Anti-proliferative therapy for HIV cure: a compound interest approach

Daniel B Reeves, Elizabeth R Duke, Sean M Hughes, Martin Prlic, Florian Hladik, Joshua T Schiffer, Daniel B Reeves, Elizabeth R Duke, Sean M Hughes, Martin Prlic, Florian Hladik, Joshua T Schiffer

Abstract

In the era of antiretroviral therapy (ART), HIV-1 infection is no longer tantamount to early death. Yet the benefits of treatment are available only to those who can access, afford, and tolerate taking daily pills. True cure is challenged by HIV latency, the ability of chromosomally integrated virus to persist within memory CD4+ T cells in a non-replicative state and activate when ART is discontinued. Using a mathematical model of HIV dynamics, we demonstrate that treatment strategies offering modest but continual enhancement of reservoir clearance rates result in faster cure than abrupt, one-time reductions in reservoir size. We frame this concept in terms of compounding interest: small changes in interest rate drastically improve returns over time. On ART, latent cell proliferation rates are orders of magnitude larger than activation and new infection rates. Contingent on subtypes of cells that may make up the reservoir and their respective proliferation rates, our model predicts that coupling clinically available, anti-proliferative therapies with ART could result in functional cure within 2-10 years rather than several decades on ART alone.

Conflict of interest statement

The authors declare that they have no competing interests.

Figures

Figure 1
Figure 1
Schematics of models for HIV dynamics on and off ART. The top panel shows all possible transitions in the model (equation (1)). The bottom shaded panel shows the available transitions for the decoupled dynamic equations when ART suppresses the virus. Model parameters are given in Table 1. HIV virus V infects susceptible cells S at rate β reduced by ART of efficacy ε to βε. The probability of latency given infection is τ. The rate of activation from latently infected cells (L) to actively infected cells (A) is ξ. Cellular proliferation and death are determined by rates α and δ for each compartment. The mechanisms of action of anti-proliferative and latency reversal therapies are to decrease αL and increase ξ, respectively.
Figure 2
Figure 2
Simulated comparisons of latent reservoir eradication strategies on standard antiretroviral (ART) treatment. Treatment thresholds (discussed in Methods) are shown as solid black lines both in the plots and color bar, which is consistent between panels. (a) One-time therapeutic reductions of the latent pool (L0). (b) Continuous therapeutic increases in the clearance rate (θL). Relatively small decreases in the clearance rate θL produce markedly faster times to cure than much larger decreases in the initial reservoir size. (ce) Latency reversal agent (LRA) and anti-proliferative (AP) therapies are given continuously for durations of weeks with potencies given in fold increase in activation rate (εLRA) and fold decrease in proliferation rate (εAP), respectively. The color bar is consistent between panels, and thresholds of cure are shown as solid black lines both on plots and on the color bar. (c) Latency reversing agent therapy (LRA) administered alone requires years and potencies above 100 to achieve the cure thresholds. (d) Anti-proliferative therapies (AP) administered alone lead to cure thresholds in 1–2 years provided potency is greater than 2–3. (e) LRA and AP therapies are administered concurrently, and the reduction in the latent pool is measured at 70 weeks. Because the proliferation rate is naturally greater than the activation rate, increasing the AP potency has a much stronger effect than increasing the LRA potency.
Figure 3
Figure 3
Simulated comparisons of anti-proliferative therapies on standard antiretroviral therapy (ART) assuming variable reservoir composition. Proliferation and death rates in Table 1. The potency of the therapy is εAP = 10 (i.e., each cell type i has proliferation rate equal to αi/10 with i∈[em,cm,n]). Plausible initial compositions of the reservoir (Li(0)) are taken from experimental measurements, , . It is assumed that the HIV activation rate ξ is equivalent across all reservoir subsets. (ac) Plots of times to therapeutic landmarks on long-term ART and anti-proliferative therapy with heterogeneous reservoir compositions consisting of effector memory (Tem), central memory (Tcm), and naïve plus stem cell-like memory (Tn + Tscm) CD4+ T cells. Tem and Tn + Tscm percentages are shown with the remaining cells representing Tcm. Times to one-year remission and functional cure are extremely sensitive to percentage of Tn + Tscm but not percentage of Tem. (d,e) Continuous 10-fold therapeutic decreases in all proliferation rates (αi) result in Hill 1-yr in (d) 3.5 years assuming Tn + Tscm = 1% and (e) 6 years assuming Tn + Tscm = 10%. The reservoir is predicted to become Tn + Tscm dominant within 2 years under both assumptions, providing an indicator to gauge the success of anti-proliferative therapy in potential experiments.
Figure 4
Figure 4
Global sensitivity analysis. We use the ranges of parameters from Supplementary Table S4. (a) 1,000 simulations drawn from Latin Hypercube sample parameter sets where R0ART<1 are shown to demonstrate the variability of latent pool dynamics with respect to all combinations of parameter ranges. (b) The time until each cure threshold, Pinkevych 1-yr (P1) and Hill cure (Hc), are calculated as the time when the latent reservoir contains fewer than 20,000 and 200 cells respectively. In some cases cures are achieved within months. In others, cure requires many years. (c) Pearson correlation coefficients indicate the correlations between each variable and time to cure. L0 is the initial number of latent cells. Ln(0)/L0 is the initial fraction of naïve cells in the latent pool. τ is the probability of latency given infection. R0ART is the basic reproductive number on ART. εART is the percent decrease in viral infectivity in the presence of ART. θL is the decay rate of latent cells. εAP is the fold reduction in proliferation rate.
Figure 5
Figure 5
Waning anti-proliferative potency over-time modulates cure. Latent reservoir dynamics on combined ART and anti-proliferative therapy simulated for waning potency of anti-proliferative therapy over time. The latent reservoir size is shown with horizontal black lines corresponding to the cure threshholds used throughout the paper. Cure thresholds are achieved within 10 years if potency decreases by less than 5% per month considering 1% naïve T cells (Ln(0)/L0 = 0.01) and initial anti-proliferative potency εAP = 5.
Figure 6
Figure 6
MMF pharmacodynamics. Pure mycophenolic acid (MPA) was added to CEM cells at varying concentrations and proliferation of CEM cells was measured to determine a dose-response curve and Hill slope. CD4+ T cells from stored peripheral blood mononuclear cell samples from 10 participants (4 HIV-infected, 6 HIV-uninfected) were stimulated to proliferate. CD4+ T cells from 3 HIV-negative subjects were sorted into effector memory (EM), central memory (CM), and naïve subsets. Pure MPA was added to these cells at varying concentrations in order to determine IC50s for MPA. (a) Dose-response curve with percentages of CEM cells proliferating at varying doses of MPA. The Hill slope is −3.7. (b) 4 samples from HIV-positive participants and 6 samples from HIV-negative participants had similar IC50s. (c) IC50s were similar among CD4+ effector memory (EM), central memory (CM), and naïve T cell subsets.

References

    1. Siliciano JD, et al. Long-term follow-up studies confirm the stability of the latent reservoir for HIV-1 in resting CD4+ T cells. Nat Med. 2003;9:727–728. doi: 10.1038/nm880.
    1. Finzi D, et al. Latent infection of CD4+ T cells provides a mechanism for lifelong persistence of HIV-1, even in patients on effective combination therapy. Nat Med. 1999;5:512–517. doi: 10.1038/8394.
    1. Martin A, Siliciano R. Progress toward HIV eradication: Case reports, current efforts, and the challenges associated with cure. Annu Rev Med. 2016;67:011514–023043. doi: 10.1146/annurev-med-011514-023043.
    1. Hütter G, et al. Long-term control of HIV by CCR5Δ32 stem-cell transplantation. N Engl J Med. 2009;360:692–698. doi: 10.1056/NEJMoa0802905.
    1. Bull ME, et al. Monotypic human immunodeficiency virus type 1 genotypes across the uterine cervix and in blood suggest proliferation of cells with provirus. J Virol. 2009;83:6020–6028. doi: 10.1128/JVI.02664-08.
    1. Jaafoura S, et al. Progressive contraction of the latent HIV reservoir around a core of less-differentiated CD4+ memory T cells. Nat Comm. 2014;5:5407. doi: 10.1038/ncomms6407.
    1. Palmer S, et al. Low-level viremia persists for at least 7 years in patients on suppressive antiretroviral therapy. Proc Natl Acad Sci USA. 2008;105:3879–3884. doi: 10.1073/pnas.0800050105.
    1. Von Stockenstrom S, et al. Longitudinal genetic characterization reveals that cell proliferation maintains a persistent HIV-1 DNA pool during effective HIV therapy. J Infect Dis. 2015;1:596–607. doi: 10.1093/infdis/jiv092.
    1. Wagner T, et al. An increasing proportion of monotypic HIV-1 DNA sequences during antiretroviral treatment suggests proliferation of HIV-infected cells. J Virol. 2013;87:1770–1778. doi: 10.1128/JVI.01985-12.
    1. Wagner TA, et al. Proliferation of cells with HIV integrated into cancer genes contributes to persistent infection. Science. 2014;345:570–573. doi: 10.1126/science.1256304.
    1. Maldarelli F, et al. HIV latency. Specific HIV integration sites are linked to clonal expansion and persistence of infected cells. Science. 2014;345:179–83. doi: 10.1126/science.1254194.
    1. Chomont N, et al. HIV reservoir size and persistence are driven by T cell survival and homeostatic proliferation. Nat Med. 2009;15:893–900. doi: 10.1038/nm.1972.
    1. Bui JK, et al. Proviruses with identical sequences comprise a large fraction of the replication-competent HIV reservoir. PLoS Path. 2017;13:e1006283. doi: 10.1371/journal.ppat.1006283.
    1. Bonhoeffer S, Coffin JM, Nowak MA. Human immunodeficiency virus drug therapy and virus load. J Virol. 1997;71:3275–3278.
    1. Callaway D, Perelson A. HIV-1 infection and low steady state viral loads. Bull Math Biol. 2002;64:29–64. doi: 10.1006/bulm.2001.0266.
    1. Shen L, et al. A critical subset model provides a conceptual basis for the high antiviral activity of major HIV drugs. Sci Transl Med. 2011;3:91ra63. doi: 10.1126/scitranslmed.3002304.
    1. Brodin J, et al. Establishment and stability of the latent HIV-1 DNA reservoir. eLife. 2016;5:e18889. doi: 10.7554/eLife.18889.
    1. Fuller DH, et al. Therapeutic DNA vaccine induces broad T cell responses in the gut and sustained protection from viral rebound and AIDS in SIV-infected rhesus macaques. PLoS One. 2012;7:e33715. doi: 10.1371/journal.pone.0033715.
    1. Aubert M, et al. Successful targeting and disruption of an integrated reporter lentivirus using the engineered homing endonuclease Y2 I-AniI. PLoS One. 2011;6:e16825. doi: 10.1371/journal.pone.0016825.
    1. Peterson C, Younan P, Jerome K, Kiem H-P. Combinatorial anti-HIV gene therapy: using a multipronged approach to reach beyond HAART. Gene Ther. 2013;20:695–702. doi: 10.1038/gt.2012.98.
    1. Hill A, Rosenbloom D, Fu F, Nowak M, Siliciano R. Predicting the outcomes of treatment to eradicate the latent reservoir for HIV-1. Proc Natl Acad Sci USA. 2014;111:15597. doi: 10.1073/pnas.1406663111.
    1. Pinkevych M, et al. HIV reactivation from latency after treatment interruption occurs on average every 5–8 days? Implications for HIV remission. PLoS Pathog. 2015;11:e1005000. doi: 10.1371/journal.ppat.1005000.
    1. Buzon MJ, et al. HIV-1 persistence in CD4+ T cells with stem cell-like properties. Nat Med. 2014;20:139–142. doi: 10.1038/nm.3445.
    1. Marino S, Hogue I, Ray C, Kirschner D. A methodology for performing global uncertainty and sensitivity analysis in systems biology. J Theor Biol. 2008;254:178–196. doi: 10.1016/j.jtbi.2008.04.011.
    1. Chapuis AG, et al. Effects of mycophenolic acid on human immunodeficiency virus infection in vitro and in vivo. Nat Med. 2000;6:762–768. doi: 10.1038/77489.
    1. Crooks AM, et al. Precise quantitation of the latent HIV-1 reservoir: implications for eradication strategies. J Infect Dis. 2015;212:1361–1365. doi: 10.1093/infdis/jiv218.
    1. Garca F, et al. Effect of mycophenolate mofetil on immune response and plasma and lymphatic tissue viral load during and after interruption of highly active antiretroviral therapy for patients with chronic HIV infection: a randomized pilot study. J Acquir Immune Defic Syndr. 2004;36:823–830. doi: 10.1097/00126334-200407010-00009.
    1. Foley GE, et al. Continuous culture of human lymphoblasts from peripheral blood of a child with acute leukemia. Cancer. 1965;18:522–529. doi: 10.1002/1097-0142(196504)18:4<522::AID-CNCR2820180418>;2-J.
    1. Conway J, Perelson A. Residual Viremia in Treated HIV+ Individuals. PLoS Comput Biol. 2016;12:e1004677. doi: 10.1371/journal.pcbi.1004677.
    1. Ho Y-C, et al. Replication-competent noninduced proviruses in the latent reservoir increase barrier to HIV-1 cure. Cell. 2013;155:540–551. doi: 10.1016/j.cell.2013.09.020.
    1. Mok C. Mycophenolate mofetil for lupus nephritis: an update. Expert Rev Clin Immunol. 2015;11:1353–1364. doi: 10.1586/1744666X.2015.1087314.
    1. Müller E, Barday Z, Mendelson M, Kahn D. HIV-positive–to–HIV-positive kidney transplantation—Results at 3 to 5 years. N Engl J Med. 2015;372:613–620. doi: 10.1056/NEJMoa1408896.
    1. Stock PG, et al. Outcomes of kidney transplantation in HIV-infected recipients. N Engl J Med. 2010;363:2004–2014. doi: 10.1056/NEJMoa1001197.
    1. Kaur R, et al. A placebo-controlled pilot study of intensification of antiretroviral therapy with mycophenolate mofetil. AIDS Res Ther. 2006;3:16. doi: 10.1186/1742-6405-3-16.
    1. Vrisekoop N, et al. Short communication: no detrimental immunological effects of mycophenolate mofetil and HAART in treatment-naive acute and chronic HIV-1-infected patients. AIDS Res Hum Retrovir. 2005;21:991–996. doi: 10.1089/aid.2005.21.991.
    1. Sankatsing SU, et al. Highly active antiretroviral therapy with or without mycophenolate mofetil in treatment-naive HIV-1 patients. AIDS. 2004;18:1925–1931. doi: 10.1097/00002030-200409240-00008.
    1. Press N, et al. Case series assessing the safety of mycophenolate as part of multidrug rescue treatment regimens. HIV Clin Trials. 2002;3:17–20. doi: 10.1310/B6T0-N98J-1J3M-EQPK.
    1. Margolis DM, et al. The addition of mycophenolate mofetil to antiretroviral therapy including abacavir is associated with depletion of intracellular deoxyguanosine triphosphate and a decrease in plasma HIV-1 RNA. J Acquir Immune Defic Syndr. 2002;31:45–49. doi: 10.1097/00126334-200209010-00006.
    1. Millan O, et al. Pharmacokinetics and pharmacodynamics of low dose mycophenolate mofetil in HIV-infected patients treated with abacavir, efavirenz and nelfinavir. Clin Pharmacokinet. 2005;44:525–538. doi: 10.2165/00003088-200544050-00006.
    1. Jurriaans S, et al. HIV-1 seroreversion in an HIV-1-seropositive patient treated during acute infection with highly active antiretroviral therapy and mycophenolate mofetil. AIDS. 2004;18:1607–1608. doi: 10.1097/01.aids.0000131367.05823.ce.
    1. Ross DM, et al. Safety and efficacy of imatinib cessation for CML patients with stable undetectable minimal residual disease: results from the TWISTER study. Blood. 2013;122:515–522. doi: 10.1182/blood-2013-02-483750.
    1. Hladik F. A new perspective on HIV cure. F1000Res. 2014;4:77.
    1. Hladik F, et al. Mucosal effects of tenofovir 1% gel. eLife. 2015;4:e04525.
    1. Rong L, Perelson A. Modeling latently infected cell activation: Viral and latent reservoir persistence, and viral blips in HIV-infected patients on potent therapy. PLoS Comput Biol. 2009;5:e1000533. doi: 10.1371/journal.pcbi.1000533.
    1. Perelson AS, Kirschner DE, De Boer R. Dynamics of HIV infection of CD4+ T cells. Math Biosci. 1993;114:81–125. doi: 10.1016/0025-5564(93)90043-A.
    1. Quah BJ, Parish CR. New and improved methods for measuring lymphocyte proliferation emphin vitro and in vivo using CFSE-like fluorescent dyes. J Immunol Methods. 2012;379:1–14. doi: 10.1016/j.jim.2012.02.012.
    1. Ritz, C., Baty, F., Streibig, J. C. & Gerhard, D. Dose-response analysis using R. PLoS One10, e0146021, journal.pone.0146021 (2015).
    1. R Core Team. R: A Language and Environment for Statistical Computing. R Foundation for Statistical Computing, Vienna, Austria (2016).
    1. Macallan DC, et al. Rapid turnover of effector-memory CD4+ T cells in healthy humans. J Exp Med. 2004;200:255–260. doi: 10.1084/jem.20040341.
    1. Markowitz M, et al. A novel antiviral intervention results in more accurate assessment of HIV-1 replication dynamics and T-cell decay in vivo. J Virol. 2003;77:5037–5038. doi: 10.1128/JVI.77.8.5037-5038.2003.
    1. Luo R, Piovoso M, Martinez-Picado J, Zurakowski R. HIV model parameter estimates from interruption trial data including drug efficacy and reservoir dynamics. PLoS One. 2012;7:e40198. doi: 10.1371/journal.pone.0040198.
    1. Hockett RD, et al. Constant mean viral copy number per infected cell in tissues regardless of high, low, or undetectable plasma HIV RNA. J Exp Med. 1999;189:1545–1554. doi: 10.1084/jem.189.10.1545.
    1. Ramratnam B, et al. Rapid production and clearance of HIV-1 and hepatitis C virus assessed by large volume plasma apheresis. Lancet. 1999;354:1782–1785. doi: 10.1016/S0140-6736(99)02035-8.

Source: PubMed

3
S'abonner