Extensive characterization of HIV-1 reservoirs reveals links to plasma viremia before and during analytical treatment interruption

Basiel Cole, Laurens Lambrechts, Zoe Boyer, Ytse Noppe, Marie-Angélique De Scheerder, John-Sebastian Eden, Bram Vrancken, Timothy E Schlub, Sherry McLaughlin, Lisa M Frenkel, Sarah Palmer, Linos Vandekerckhove, Basiel Cole, Laurens Lambrechts, Zoe Boyer, Ytse Noppe, Marie-Angélique De Scheerder, John-Sebastian Eden, Bram Vrancken, Timothy E Schlub, Sherry McLaughlin, Lisa M Frenkel, Sarah Palmer, Linos Vandekerckhove

Abstract

The HIV-1 reservoir is composed of cells harboring latent proviruses that have the potential to contribute to viremia upon antiretroviral treatment (ART) interruption. While this reservoir is known to be maintained by clonal expansion of infected cells, the contribution of these cell clones to residual viremia and viral rebound remains underexplored. Here, we conducted an extensive analysis on four ART-treated individuals who underwent an analytical treatment interruption (ATI), characterizing the proviral genomes and associated integration sites of large infected clones and phylogenetically linking these to plasma viremia. We show discrepancies between different assays in their ability to assess clonal expansion. Furthermore, we demonstrate that proviruses could phylogenetically be linked to plasma virus obtained before or during an ATI. This study highlights a role for HIV-infected cell clones in the maintenance of the replication-competent reservoir and suggests that infected cell clones can directly contribute to rebound viremia upon ATI.

Trial registration: ClinicalTrials.gov NCT02641756.

Keywords: CP: Microbiology; HIV; analytical treatment interruption; antiretroviral therapy; cellular proliferation; clonal expansion; full-length HIV sequencing; integration site sequencing; latency; rebound; replication competency.

Conflict of interest statement

Declaration of interests The authors declare that no conflict of interest exists.

Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.

Figures

Figure 1.. Overview of the workflow for…
Figure 1.. Overview of the workflow for HIV-1 reservoir characterization and viral loads at each time point of sample collection
(A) Workflow of HIV-1 reservoir characterization by single-genome sequencing (SGS), full-length individual proviral sequencing (FLIPS), integration site loop amplification (ISLA), and multiple displacement amplification (MDA). In a first step, potentially clonal HIV-1-infected cells were identified by SGS, FLIPS, and ISLA on lysed sorted CD4 T cell subsets. In a second step, MDA with subsequent SGS and ISLA was performed on selected sorted cell lysates. In the final step, MDA reactions containing a potentially clonal provirus were identified, and the near full-length (NFL) genome of the according provirus was amplified and sequenced. (B) Viral load (copies/mL) at each time point of sample collection for all participants. The day of analytical treatment interruption (ATI) initiation is indicated with a vertical red line. The plasma was sampled during ART (time point 1, T1), 8 to 14 days after ATI (time point 2, T2), at the first detectable viral load (time point 3, T3), and at later rebound (time point 4, T4). Note that T1 is not shown to scale. The horizontal dashed lines indicate the limit of detection at 20 copies/mL. See also Figure S4 and Tables S1, S6, and S7.
Figure 2.. Classification of HIV-1 integration sites…
Figure 2.. Classification of HIV-1 integration sites and proviral near full-length genome sequences across different cell subsets before ATI
(A) Proportions of integration sites (IS) retrieved by integration site loop amplification (ISLA) for participants STAR 9, STAR 10, and STAR 11 from TCM/TTM and TEM subsets from peripheral blood. IS found more than once are defined as “clonal” and are shown in color as proportion of all IS. Identical IS found in both subsets are linked with dashed lines. p values test was used for a difference in the proportion of unique IS between TCM/TTM and TEM by “prop.test” in R. (B) Proportions of intact and defective near full-length sequences from full-length individual proviral sequencing (FLIPS) within TCM/TTM and TEM peripheral blood for each participant. See Figures S1 and S2A for figures including all FLIPS data. (C) Proportions of expansions of identical sequences (EIS) found in TCM/TTM and TEM peripheral blood subsets based on FLIPS data for each participant. EIS consisting of a detective and intact provirus are shown in shades of red and green, respectively, while unique proviruses are represented in white. The “prop.test” in R was used to test for differences in the proportion of unique proviruses between TCM/TTM and TEM. TCM/TTM, central/transitional memory CD4 T cell; TEM, effector memory CD4 T cell; PSI, packaging signal; MSD, major splice donor. See also Figures S1 and S2 and Tables S2 and S4.
Figure 3.. Near full-length proviral HIV-1 genomes…
Figure 3.. Near full-length proviral HIV-1 genomes and associated integration sites recovered from the peripheral blood by MDA
(A) For each participant, the recovered proviral genome structures are shown aligned to the HXB2 reference genome, and corresponding integration sites are listed on the right. For more information, see Table S3. Each provirus is colored according to their structural category. (B–D) For each participant, the number of integration site (IS) and near full-length (NFL) proviruses generated via full-length individual proviral sequencing (FLIPS) linked to clones characterized by multiple displacement amplification (MDA) is shown together with their corresponding cellular subset. TCM/TTM, central/transitional memory CD4 T cell; TEM, effector memory CD4 T cell; PSI, packaging signal; MSD, major splice donor. See also Table S3.
Figure 4.. Comparison of assays to identify…
Figure 4.. Comparison of assays to identify potentially clonal HIV-1-infected cell populations
Clonality assessments are shown for integration site loop amplification (ISLA), single-genome sequencing (SGS), and full-length individual proviral sequencing (FLIPS). Identical sequences found multiple times within the same assay are shown as light gray slices. The total number of examined integration sites (IS), V1–V3 env sequences, and near full-length (NFL) proviral sequences is noted in the middle of each donut plot. When an IS, V1–V3 env SGS, or NFL FLIPS sequence could be unambiguously linked to data from previously identified infected cell clones by multiple displacement amplification (MDA), they were given a distinct standout color and chromosome designation as indicated in the legend. Dark gray slices show identical sequences that did not allow for overlap detection as (1) not all IS could be linked via MDA to a V1–V3 env or NFL sequence and (2) not all NFL genomes contained the V1–V3 env region. Arrows are used to indicate discrepancies between the different assays. EIS, expansion of identical sequences. See also Table S5.
Figure 5.. Maximum-likelihood phylogenetic trees based on…
Figure 5.. Maximum-likelihood phylogenetic trees based on the V1–V3 env region from proviral and plasma sequences before and during ATI
Proviral sequences collected before ATI from full-length individual proviral sequencing (FLIPS) and multiple displacement amplification (MDA) are shown as squares and circles, respectively. The integration sites (IS) associated with MDA-derived proviruses are noted if available. Plasma sequences are shown as triangles (V1–V3 env) or diamonds (3′-half genome) where the color indicates the time point during analytical treatment interruption (ATI). Arrows indicate identical matches between proviral and plasma V1–V3 env sequences. All trees are rooted to the HXB2 reference sequence. (A) In participant STAR 10, three identical matches between defective proviral and plasma rebound sequences were found. For two, the corresponding IS ZBTB20 and Chr8:100792121 could be recovered. (B) In participant STAR 4, only one match between a unique major splice donor (MSD) deleted provirus and plasma sequences was observed. (C) In STAR 9, a match between a unique intact provirus and multiple plasma sequences from different time points were found. (D) In STAR 11, a rebounding plasma sequence could be linked to an expansion of identical intact near full-length (NFL) genomes located in the ZNF141 gene. One unique intact provirus can be linked to a residual plasma sequence from T1. SGS, single-genome sequencing; PSI, packaging signal. See also Figure S3.

References

    1. Aamer HA, Mcclure J, Ko D, Maenza J, Collier AC, Mullins JI, and Frenkel LM (2020). Cells producing residual viremia during antiretroviral treatment appear to contribute to rebound viremia following interruption of treatment. PLoS Pathogens 16, e1008791.
    1. Artesi M, Hahaut V, Cole B, Lambrechts L, Ashrafi F, Marçais A, Hermine O, Griebel P, Arsic N, van der Meer F, et al. (2021). PCIP-seq: simultaneous sequencing of integrated viral genomes and their insertion sites with long reads. Genome Biol. 22, 97.
    1. Bailey JR, Sedaghat AR, Kieffer T, Brennan T, Lee PK, Wind-rotolo M, Haggerty CM, Kamireddi AR, Liu Y, Lee J, et al. (2006). Residual human immunodeficiency virus type 1 viremia in some patients on antiretroviral therapy is dominated by a small number of invariant clones rarely found in circulating CD4+ T cells. J. Virol. 80, 6441–6457.
    1. Bertagnolli LN, Varriale J, Sweet S, Brockhurst J, Simonetti FR, White J, Beg S, Lynn K, Mounzer K, Frank I, et al. (2020). Autologous IgG antibodies block outgrowth of a substantial but variable fraction of viruses in the latent reservoir for HIV-1. Proc. Natl. Acad. Sci. U S A 117, 32066–32077.
    1. Boritz EA, Darko S, Swaszek L, Wolf G, Wells D, Wu X, Henry AR, Laboune F, Hu J, Ambrozak D, et al. (2016). Multiple origins of virus persistence during natural control of HIV infection. Cell 166, 1004–1015.
    1. Brennan TP, Woods JO, Sedaghat AR, Siliciano JD, Siliciano RF, and Wilke CO (2009). Analysis of human immunodeficiency virus type 1 viremia and provirus in resting CD4+ T cells reveals a novel source of residual viremia in patients on antiretroviral therapy. J. Virol. 83, 8470–8481.
    1. Cesana D, Santoni de Sio FR, Rudilosso L, Gallina P, Calabria A, Beretta S, Merelli I, Bruzzesi E, Passerini L, Nozza S, et al. (2017). HIV-1-mediated insertional activation of STAT5B and BACH2 trigger viral reservoir in T regulatory cells. Nat. Commun. 8, 498.
    1. Chun T, and Fauci AS (1999). Perspective Latent reservoirs of HIV: Obstacles to the eradication of virus. Proc. Natl. Acad. Sci. U S A 96, 10958–10961.
    1. Chun TW, Engel D, Berrey MM, Shea T, Corey L, and Fauci AS (1998). Early establishment of a pool of latently infected, resting CD4(+) T cells during primary HIV-1 infection. Proc. Natl. Acad. Sci. U S A 95, 8869–8873.
    1. Chun TW, Stuyver L, Mizell SB, Ehler LA, Mican JAM, Baseler M, Lloyd AL, Nowak MA, and Fauci AS (1997). Presence of an inducible HIV-1 latent reservoir during highly active antiretroviral therapy. Proc. Natl. Acad. Sci. U S A 94, 13193–13197.
    1. Clarridge KE, Blazkova J, Einkauf K, Petrone M, Refsland W, Justement JS, Shi V, Huiting ED, Seamon CA, Lee GQ, et al. (2018). Effect of analytical treatment interruption and reinitiation of antiretroviral therapy on HIV reservoirs and immunologic parameters in infected individuals. PLoS Pathogens 14, e1006792.
    1. Cohen YZ, Lorenzi JCC, Krassnig L, Barton JP, Burke L, Pai J, Lu CL, Mendoza P, Oliveira TY, Sleckman C, et al. (2018). Relationship between latent and rebound viruses in a clinical trial of anti-HIV-1 antibody 3BNC117. J. Exp. Med. 215, 2311–2324.
    1. Cohn LB, Silva IT, Oliveira TY, Rosales RA, Parrish EH, Learn GH, Hahn BH, Czartoski JL, McElrath MJ, Lehmann C, et al. (2015). HIV-1 integration landscape during latent and active infection. Cell 160, 420–432.
    1. Cole B, Lambrechts L, Gantner P, Noppe Y, Bonine N, Witkowski W, Chen L, Palmer S, Mullins JI, Chomont N, et al. (2021). In-depth single-cell analysis of translation-competent HIV-1 reservoirs identifies cellular sources of plasma viremia. Nat. Commun. 12, 3727.
    1. De Scheerder M-A, Vrancken B, Dellicour S, Schlub T, Lee E, Shao W, Rutsaert S, Verhofstede C, Kerre T, Malfait T, et al. (2019). HIV rebound is predominantly fueled by genetically identical viral expansions from diverse reservoirs. Cell Host Microbe 26, 347–358.
    1. Einkauf KB, Lee GQ, Gao C, Sharaf R, Sun X, Hua S, Chen SM, Jiang C, Lian X, Chowdhury FZ, et al. (2019). Intact HIV-1 proviruses accumulate at distinct chromosomal positions during prolonged antiretroviral therapy. J. Clin. Invest. 129, 988–998.
    1. Estes JD, Kityo C, Ssali F, Swainson L, Makamdop KN, Del Prete GQ, Deeks SG, Luciw PA, Chipman JG, Beilman GJ, et al. (2017). Defining total-body AIDS-virus burden with implications for curative strategies. Nat. Med. 23, 1271–1276.
    1. Finzi D, Hermankova M, Pierson T, Carruth LM, Buck C, Chaisson RE, Quinn TC, Chadwick K, Margolick J, Brookmeyer R, et al. (1997). Identification of a reservoir for HIV-1 in patients on highly active antiretroviral therapy. Science 278, 1295–1300.
    1. Garner SA, Rennie S, Ananworanich J, Dube K, Margolis DM, Sugarman J, Tressler R, Gilbertson A, and Dawson L (2017). Interrupting antiretroviral treatment in HIV cure research: scientific and ethical considerations. J. Virus Eradication 3, 82–84.
    1. Guindon S, Dufayard J-F, Lefort V, Anisimova M, Hordijk W, and Gascuel O (2010). New algorithms and methods to estimate maximum-likelihood phylogenies: assessing the performance of PhyML 3.0. Syst. Biol. 59, 307–321.
    1. Halvas EK, Hughes SH, Mellors JW, Halvas EK, Joseph KW, Brandt LD, Guo S, Sobolewski MD, Jacobs JL, Tumiotto C, et al. (2020). HIV-1 viremia not suppressible by antiretroviral therapy can originate from large T cell clones producing infectious virus. J. Clin. Invest. 130, 5847–5857.
    1. Hiener B, Horsburgh BA, Eden JS, Barton K, Schlub TE, Lee E, von Stockenstrom S, Odevall L, Milush JM, Liegler T, et al. (2017). Identification of genetically intact HIV-1 proviruses in specific CD4+T cells from effectively treated participants. Cell Rep. 21, 813–822.
    1. Hosmane NN, Kwon KJ, Bruner KM, Capoferri AA, Beg S, Rosenbloom DIS, Keele BF, Ho Y-C, Siliciano JD, and Siliciano RF (2017). Proliferation of latently infected CD4 + T cells carrying replication-competent HIV-1: potential role in latent reservoir dynamics. J. Exp. Med. 214, 959–972.
    1. Huang AS, Ramos V, Oliveira TY, Gaebler C, Jankovic M, Nussenzweig MC, and Cohn LB (2021). Integration features of intact latent HIV-1 in CD4+ T cell clones contribute to viral persistence. J. Exp. Med. 218, e20211427.
    1. Imamichi H, Smith M, Adelsberger JW, Izumi T, Scrimieri F, Sherman BT, Rehm CA, Imamichi T, Pau A, Catalfamo M, et al. (2020). Defective HIV-1 proviruses produce viral proteins. Proc. Natl. Acad. Sci. U S A 117, 3704–3710.
    1. Jiang C, Lian X, Gao C, Sun X, Einkauf KB, Chevalier JM, Chen SMY, Hua S, Rhee B, Chang K, et al. (2020). Distinct viral reservoirs in individuals with spontaneous control of HIV-1. Nature 585, 261–267.
    1. John F, and Sanford W (2019). An R Companion to Applied Regression (Sage; ).
    1. Josefsson L, Eriksson S, Sinclair E, Ho T, Killian M, Epling L, Shao W, Lewis B, Bacchetti P, Loeb L, et al. (2012). Hematopoietic precursor cells isolated from patients on long-term suppressive HIV therapy did not contain HIV-1 DNA. J. Infect. Dis. 206, 28–34.
    1. Josefsson L, von Stockenstrom S, Faria NR, Sinclair E, Bacchetti P, Killian M, Epling L, Tan A, Ho T, Lemey P, et al. (2013). 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 110, E4987–E4996.
    1. Katoh K, Misawa K, Kuma K, and Miyata T (2002). MAFFT: a novel method for rapid multiple sequence alignment based on fast Fourier transform. Nucleic Acids Res. 30, 3059–3066.
    1. Kearney MF, Wiegand A, Shao W, Coffin JM, Mellors JW, Lederman M, Gandhi RT, Keele BF, and Li JZ (2016). Origin of rebound plasma HIV Includes cells with identical proviruses that are Transcriptionally active before stopping of antiretroviral therapy. J. Virol. 90, 1369–1376.
    1. Kumar S, Stecher G, and Tamura K (2016). MEGA7: molecular evolutionary genetics analysis version 7.0 for bigger datasets. Mol. Biol. Evol. 33, 1870–1874.
    1. Lambrechts L, Cole B, Rutsaert S, Trypsteen W, and Vandekerckhove L (2020). Emerging PCR-based techniques to study HIV-1 reservoir persistence. Viruses 12, 1–12.
    1. Laskey SB, Pohlmeyer CW, Bruner KM, and Siliciano RF (2016). Evaluating clonal expansion of HIV-infected cells: Optimization of PCR strategies to predict clonality. PLoS Pathog. 12, e1005689.
    1. Lee GQ, Orlova-Fink N, Einkauf K, Chowdhury FZ, Sun X, Harrington S, Kuo H-H, Hua S, Chen H-R, Ouyang Z, et al. (2017). Clonal expansion of genome-intact HIV-1 in functionally polarized Th1 CD4+ T cells. J. Clin. Invest. 127, 2689–2696
    1. Letunic I, and Bork P (2019). Interactive Tree of Life (iTOL) v4: recent updates and new developments. Nucleic Acids Res. 47, W256–W259.
    1. Li D, Liu C-M, Luo R, Sadakane K, and Lam T-W (2015). MEGAHIT: an ultra-fast single-node solution for large and complex metagenomics assembly via succinct de Bruijn graph. Bioinformatics 31, 1674–1676.
    1. Liu R, Simonetti FR, and Ho Y (2020). The forces driving clonal expansion of the HIV-1 latent reservoir. Virol. J 17.
    1. Lu C, Pai JA, Nogueira L, Mendoza P, Gruell H, Oliveira TY, and Barton J (2018). Relationship between intact HIV-1 proviruses in circulating CD4 + T cells and rebound viruses emerging during treatment interruption. Proc. Natl. Acad. Sci. U S A 115, 11341–11348.
    1. Maldarelli F, Wu X, Su L, Simonetti FR, Shao W, Hill S, Spindler J, Ferris AL, Mellors JW, Kearney MF, et al. (2014). Specific HIV integration sites are linked to clonal expansion and persistence of infected cells. Science 345, 179–183.
    1. Mcmanus WR, Coffin JM, Kearney MF, Mcmanus WR, Bale MJ, Spindler J, Wiegand A, Musick A, Patro SC, Sobolewski MD, et al. (2019). HIV-1 in lymph nodes is maintained by cellular proliferation during antiretroviral therapy. J. Clin. Invest. 129, 4629–4642.
    1. Minin VN, Dorman KS, Fang F, and Suchard MA (2005). Dual multiple change-point model leads to more accurate recombination detection. Bioinformatics 21, 3034–3042.
    1. Pannus P, Rutsaert S, De Wit S, Allard SD, Vanham G, Cole B, Nescoi C, Aerts J, De Spiegelaere W, Tsoumanis A, et al. (2020). Rapid viral rebound after analytical treatment interruption in patients with very small HIV reservoir and minimal on-going viral transcription. J. Int. AIDS Soc. 23, e25453.
    1. Patro SC, Brandt LD, Bale MJ, Halvas EK, Joseph KW, Shao W, and Wu X (2019). Combined HIV-1 sequence and integration site analysis informs viral dynamics and allows reconstruction of replicating viral ancestors. Proc. Natl. Acad. Sci. U S A 116, 25891–25899.
    1. Peluso MJ, Laird GM, Deeks SG, Peluso MJ, Bacchetti P, Ritter KD, Beg S, Lai J, Martin JN, Hunt PW, et al. (2020). Differential decay of intact and defective proviral DNA in HIV-1 – infected individuals on suppressive antiretroviral therapy. JCI Insight 5, e132997.
    1. Pinzone MR, Vanbelzen DJ, Weissman S, Bertuccio MP, Cannon L, Venanzi-rullo E, Migueles S, Jones RB, Mota T, Joseph SB, et al. (2019). Longitudinal HIV sequencing reveals reservoir expression leading to decay which is obscured by clonal expansion. Nat. Commun. 10, 728.
    1. Pollack RA, Jones RB, Pertea M, Bruner KM, Martin AR, Thomas AS, Capoferri AA, Beg SA, Huang SH, Karandish S, et al. (2017). Defective HIV-1 proviruses are expressed and can Be recognized by cytotoxic T lymphocytes, which shape the proviral landscape. Cell Host Microbe 21, 494–506.
    1. R Core Team (2020). R: A language and environment for statistical computing (R Foundation for Statistical Computing; ).
    1. Rutsaert S, De Spiegelaere W, De Clercq L, and Vandekerckhove L (2019). Evaluation of HIV-1 reservoir levels as possible markers for virological failure during boosted darunavir monotherapy. J. Antimicrob. Chemother. 74, 3030–3034.
    1. Salantes DB, Tebas P, Bar KJ, Salantes DB, Zheng Y, Mampe F, Srivastava T, Beg S, Lai J, Li JZ, et al. (2018). HIV-1 latent reservoir size and diversity are stable following brief treatment interruption. J. Clin. Invest. 128, 3102–3115.
    1. Simonetti FR, Sobolewski MD, Fyne E, Shao W, Spindler J, Hattori J, Anderson EM, Watters SA, Hill S, Wu X, et al. (2016). Clonally expanded CD4+ T cells can produce infectious HIV-1 in vivo. Proc. Natl. Acad. Sci. U S A 113, 1883–1888.
    1. Tobin NH, Learn GH, Holte SE, Wang Y, Melvin AJ, McKernan JL, Pawluk DM, Mohan KM, Lewis PF, Mullins JI, et al. (2005). Evidence that low-level viremias during effective highly active antiretroviral therapy result from two processes: expression of archival virus and replication of virus. J. Virol. 79, 9625–9634.
    1. Trypsteen W, Vynck M, De Neve J, Bonczkowski P, Kiselinova M, Malatinkova E, Vervisch K, Thas O, Vandekerckhove L, and De Spiegelaere W (2015). ddpcRquant: threshold determination for single channel droplet digital PCR experiments. Anal. Bioanal. Chem. 407, 5827–5834.
    1. Vibholm LK, Lorenzi JCC, Pai JA, Cohen YZ, Oliveira TY, Barton JP, Garcia Noceda M, Lu C-L, Ablanedo-Terrazas Y, Del Rio Estrada PM, et al. (2019). Characterization of intact proviruses in blood and lymph node from HIV-infected individuals undergoing analytical treatment interruption. J. Virol. 93, e01920–18.
    1. Von Stockenstrom S, Odevall L, Lee E, Sinclair E, Bacchetti P, Killian M, Epling L, Shao W, Hoh R, Ho T, et al. (2015). Longitudinal genetic characterization reveals that cell proliferation maintains a persistent HIV Type 1 DNA pool during effective HIV therapy. J. Infect. Dis. 212, 596–607.
    1. Wagner TA, McKernan JL, Tobin NH, Tapia KA, Mullins JI, Frenkel M, and Frenkel LM (2013). An increasing proportion of monotypic HIV-1 DNA sequences during antiretroviral treatment suggests proliferation of HIV-infected cells. J. Virol. 87, 1770–1778.
    1. Wagner TA, McLaughlin S, Garg K, Cheung CYK, Larsen BB, Styrchak S, Huang HC, Edlefsen PT, Mullins JI, and Frenkel LM (2014). Proliferation of cells with HIV integrated into cancer genes contributes to persistent infection. Science 345, 570–573.
    1. Wang Z, Gurule EE, Brennan TP, Gerold JM, Kwon KJ, Hosmane NN, Kumar MR, Beg SA, Capoferri AA, Ray SC, et al. (2018). Expanded cellular clones carrying replication-competent HIV-1 persist, wax, and wane. Proc. Natl. Acad. Sci. U S A 115, E2575–E2584.

Source: PubMed

3
Iratkozz fel