Rapid viral rebound after analytical treatment interruption in patients with very small HIV reservoir and minimal on-going viral transcription

Pieter Pannus, Sofie Rutsaert, Stéphane De Wit, Sabine D Allard, Guido Vanham, Basiel Cole, Coca Nescoi, Joeri Aerts, Ward De Spiegelaere, Achilleas Tsoumanis, Marie-Madeleine Couttenye, Natacha Herssens, Marie-Angélique De Scheerder, Linos Vandekerckhove, Eric Florence, Pieter Pannus, Sofie Rutsaert, Stéphane De Wit, Sabine D Allard, Guido Vanham, Basiel Cole, Coca Nescoi, Joeri Aerts, Ward De Spiegelaere, Achilleas Tsoumanis, Marie-Madeleine Couttenye, Natacha Herssens, Marie-Angélique De Scheerder, Linos Vandekerckhove, Eric Florence

Abstract

Introduction: Viral remission after analytical treatment interruption (ATI), termed post-treatment control, has been described in a small proportion of HIV-positive patients. This phenomenon has been separately associated to both low levels of HIV-1 proviral DNA as well as cell-associated RNA. We investigated whether the combination of both parameters could help predict delayed viral rebound after treatment interruption (TI).

Methods: We conducted an open single-arm ATI study in four Belgian HIV reference centres from January 2016 to July 2018. Eligible participants were adults who had fewer than 50 HIV-1 RNA copies/mL for more than two years, more than 500 CD4 cells/µL for more than three months, and were in general good health. Consenting participants who had fewer than 66 copies total HIV-1 DNA (t-DNA) and fewer than 10 copies cell-associated HIV-1 unspliced RNA (US-RNA) per million peripheral blood mononuclear cells (PBMCs), interrupted therapy and were monitored closely. Antiretroviral therapy (ART) was resumed after two consecutive viral loads exceeding 1000 copies or one exceeding 10,000 copies/mL. The primary outcome was the proportion of participants with fewer than 50 HIV-1 RNA copies/mL 48 weeks after TI. Secondary outcomes were time to viral rebound, the frequency of serious adverse events (AEs) and evolution of t-DNA and US-RNA after TI.

Results: All 16 consenting participants who interrupted therapy experienced rapid viral rebound two to eight weeks after TI. No serious AEs were observed. Levels of t-DNA and US-RNA increased after TI but returned to pre-ATI levels after treatment restart. None of the studied demographic, clinical and biological parameters were predictive of time of viral rebound.

Conclusions: The combination of low levels of t-DNA and US-RNA in PBMCs, corresponding respectively to a small and transcriptionally silent viral reservoir, is not predictive of viral remission after TI in patients on ART.

Trial registration: ClinicalTrials.gov NCT02590354.

Keywords: HIV; cell-associated HIV RNA; post-treatment control; total HIV DNA; treatment interruption; viral reservoir.

© 2020 The Authors. Journal of the International AIDS Society published by John Wiley & Sons Ltd on behalf of the International AIDS Society.

Figures

Figure 1
Figure 1
(a) Overview of stage 1 and 2 of the study. Timepoints at which blood was drawn for plasma and PBMC separation are indicated with yellow arrows. ART is restarted when treatment resumption criteria are reached or 48 weeks after treatment interruption. (b) t‐DNA values in copies per million PBMCs for all stage 1 participants. Participants with t‐DNA values >66 copies per million PBMCs are indicated with grey dots (n = 75). Patients with t‐DNA values below 66 copies but US‐RNA values above 10 copies per million PBMCs are indicated with green dots (n = 3). Patients with t‐DNA and US‐RNA values below 66 and 10 copies per million PBMCs respectively are indicated with blue dots (n = 38) and are eligible for stage 2 participation. (c) Flow chart with number of patients screened until number of patients who completed the study. ART, antiretroviral therapy; ATI, analytical treatment interruption; PBMC, peripheral blood mononuclear cell; pVL, plasma viral load; t‐DNA, total HIV‐1 DNA; US‐RNA, cell‐associated HIV‐1 unspliced RNA.
Figure 2
Figure 2
(a) Plasma viral rebound dynamics after ATI until ART restart in 16 selected patients with very small viral reservoir, (b, c, d) dynamics of total and integrated HIV‐1 DNA and LTR HIV‐1 CA‐RNA over time in stage 2 patients. p‐Values are from Friedman Rank Sum tests and post‐hoc Nemenyi test. ART, antiretroviral therapy; ATI, analytical treatment interruption; LTR, long terminal repeat; PBMC, peripheral blood mononuclear cell; PW12: 12 weeks after restart ART; VR, timepoint at viral rebound just before restart ART, W, weeks; W0, timepoint at stop ART.
Figure 3
Figure 3
Additional viral parameters before ATI. (a) Ultra‐sensitive plasma viral load (in HIV‐1 RNA copies/mL). (b) Replication competent fraction (in infectious units per million CD4+ T cells, IUPM). (c, d) Viral release from 15 × 106 PBMCs (c) and 5 × 106 CD4+ T cells (d), either spontaneous or after stimulation with PMA + ionomycin (in log copies/mL). p Values are from Wilcoxon matched‐pairs signed‐rank test. ATI, analytical treatment interruption; PBMC, peripheral blood mononuclear cell.
Figure 4
Figure 4
Maximum likelihood phylogenetic trees of V1‐V3 env sequences from rebound plasma and positive quantitative viral outgrowth assay wells (outgrowth) for participants 4012, 3034 and 1011.

References

    1. Finzi D, Hermankova M, Pierson T, Carruth LM, Buck C, Chaisson RE, et al. Identification of a reservoir for HIV‐1 in patients on highly active antiretroviral therapy. Science. 1997;278(5341):1295–300.
    1. Deeks SG, Lewin SR, Ross AL, Ananworanich J, Benkirane M, Cannon P, et al. International AIDS Society global scientific strategy: towards an HIV cure 2016. Nat Med. 2016;22:839.
    1. Hocqueloux L, Prazuck T, Avettand‐Fenoel V, Lafeuillade A, Cardon B, Viard JP, et al. Long‐term immunovirologic control following antiretroviral therapy interruption in patients treated at the time of primary HIV‐1 infection. AIDS. 2010;24(10):1598–601.
    1. Van Gulck E, Heyndrickx L, Bracke L, Coppens S, Florence E, Buve A, et al. Control of viral replication after cessation of HAART. AIDS Res Ther. 2011;8(1):6.
    1. Lau JSY, Smith MZ, Lewin SR, McMahon JH. Clinical trials of antiretroviral treatment interruption in HIV‐infected individuals. AIDS. 2019;33(5):773–91.
    1. Namazi G, Fajnzylber JM, Aga E, Bosch R, Acosta EP, Sharaf R, et al. The Control of HIV after Antiretroviral Medication Pause (CHAMP) study: post‐treatment controllers identified from 14 clinical studies. J Infect Dis. 2018;218(12):1954–63.
    1. Maggiolo F, Di Filippo E, Comi L, Callegaro A. Post treatment controllers after treatment interruption in chronically HIV infected patients. AIDS. 2018;32:623–8.
    1. Saez‐Cirion A, Bacchus C, Hocqueloux L, Avettand‐Fenoel V, Girault I, Lecuroux C, et al. Post‐treatment HIV‐1 controllers with a long‐term virological remission after the interruption of early initiated antiretroviral therapy ANRS VISCONTI Study. PLoS Pathog. 2013;9:e1003211.
    1. Williams JP, Hurst J, Stohr W, Robinson N, Brown H, Fisher M, et al. HIV‐1 DNA predicts disease progression and post‐treatment virological control. eLife. 2014;3:e03821.
    1. Assoumou L, Weiss L, Piketty C, Burgard M, Melard A, Girard PM, et al. A low HIV‐DNA level in peripheral blood mononuclear cells at antiretroviral treatment interruption predicts a higher probability of maintaining viral control. AIDS. 2015;29(15):2003–7.
    1. Li JZ, Etemad B, Ahmed H, Aga E, Bosch RJ, Mellors JW, et al. The size of the expressed HIV reservoir predicts timing of viral rebound after treatment interruption. AIDS. 2016;30(3):343–53.
    1. Vanham G, Buve A, Florence E, Seguin‐Devaux C, Saez‐Cirion A. What is the significance of posttreatment control of HIV infection vis‐a‐vis functional cure? AIDS. 2014;28(4):603–5.
    1. Piketty C, Weiss L, Assoumou L, Burgard M, Melard A, Ragnaud JM, et al. A high HIV DNA level in PBMCs at antiretroviral treatment interruption predicts a shorter time to treatment resumption, independently of the CD4 nadir. J Med Virol. 2010;82(11):1819–28.
    1. Leal L, Guardo AC, Moron‐Lopez S, Salgado M, Mothe B, Heirman C, et al. Phase I clinical trial of an intranodally administered mRNA based therapeutic vaccine against HIV‐1 infection. AIDS. 2018;32(17):2533–45.
    1. Schvachsa N, Turk G, Burgard M, Dilernia D, Carobene M, Pippo M, et al. Examination of real‐time PCR for HIV‐1 RNA and DNA quantitation in patients infected with HIV‐1 BF intersubtype recombinant variants. J Virol Methods. 2007;140(1–2):222–7.
    1. Cillo AR, Hong F, Tsai A, Irrinki A, Kaur J, Sloan DD, et al. Blood biomarkers of expressed and inducible HIV‐1. AIDS. 2018;32(6):699–708.
    1. Laird GM, Rosenbloom DI, Lai J, Siliciano RF, Siliciano JD. Measuring the frequency of latent HIV‐1 in resting CD4(+) T cells using a limiting dilution coculture assay. Methods Mol Biol. 2016;1354:239–53.
    1. Kiselinova M, Geretti AM, Malatinkova E, Vervisch K, Beloukas A, Messiaen P, et al. HIV‐1 RNA and HIV‐1 DNA persistence during suppressive ART with PI‐based or nevirapine‐based regimens. J Antimicrob Chemother. 2015;70(12):3311–6.
    1. Horng S, Grady C. Misunderstanding in clinical research: distinguishing therapeutic misconception, therapeutic misestimation, and therapeutic optimism. Irb. 2003;25(1):11–6.
    1. Division of AIDS (DAIDS) . Table for grading the severity of adult and pediatric adverse events. Version 1.0. 2004. [cited 2020 Feb 10]; [updated 2009]. Available from:
    1. Yukl SA, Kaiser P, Kim P, Telwatte S, Joshi SK, Vu M, et al. HIV latency in isolated patient CD4(+) T cells may be due to blocks in HIV transcriptional elongation, completion, and splicing. Sci Transl Med. 2018;10(430): eaap9927.
    1. Palmer S, Wiegand AP, Maldarelli F, Bazmi H, Mican JM, Polis M, 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(10):4531–6.
    1. Josefsson L, von Stockenstrom S, Faria NR, Sinclair E, Bacchetti P, Killian M, et al. The HIV‐1 reservoir in eight patients on long‐term suppressive antiretroviral therapy is stable with few genetic changes over time. Proc Natl Acad Sci U S A. 2013;110(51):E4987–96.
    1. von Stockenstrom S, Odevall L, Lee E, Sinclair E, Bacchetti P, Killian M, et al. Longitudinal genetic characterization reveals that cell proliferation maintains a persistent HIV Type 1 DNA pool during effective HIV therapy. J Infect Dis. 2015;212(4):596–607.
    1. Edgar RC. MUSCLE: multiple sequence alignment with high accuracy and high throughput. Nucleic Acids Res. 2004;32(5):1792–7.
    1. Guindon S, Dufayard JF, Lefort V, Anisimova M, Hordijk W, Gascuel O. New algorithms and methods to estimate maximum‐likelihood phylogenies: assessing the performance of PhyML 3.0. Syst Biol. 2010;59(3):307–21.
    1. Laskey SB, Pohlmeyer CW, Bruner KM, Siliciano RF. Evaluating clonal expansion of HIV‐infected cells: optimization of PCR strategies to predict clonality. PLoS Pathog. 2016;12:e1005689.
    1. Calin R, Hamimi C, Lambert‐Niclot S, Carcelain G, Bellet J, Assoumou L, et al. Treatment interruption in chronically HIV‐infected patients with an ultralow HIV reservoir. AIDS. 2016;30(5):761–9.
    1. Colby DJ, Trautmann L, Pinyakorn S, Leyre L, Pagliuzza A, Kroon E, et al. Rapid HIV RNA rebound after antiretroviral treatment interruption in persons durably suppressed in Fiebig I acute HIV infection. Nat Med. 2018;24(7):923–6.
    1. Strongin Z, Sharaf R, VanBelzen DJ, Jacobson JM, Connick E, Volberding P, et al. Effect of short‐term ART interruption on levels of integrated HIV‐DNA. J Virol. 2018;92:e00285‐18.
    1. Mendoza P, Gruell H, Nogueira L, Pai JA, Butler AL, Millard K, et al. Combination therapy with anti‐HIV‐1 antibodies maintains viral suppression. Nature. 2018;561(7724):479–84.
    1. Salantes DB, Zheng Y, Mampe F, Srivastava T, Beg S, Lai J, et al. HIV‐1 latent reservoir size and diversity are stable following brief treatment interruption. J Clin Investig. 2018;128(7):3102–15.
    1. Lelievre JD, Hocqueloux L. Unintended HIV‐1 transmission to a sex partner in a study of a therapeutic vaccine candidate. J Infect Dis. 2019;220 Suppl 1:S5–6.
    1. Salantes DB, Zheng Y, Mampe F, Srivastava T, Beg S, Lai J, et al. HIV‐1 latent reservoir size and diversity are stable following brief treatment interruption. J Clin Invest. 2018;128(7):3102–15.
    1. Winckelmann A, Morcilla V, Shao W, Schleimann MH, Hojen JF, Schlub TE, et al. Genetic characterization of the HIV‐1 reservoir after Vacc‐4x and romidepsin therapy in HIV‐1‐infected individuals. AIDS. 2018;32(13):1793–802.
    1. Cohn LB, da Silva IT, Valieris R, Huang AS, Lorenzi JCC, Cohen YZ, et al. Clonal CD4+ T cells in the HIV‐1 latent reservoir display a distinct gene profile upon reactivation. Nat Med. 2018;24(5):604–9.
    1. De Scheerder MA, Vrancken B, Dellicour S, Schlub T, Lee E, Shao W, et al. Rebound is predominantly fueled by genetically identical viral expansions from diverse reservoirs. Cell Host Microbe. 2019;26(3):347–58.
    1. Wang Z, Gurule EE, Brennan TP, Gerold JM, Kwon KJ, Hosmane NN, et al. Expanded cellular clones carrying replication‐competent HIV‐1 persist, wax, and wane. Proc Natl Acad Sci U S A. 2018;115(11):E2575–84.

Source: PubMed

3
S'abonner