hIL-7-hyFc, A Long-Acting IL-7, Increased Absolute Lymphocyte Count in Healthy Subjects

Sang Won Lee, Donghoon Choi, MinKyu Heo, Eui-Cheol Shin, Su-Hyung Park, So Jeong Kim, Yeon-Kyung Oh, Byung Ha Lee, Se Hwan Yang, Young Chul Sung, Howard Lee, Sang Won Lee, Donghoon Choi, MinKyu Heo, Eui-Cheol Shin, Su-Hyung Park, So Jeong Kim, Yeon-Kyung Oh, Byung Ha Lee, Se Hwan Yang, Young Chul Sung, Howard Lee

Abstract

A low lymphocyte count puts immune-compromised patients at risk of mortality. hIL-7-hyFc is a homodimeric interleukin-7 (IL-7), a potent T-cell amplifier, fused to the hybridizing IgD/IgG4 immunoglobulin domain. We performed a randomized, double-blind, placebo-controlled, dose-escalation, phase I study to assess the pharmacokinetic, pharmacodynamic, safety, tolerability, and immunogenicity profiles of hIL-7-hyFc administered s.c. and i.m. to healthy volunteers. Thirty subjects randomly received hIL-7-hyFc or its matching placebo in an 8:2 ratio at 20, 60 μg/kg s.c., or 60 μg/kg i.m. The hIL-7-hyFc was slowly absorbed and its terminal half-life was 63.26 hours after i.m. administration. The hIL-7-hyFc increased absolute lymphocyte count, mostly in T-cells, which peaked 3 weeks after administration and then lasted for several additional weeks. The hIL-7-hyFc was well-tolerated after a single s.c. and i.m. administration. Injection site reaction was the most common treatment-emergent adverse event, which resolved spontaneously without treatment. The hIL-7-hyFc can be developed into a beneficial treatment option for patients with compromised T-cell immunity. This trial was registered at www.clinicaltrials.gov as #NCT02860715.

Conflict of interest statement

D.C., B.H.L., and S.H.Y. are employees of NeoImmuneTech, Inc. M.H., Y.‐K.O., and Y.C. Sung are employees of Genexine, Inc. All other authors declared no competing interests for this work.

© 2020 The Authors. Clinical and Translational Science published by Wiley Periodicals LLC on behalf of the American Society of Clinical Pharmacology and Therapeutics.

Figures

Figure 1
Figure 1
Mean serum concentration‐time profiles of interleukin (IL)‐7 after a single subcutaneous or intramuscular administration of hIL‐7‐hyFc. The error bars denote the SDs (● = 20 μg/kg s.c., N = 8; □ = 60 μg/kg s.c., N = 8; ▲ = 60 μg/kg i.m., N = 8).
Figure 2
Figure 2
Time course of mean pharmacodynamic parameters after a single subcutaneous or intramuscular administration of hIL‐7‐hyFc or placebo. (a) Absolute lymphocyte count. (b) Percent change from baseline in absolute lymphocyte count. (c) Absolute T‐cell count. (d) Percent change from baseline in absolute T‐cell count. (e) Absolute B‐cell count. (f) Percent change from baseline in absolute B‐cell count. The error bars denote the SDs. Upper dotted line: 50% change from baseline; middle dotted line: 0% change from baseline; lower dotted line: −50% change from baseline (● = 20 μg/kg s.c., N = 8; □ = 60 μg/kg s.c., N = 8; ▲ = 60 μg/kg i.m., N = 8; ▽ = placebo, N = 6).
Figure 3
Figure 3
Baseline‐corrected pharmacodynamic parameters after a single subcutaneous or intramuscular administration of hIL‐7‐hyFc or placebo. (a) maximum absolute lymphocyte count (ΔEmax), (b) area under the absolute lymphocyte count‐time curve until the last sampling time (ΔAUEC). The error bars represent the standard deviations (*P value < 0.05 vs. placebo; **P value < 0.01 vs. placebo).
Figure 4
Figure 4
Relationships between individual pharmacokinetic parameters vs. pharmacodynamic parameters. (a) maximum concentration (Cmax) of interleukin (IL)‐7 vs. maximum absolute lymphocyte count (ΔEmax), (b) Cmax of IL‐7 vs. absolute lymphocyte count‐time curve until the last sampling time (ΔAUEC), (c) area under the serum concentration vs. time curve (AUC) from time zero to the last measurable time‐point (AUClast) of IL‐7 vs. ΔEmax, (d) AUClast of IL‐7 vs. ΔAUEC of absolute lymphocyte count after a single subcutaneous or intramuscular administration of IL‐7 (● = 20 μg/kg s.c., N = 8; □ = 60 μg/kg s.c., N = 8; ▲ = 60 μg/kg i.m., N = 8).
Figure 5
Figure 5
Percentages of subjects positive on anti‐drug antibody against hIL‐7‐hyFc over time after administration by treatment group. (a) anti‐drug antibody (ADA), (b) neutralizing antibody (nAb).

References

    1. Péron, J. et al. CD4 lymphopenia to identify end‐of‐life metastatic cancer patients. Eur. J. Cancer 49, 1080–1089 (2013).
    1. Grossman, S.A. et al. Survival in patients with severe lymphopenia following treatment with radiation and chemotherapy for newly diagnosed solid tumors. J. Natl. Compr. Canc. Netw. 13, 1225–1231 (2015).
    1. Ray‐Coquard, I. et al. Lymphopenia as a prognostic factor for overall survival in advanced carcinomas, sarcomas, and lymphomas. Cancer Res. 69, 5383–5391 (2009).
    1. He, J.R. et al. Pretreatment levels of peripheral neutrophils and lymphocytes as independent prognostic factors in patients with nasopharyngeal carcinoma. Head Neck 34, 1769–1776 (2012).
    1. Gong, J. , Chehrazi‐Raffle, A. , Reddi, S. & Salgia, R. Development of PD‐1 and PD‐L1 inhibitors as a form of cancer immunotherapy: a comprehensive review of registration trials and future considerations. J. Immunotherapy Cancer 6, 8 (2018).
    1. Smith, D.K. , Neal, J.J. & Holmberg, S.D. Unexplained opportunistic infections and CD4+ T‐lymphocytopenia without HIV infection–an investigation of cases in the United States. N. Engl. J. Med. 328, 373–379 (1993).
    1. Walker, U.A. & Warnatz, K. Idiopathic CD4 lymphocytopenia. Curr. Opin. Rheumatol. 18, 389–395 (2006).
    1. Hequet, O. et al. Multifocal progressive leukoencephalopathy occurring after refractory anemia and multiple infectious disorders consecutive to severe lymphopenia. Ann. Hematol. 81, 340–342 (2002).
    1. Glück, T. , Kiefmann, B. , Grohmann, M. , Falk, W. , Straub, R.H. & Schölmerich, J. Immune status and risk for infection in patients receiving chronic immunosuppressive therapy. J. Rheumatol. 32, 1473–1480 (2005).
    1. Duncan, R.A. , von Reyn, C.F. , Alliegro, G.M. , Toossi, Z. , Sugar, A.M. & Levitz, S.M. Idiopathic CD4+ T‐lymphocytopenia–four patients with opportunistic infections and no evidence of HIV infection. N. Engl. J. Med. 328, 393–398 (1993).
    1. Cunningham‐Rundles, C. & Bodian, C. Common variable immunodeficiency: clinical and immunological features of 248 patients. Clin. Immunol. 92, 34–48 (1999).
    1. Valiathan, R. , Deeb, K. , Diamante, M. , Ashman, M. , Sachdeva, N. & Asthana, D. Reference ranges of lymphocyte subsets in healthy adults and adolescents with special mention of T cell maturation subsets in adults of South Florida. Immunobiology 219, 487–496 (2014).
    1. Park, J.‐H. et al. Suppression of IL7Rα transcription by IL‐7 and other prosurvival cytokines: a novel mechanism for maximizing IL‐7‐dependent T cell survival. Immunity 21, 289–302 (2004).
    1. Fry, T.J. et al. IL‐7 therapy dramatically alters peripheral T‐cell homeostasis in normal and SIV‐infected nonhuman primates. Blood 101, 2294–2299 (2003).
    1. Sportès, C. et al. Phase I study of recombinant human interleukin‐7 administration in subjects with refractory malignancy. Clin. Cancer Res. 16, 727–735 (2010).
    1. Andersson, Å. et al. IL‐7 promotes CXCR3 ligand‐dependent T cell antitumor reactivity in lung cancer. J. Immunol. 182, 6951–6958 (2009).
    1. Tan, J.T. et al. IL‐7 is critical for homeostatic proliferation and survival of naive T cells. Proc. Natl. Acad. Sci. 98, 8732–8737 (2001).
    1. Sheikh, V. et al. Administration of interleukin‐7 increases CD4 T cells in idiopathic CD4 lymphocytopenia. Blood 127, 977–988 (2016).
    1. Levy, Y. et al. Enhanced T cell recovery in HIV‐1–infected adults through IL‐7 treatment. J. Clin. Investig. 119, 997–1007 (2009).
    1. Alstadhaug, K.B. et al. Treatment of progressive multifocal leukoencephalopathy with interleukin 7. JAMA Neurol. 71, 1030–1035 (2014).
    1. Zhong, H. et al. Interleukin‐7 in patients with chronic hepatitis B may have effect on T follicular helper cells and specific cellular immunity. Hepatitis Monthly 16, e36068 (2016).
    1. Hou, H. , Kang, Y. , Zeng, Y. , Li, Y. & Shang, J. Interleukin‐7 augments CD8+ T cells function and promotes viral clearance in chronic hepatitis C virus infection. Cytokine 102, 26–33 (2018).
    1. Bocci, V. Interleukins. Clin. Pharmacokinet. 21, 274–284 (1991).
    1. Huhn, R.D.C. Clinical pharmacology and development of recombinant human biological agents In: Biotechnology and Safety Assessment (eds. Thomas J.A. & Myers L.A.) 59–78 (Raven Press, New York, 1993).
    1. Sockolosky, J.T. & Szoka, F.C. The neonatal Fc receptor, FcRn, as a target for drug delivery and therapy. Adv. Drug Deliv. Rev. 91, 109–124 (2015).
    1. Sereti, I. et al. IL‐7 administration drives T cell–cycle entry and expansion in HIV‐1 infection. Blood 113, 6304–6314 (2009).
    1. Knauf, M.J. , Bell, D.P. , Hirtzer, P. , Luo, Z.‐P. , Young, J.D. & Katre, N.V. Relationship of effective molecular size to systemic clearance in rats of recombinant interleukin‐2 chemically modified with water‐soluble polymers. J. Biol. Chem. 263, 15064–15070 (1988).
    1. Beq, S. et al. Injection of glycosylated recombinant simian IL‐7 provokes rapid and massive T‐cell homing in rhesus macaques. Blood 114, 816–825 (2009).
    1. Kovacs, J.A. et al. Interleukin‐2 induced immune effects in human immunodeficiency virus‐infected patients receiving intermittent interleukin‐2 immunotherapy. Eur. J. Immunol. 31, 1351–1360 (2001).
    1. Sportès, C. 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. 205, 1701–1714 (2008).
    1. LeBien, T.W. Fates of human B‐cell precursors. Blood 96, 9–23 (2000).
    1. Dittel, B.N. & LeBien, T.W. The growth response to IL‐7 during normal human B cell ontogeny is restricted to B‐lineage cells expressing CD34. J. Immunol. 154, 58–67 (1995).
    1. Johnson, S.E. , Shah, N. , Panoskaltsis‐Mortari, A. & LeBien, T.W. Murine and human IL‐7 activate STAT5 and induce proliferation of normal human pro‐B cells. J. Immunol. 175, 7325–7331 (2005).
    1. Mould, D.R. & Sweeney, K.R. The pharmacokinetics and pharmacodynamics of monoclonal antibodies‐mechanistic modeling applied to drug development. Curr. Opin. Drug Discov. Devel. 10, 84 (2007).
    1. Im, S.J. et al. Natural form of noncytolytic flexible human Fc as a long‐acting carrier of agonistic ligand, erythropoietin. PLoS One 6, e24574 (2011).
    1. Levy, Y. et al. 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. Clin. Infect. Dis. 55, 291–300 (2012).
    1. Perales, M.‐A. et al. Recombinant human interleukin‐7 (CYT107) promotes T‐cell recovery after allogeneic stem cell transplantation. Blood 120, 4882–4891 (2012).

Source: PubMed

3
구독하다