Effects of recombinant human interleukin 7 on T-cell recovery and thymic output in HIV-infected patients receiving antiretroviral therapy: results of a phase I/IIa randomized, placebo-controlled, multicenter study

Y Lévy, I Sereti, G Tambussi, J P Routy, J D Lelièvre, J F Delfraissy, J M Molina, M Fischl, C Goujard, B Rodriguez, C Rouzioux, V Avettand-Fenoël, T Croughs, S Beq, M Morre, J F Poulin, R P Sekaly, R Thiebaut, M M Lederman, Y Lévy, I Sereti, G Tambussi, J P Routy, J D Lelièvre, J F Delfraissy, J M Molina, M Fischl, C Goujard, B Rodriguez, C Rouzioux, V Avettand-Fenoël, T Croughs, S Beq, M Morre, J F Poulin, R P Sekaly, R Thiebaut, M M Lederman

Abstract

Background: The immune deficiency of human immunodeficiency virus (HIV) infection is not fully corrected with ARV therapy. Interleukin-7 (IL-7) can boost CD4 T-cell counts, but optimal dosing and mechanisms of cellular increases need to be defined.

Methods: We performed a randomized placebo-controlled dose escalation (10, 20 and 30 µg/kg) trial of 3 weekly doses of recombinant human IL-7 (rhIL-7) in ARV-treated HIV-infected persons with CD4 T-cell counts between 101 and 400 cells/µL and plasma HIV levels <50 copies/mL. Toxicity, activity and the impact of rhIL-7 on immune reconstitution were monitored.

Results: Doses of rhIL-7 up to 20 µg/kg were well tolerated. CD4 increases of predominantly naive and central memory T cells were brisk (averaging 323 cells/µL at 12 weeks) and durable (up to 1 year). Increased cell cycling and transient increased bcl-2 expression were noted. Expanded cells did not have the characteristics of regulatory or activated T cells. Transient low-level HIV viremia was seen in 6 of 26 treated patients; modest increases in total levels of intracellular HIV DNA were proportional to CD4 T-cell expansions. IL-7 seemed to increase thymic output and tended to improve the T-cell receptor (TCR) repertoire in persons with low TCR diversity.

Conclusions: Three weekly doses of rhIL-7 at 20 µg/kg are well tolerated and lead to a dose-dependent CD4 T-cell increase and the broadening of TCR diversity in some subjects. These data suggest that this rhIL-7 dose could be advanced in future rhIL-7 clinical studies.

Clinical trials registration: NCT0047732.

Figures

Figure 1.
Figure 1.
Changes in T-cell counts after recombinant human interleukin 7 (rhIL-7) administration. A, B, CD4 (A) and CD8 (B) T-cell counts (median, Quarter 1–Quarter 3) increased significantly at week 12 (day 77) after a rhIL-7 cycle (1 injection weekly for 3 weeks) (P < .002, representing changes from baseline for each dose group compared with placebo). The increase was dose-dependent for both CD4 and CD8 T cells and remained significant at week 52 (P < .005 and P < .01 for CD4 and CD8, respectively). CF, Naive and central memory CD4 and CD8 T-cell subsets increased after rhIL-7 administration (P < .05 represents comparisons between the 20 and 30 µg/kg dose groups and placebo group by Wilcoxon rank sum test). G, Administration of rhIL-7 increased absolute counts of regulatory T cells (Tregs), but their frequency was significantly decreased. Abbreviations: CM, central memory; N, naive.
Figure 2.
Figure 2.
Cycling of CD4 and CD8 T-cell subsets in response to recombinant human interleukin 7 (rhIL-7). A, Flow cytometry was used to determine Ki-67 expression in T-cell subsets. Increased naive, central memory (CM), and effector memory T-cell cycling was seen on day 7 (pooled rhIL-7 groups compared with placebo, P < .001), returning to baseline by day 28 (median, Quarter 1–Quarter 3). The increase was not dose dependent and occurred in both CD4 and CD8 subsets. B, PD-1 expression decreased on both CD4 and CD8 T cells 28 days after 20 and 30 µg/kg rhIL-7 administration (P < .05 for each). The decrease persisted until day 77 (P < .05 ). C, CM subsets showed decreased expression of PD-1. Abbreviations: CM, central memory; EM, effector memory; N, naive.
Figure 3.
Figure 3.
Recombinant human interleukin 7 (rhIL-7) therapy increases numbers of naive and recent thymic emigrants (RTE) CD4 T cells and broadens the T-cell receptor (TCR) repertoire in a few subjects. Data shown are for subjects who received 3 doses of rhIL-7 at 20 or 30 µg/kg. A, Thymic activity was evaluated using the sj/β T-cell receptor excision circles (TREC) ratio. Treatment with rhIL-7 increased the number of naive and RTE cells in the peripheral blood on day 77 (P < .05, 20 and 30 µg/kg pooled data). The sj/β TREC ratio tended to increase (P = .06, pooled data for 20 and 30 µg/kg). B, The CD4 T-cell count at day 0 and CD4 nadir were associated with a change in the sj/β TREC ratio between days 0 and 77 (P = .01; Spearman's test). C, Subjects with lower CD4 T-cell counts experienced greater increases in thymopoiesis after rhIL-7 treatment. TCR repertoire diversity was assessed by quantifying all possible combinations between Vβ and Jβ segments. Shaded area represents range of diversity in historical controls. Diversity increased in all rhIL-7–treated subjects with diversity below the normal range but not significantly for the treated groups overall. D, Distribution of peaks for each Vβ-Jβ rearrangement for a placebo-treated and a rhIL-7–treated patient. Abbreviations: RTE, recent thymic emigrant; TREC, T-cell reception excision circle.

References

    1. von Freeden-Jeffry U, Vieira P, Lucian LA, McNeil T, Burdach SE, Murray R. Lymphopenia in interleukin (IL)-7 gene-deleted mice identifies IL-7 as a nonredundant cytokine. J Exp Med. 1995;181:1519–26.
    1. Beq S, Delfraissy JF, Theze J. Interleukin-7 (IL-7): immune function, involvement in the pathogenesis of HIV infection and therapeutic potential. Euro Cytokine Netw. 2004;15:279–89.
    1. Puel A, Ziegler SF, Buckley RH, Leonard WJ. Defective IL7R expression in T-B+NK+ severe combined immunodeficiency. Nat Gene. 1998;20:394–7.
    1. Noguchi M, Nakamura Y, Russell SM, et al. Interleukin-2 receptor gamma chain: a functional component of the interleukin-7 receptor. Science. 1993;262:1877–80.
    1. Bolotin E, Annett G, Parkman R, Weinberg K. Serum levels of IL-7 in bone marrow transplant recipients: relationship to clinical characteristics and lymphocyte count. Bone Marrow Transplant. 1999;23:783–8.
    1. Napolitano LA, Grant RM, Deeks SG, et al. Increased production of IL-7 accompanies HIV-1-mediated T-cell depletion: implications for T-cell homeostasis. Nat Med. 2001;7:73–9.
    1. Beq S, Nugeyre MT, Ho Tsong Fang R, et al. IL-7 induces immunological improvement in SIV-infected rhesus macaques under antiviral therapy. J Immunol. 2006;176:914–22.
    1. Fry TJ, Moniuszko M, Creekmore S, et al. IL-7 therapy dramatically alters peripheral T-cell homeostasis in normal and SIV-infected nonhuman primates. Blood. 2003;101:2294–9.
    1. Mackall CL, Fry TJ, Bare C, Morgan P, Galbraith A, Gress RE. IL-7 increases both thymic-dependent and thymic-independent T-cell regeneration after bone marrow transplantation. Blood. 2001;97:1491–7.
    1. Storek J, Gillespy T, 3rd, Lu H, et al. Interleukin-7 improves CD4 T-cell reconstitution after autologous CD34 cell transplantation in monkeys. Blood. 2003;101:4209–18.
    1. Beq S, Rozlan S, Gautier D, et al. Injection of glycosylated recombinant simian IL-7 provokes rapid and massive T-cell homing in rhesus macaques. Blood. 2009;114:816–25.
    1. Levy Y, Lacabaratz C, Weiss L, et al. Enhanced T cell recovery in HIV-1-infected adults through IL-7 treatment. J Clin Invest. 2009;119:997–1007.
    1. Rosenberg SA, Sportes C, Ahmadzadeh M, et al. IL-7 administration to humans leads to expansion of CD8+ and CD4+ cells but a relative decrease of CD4+ T-regulatory cells. J Immunother. 2006;29:313–9.
    1. Sereti I. Where have all the T cells gone? Blood. 2009;114:751–2.
    1. Sportes C, Hakim FT, Memon SA, et al. Administration of rhIL-7 in humans increases in vivo TCR repertoire diversity by preferential expansion of naive T cell subsets. J Exp Med. 2008;205:1701–14.
    1. Lelievre JD, Levy Y. Perspectives on interleukin-7 therapy in HIV infection. Curr Opin HIV AIDS. 2007;2:228–33.
    1. Mackall CL, Fry TJ, Gress RE. Harnessing the biology of IL-7 for therapeutic application. Nat Rev Immunol. 2011;11:330–42.
    1. Dion ML, Sekaly RP, Cheynier R. Estimating thymic function through quantification of T-cell receptor excision circles. Methods Mol Biol. 2007;380:197–213.
    1. Marodon G, Desjardins D, Mercey L, et al. High diversity of the immune repertoire in humanized NOD.SCID.gamma c-/- mice. Eur J Immunol. 2009;39:2136–45.
    1. Pasqual N, Gallagher M, Aude-Garcia C, et al. Quantitative and qualitative changes in V-J alpha rearrangements during mouse thymocyte differentiation: implication for a limited T cell receptor alpha chain repertoire. J Exp Med. 2002;196:1163–73.
    1. Avettand-Fenoel V, Chaix ML, Blanche S, et al. LTR real-time PCR for HIV-1 DNA quantitation in blood cells for early diagnosis in infants born to seropositive mothers treated in HAART area (ANRS CO 01) J Med Virol. 2009;81:217–23.
    1. Abrams D, Levy Y, Losso MH, et al. Interleukin-2 therapy in patients with HIV infection. Engl J Med. 2009;361:1548–59.
    1. Sereti I, Imamichi H, Natarajan V, et al. In vivo expansion of CD4CD45RO-CD25 T cells expressing foxP3 in IL-2-treated HIV-infected patients. J Clin Invest. 2005;115:1839–47.
    1. Weiss L, Letimier FA, Carriere M, et al. In vivo expansion of naive and activated CD4+CD25+FOXP3+ regulatory T cell populations in interleukin-2-treated HIV patients. Proc Natl Acad Sci U S A. 2010;107:10632–7.
    1. Said EA, Dupuy FP, Trautmann L, et al. Programmed death-1-induced interleukin-10 production by monocytes impairs CD4+ T cell activation during HIV infection. Nat Med. 2010;16:452–9.
    1. Sereti I, Dunham RM, Spritzler J, et al. IL-7 administration drives T cell-cycle entry and expansion in HIV-1 infection. Blood. 2009;113:6304–14.
    1. Sieg SF, Harding CV, Lederman MM. HIV-1 infection impairs cell cycle progression of CD4+ T cells without affecting early activation responses. J Clin Invest. 2001;108:757–64.
    1. Sereti I, Anthony KB, Martinez-Wilson H, et al. IL-2-induced CD4+ T-cell expansion in HIV-infected patients is associated with long-term decreases in T-cell proliferation. Blood. 2004;104:775–80.
    1. Kovacs JA, Lempicki RA, Sidorov IA, et al. Induction of prolonged survival of CD4+ T lymphocytes by intermittent IL-2 therapy in HIV-infected patients. J Clin Invest. 2005;115:2139–48.
    1. Chomont N, El-Far M, Ancuta P, et al. HIV reservoir size and persistence are driven by T cell survival and homeostatic proliferation. Nat Med. 2009;15:893–900.
    1. Bruyand M, Thiebaut R, Lawson-Ayayi S, et al. Role of uncontrolled HIV RNA level and immunodeficiency in the occurrence of malignancy in HIV-infected patients during the combination antiretroviral therapy era: Agence Nationale de Recherche sur le Sida (ANRS) CO3 Aquitaine Cohort. Clin Infect Dis. 2009;49:1109–16.
    1. Guiguet M, Boue F, Cadranel J, Lang JM, Rosenthal E, Costagliola D. Effect of immunodeficiency, HIV viral load, and antiretroviral therapy on the risk of individual malignancies (FHDH-ANRS CO4): a prospective cohort study. Lancet Oncol. 2009;10:1152–9.
    1. Lewden C, Chene G, Morlat P, et al. HIV-infected adults with a CD4 cell count greater than 500 cells/mm3 on long-term combination antiretroviral therapy reach same mortality rates as the general population. J Acquir Immune Defic Syndr. 2007;46:72–7.

Source: PubMed

3
Prenumerera