
    1. Kassu A, Marcus RA, D’Souza MB. et al. Regulation of virus-specific CD4+ T cell function by multiple costimulatory receptors during chronic HIV infection. J Immunol 2010; 185: 3007–18.
    1. Stöhr W, Dunn DT, Arenas-Pinto A. et al. Factors associated with virological rebound in HIV-infected patients receiving protease inhibitor monotherapy. AIDS 2016; 30: 2617–24.
    1. Gianotti N, Cozzi-Lepri A, Antinori A. et al. Refining criteria for selecting candidates for a safe lopinavir/ritonavir or darunavir/ritonavir monotherapy in HIV-infected virologically suppressed patients. PLoS One 2017; 12: e0171611..

Source: PubMed

3
Předplatit