NHS-IL2 combined with radiotherapy: preclinical rationale and phase Ib trial results in metastatic non-small cell lung cancer following first-line chemotherapy

Michel M van den Heuvel, Marcel Verheij, Rogier Boshuizen, José Belderbos, Anne-Marie C Dingemans, Dirk De Ruysscher, Julien Laurent, Robert Tighe, John Haanen, Sonia Quaratino, Michel M van den Heuvel, Marcel Verheij, Rogier Boshuizen, José Belderbos, Anne-Marie C Dingemans, Dirk De Ruysscher, Julien Laurent, Robert Tighe, John Haanen, Sonia Quaratino

Abstract

Background: NHS-IL2 (selectikine, EMD 521873, MSB0010445) consists of human NHS76 (antibody specific for necrotic DNA) fused to genetically modified human interleukin 2 (IL-2) and selectively activates the high-affinity IL-2 receptor. Based on an evolving investigational concept to prime the tumor microenvironment with ionizing radiation prior to initiating immunotherapy, 2 related studies were conducted and are reported here. The first, a preclinical study, tests the systemic effect of the immunocytokine NHS-IL2 and radiotherapy in a lung carcinoma animal model; the second, a phase Ib trial in patients with metastatic non-small cell lung carcinoma (NSCLC), was designed to determine the safety and tolerability of NHS-IL2 in combination with radiotherapy directly following first-line palliative chemotherapy.

Methods: Tumor-bearing C57Bl/6 mice were treated with NHS-IL2 alone (5 mg/kg; days 7-9), fractionated radiotherapy (3.6 Gy; days 0-4) plus cisplatin (4 mg/kg; day 0), or the triple combination. Metastatic NSCLC patients who achieved disease control with first-line palliative chemotherapy were enrolled in the phase Ib trial. Patients received local irradiation (5x 4 Gy) of a single pulmonary nodule. Dose-escalated NHS-IL2 was administered as 1-h intravenous infusion on 3 consecutive days every 3 weeks.

Results: NHS-IL2 plus radiotherapy induced immune response activation and complete tumor growth regressions in 80%-100% of mice. In patients with metastatic NSCLC treated with NHS-IL2 (3, 3, and 7 patients in the 0.15-mg/kg, 0.30-mg/kg, and 0.45-mg/kg cohorts, respectively), maximum tolerated dose was not reached. Most frequently reported adverse events were fatigue, anorexia, and rash. Transient increases in leukocyte subsets were observed. In 3 patients, thyroid gland dysfunction occurred. No objective responses were reported; long-term survival was observed in 2 patients, including 1 patient with long-term tumor control.

Conclusions: Combining NHS-IL2 with radiotherapy achieved synergistic antitumor activity in preclinical studies, supporting the use in lung cancer patients. This combination was well tolerated and 2 of 13 patients achieved long-term survival.

Trial registration: ClinicalTrials.gov NCT00879866.

Figures

Figure 1
Figure 1
Mouse model (LLC in C57/Bl6) study design (A) and observed tumor growth inhibition (B). Mice received the indicated treatments when tumors had reached volume of 81 mm3 to 137 mm3 7 days after implantation. Cisplatin 4 mg/kg was administered only on day 0. The dose of radiotherapy was 3.6 Gy and the dose of NHS-IL2 was 5 mg/kg. Each treatment group comprised 10 mice through day 14, and 6 mice thereafter; 4 mice from each group, selected around the mean, were sacrificed on day 14 for mechanistic endpoints. The experiment was repeated 3 times with similar results. Cis, cisplatin; RTX, radiotherapy.
Figure 2
Figure 2
Clinical trial design (A) and observed NHS-IL2 serum concentrations (B). Patients with metastatic NSCLC after first-line platinum-based therapy (4–6 cycles) were treated with radiotherapy followed by NHS-IL2 at escalating doses of 0.15 mg/kg, 0.30 mg/kg, or 0.45 mg/kg. Mean serum concentration of NHS-IL2 is depicted by dose level during the first, second, and fourth treatment cycles. CTX, chemotherapy; NSCLC, non-small cell lung cancer; PR, partial response; Pt, platinum; SD, stable disease.
Figure 3
Figure 3
Time course of leukocyte subpopulations in metastatic NSCLC patients treated with NHS-IL2 (by dose group). Absolute counts (109 cells/mm3) are shown for total leukocytes, lymphocytes, neutrophils, monocytes, basophils, and eosinophils from NSCLC patients treated with NHS-IL2 at dose levels 0.15 mg/kg, 0.30 mg/kg, and 0.45 mg/kg. Geometric mean values of absolute counts are shown for each dose level. Blood was drawn 7 days before start of treatment and at days 1 and 8 of each cycle until discontinuation. The arrow identifies the first of the 3 consecutive days of NHS-IL2 infusion. NSCLC, non-small cell lung cancer.
Figure 4
Figure 4
Time course of lymphocyte subsets (median by treatment group). Percentage change from baseline (day –7) based on absolute count values (cells/mm3) on days 1 and 8 during the first and second treatment cycles for: (A) proliferative CD4 T cells (CD3 + CD4 + Ki67+) and memory CD4 T cells (CD3 + CD4 + CD197–CD45RA–); (B) proliferative CD8 T cells (CD3 + CD8 + Ki67+) and memory CD8 T cells (CD3 + CD8 + CD197–CD45RA–); and (C) immature NK cells (CD16–CD56brightCD3–) and mature NK cells CD16 + CD56 + CD3–. (D) sIL-2Ralpha serum concentrations (pg/mL) at baseline and on days 1 and 8 during the first cycle. Graphs show the individual values from 12 NSCLC patients treated with NHS-IL2 at doses of 0.15 mg/kg, 0.30 mg/kg, and 0.45 mg/kg. Bars represent geometric mean values. NK, natural killer; NSCLC, non-small cell lung cancer; sIL-2Ralpha, soluble IL-2 receptor alpha.

References

    1. Mok TS, Wu YL, Thongprasert S, Yang CH, Chu DT, Saijo N, et al. Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma. N Engl J Med. 2009;361:947–57. doi: 10.1056/NEJMoa0810699.
    1. Camidge DR, Bang YJ, Kwak EL, Iafrate AJ, Varella-Garcia M, Fox SB, et al. Activity and safety of crizotinib in patients with ALK-positive non-small-cell lung cancer: updated results from a phase 1 study. Lancet Oncol. 2012;13:1011–9. doi: 10.1016/S1470-2045(12)70344-3.
    1. Butts C, Murray N, Maksymiuk A, Goss G, Marshall E, Soulieres D, et al. Randomized phase IIB trial of BLP25 liposome vaccine in stage IIIB and IV non-small-cell lung cancer. J Clin Oncol. 2005;23:6674–81. doi: 10.1200/JCO.2005.13.011.
    1. Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 2012;366:2443–54. doi: 10.1056/NEJMoa1200690.
    1. Lynch TJ, Bondarenko I, Luft A, Serwatowski P, Barlesi F, Chacko R, et al. Ipilimumab in combination with paclitaxel and carboplatin as first-line treatment in stage IIIB/IV non-small-cell lung cancer: results from a randomized, double-blind, multicenter phase II study. J Clin Oncol. 2012;30:2046–54. doi: 10.1200/JCO.2011.38.4032.
    1. Van den Heuvel MM, Burgers SA, van Zandwijk N. Immunotherapy in non-small-cell lung carcinoma: from inflammation to vaccination. Clin Lung Cancer. 2009;10:99–105. doi: 10.3816/CLC.2009.n.012.
    1. Tester WJ, Kim KM, Krigel RL, Bonomi PD, Glick JH, Asbury RF, et al. A randomized phase II study of interleukin-2 with and without beta-interferon for patients with advanced non-small cell lung cancer: an eastern cooperative oncology group study (PZ586) Lung Cancer. 1999;25:199–206. doi: 10.1016/S0169-5002(99)00054-9.
    1. Bersanelli M, Buti S, Camisa R, Brighenti M, Lazzarelli S, Mazza G, et al. Gefitinib plus interleukin-2 in advanced non-small cell lung cancer patients previously treated with chemotherapy. Cancers (Basel) 2014;6:2035–48. doi: 10.3390/cancers6042035.
    1. Kimura H, Yamaguchi Y. A phase III randomized study of interleukin-2 lymphokine-activated killer cell immunotherapy combined with chemotherapy or radiotherapy after curative or noncurative resection of primary lung carcinoma. Cancer. 1997;80:42–9. doi: 10.1002/(SICI)1097-0142(19970701)80:1<42::AID-CNCR6>;2-H.
    1. Gillies SD, Lan Y, Hettmann T, Brunkhorst B, Sun Y, Mueller SO, et al. A low-toxicity IL-2-based immunocytokine retains antitumor activity despite its high degree of IL-2 receptor selectivity. Clin Cancer Res. 2011;17:3673–85. doi: 10.1158/1078-0432.CCR-10-2921.
    1. Gillessen S, Gnad-Vogt US, Gallerani E, Beck J, Sessa C, Omlin A, et al. A phase I dose-escalation study of the immunocytokine EMD 521873 (Selectikine) in patients with advanced solid tumours. Eur J Cancer. 2013;49:35–44. doi: 10.1016/j.ejca.2012.07.015.
    1. Demaria S, Formenti SC. Radiation as an immunological adjuvant: current evidence on dose and fractionation. Front Oncol. 2012;2:153.
    1. Postow MA, Callahan MK, Barker CA, Yamada Y, Yuan J, Kitano S, et al. Immunologic correlates of the abscopal effect in a patient with melanoma. N Engl J Med. 2012;366:925–31. doi: 10.1056/NEJMoa1112824.
    1. Demaria S, Bhardwaj N, McBride WH, Formenti SC. Combining radiotherapy and immunotherapy: a revived partnership. Int J Radiat Oncol Biol Phys. 2005;63:655–66. doi: 10.1016/j.ijrobp.2005.06.032.
    1. Kudo-Saito C, Schlom J, Camphausen K, Coleman CN, Hodge JW. The requirement of multimodal therapy (vaccine, local tumor radiation, and reduction of suppressor cells) to eliminate established tumors. Clin Cancer Res. 2005;11:4533–44. doi: 10.1158/1078-0432.CCR-04-2237.
    1. Reits EA, Hodge JW, Herberts CA, Groothuis TA, Chakraborty M, Wansley EK, et al. Radiation modulates the peptide repertoire, enhances MHC class I expression, and induces successful antitumor immunotherapy. J Exp Med. 2006;203:1259–71. doi: 10.1084/jem.20052494.
    1. Formenti SC, Demaria S. Combining radiotherapy and cancer immunotherapy: a paradigm shift. J Natl Cancer Inst. 2013;105:256–65. doi: 10.1093/jnci/djs629.
    1. Laurent J, Touvrey C, Gillessen S, Joffraud M, Vicari M, Bertrand C, et al. T-cell activation by treatment of cancer patients with EMD 521873 (Selectikine), an IL-2/anti-DNA fusion protein. J Transl Med. 2013;11:5. doi: 10.1186/1479-5876-11-5.
    1. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144:646–74. doi: 10.1016/j.cell.2011.02.013.
    1. Seung SK, Curti BD, Crittenden M, Walker E, Coffey T, Siebert JC, et al. Phase 1 study of stereotactic body radiotherapy and interleukin-2--tumor and immunological responses. Sci Transl Med. 2012;4:137–74. doi: 10.1126/scitranslmed.3003649.
    1. Crowley J, Ankerst DP. Handbook of Statistics in Clinical Oncology. 2. Boca Raton: Boca RatonChapman & Hall/CRC Taylor & Francis Group; 2006.
    1. De Ruysscher D, Faivre-Finn C, Nestle U, Hurkmans CW, Le Pechoux C, Price A, et al. European organisation for research and treatment of cancer recommendations for planning and delivery of high-dose, high-precision radiotherapy for lung cancer. J Clin Oncol. 2010;28:5301–10. doi: 10.1200/JCO.2010.30.3271.
    1. National Cancer Institute: Common Terminology Criteria for Adverse Events v3.0 (CTCAE). [].
    1. Therasse P, Eisenhauer EA, Verweij J. RECIST revisited: a review of validation studies on tumour assessment. Eur J Cancer. 2006;42:1031–9. doi: 10.1016/j.ejca.2006.01.026.
    1. Demaria S, Formenti SC. Role of T lymphocytes in tumor response to radiotherapy. Front Oncol. 2012;2:95.
    1. Matsumura S, Wang B, Kawashima N, Braunstein S, Badura M, Cameron TO, et al. Radiation-induced CXCL16 release by breast cancer cells attracts effector T cells. J Immunol. 2008;181:3099–107. doi: 10.4049/jimmunol.181.5.3099.
    1. Suzuki Y, Mimura K, Yoshimoto Y, Watanabe M, Ohkubo Y, Izawa S, et al. Immunogenic tumor cell death induced by chemoradiotherapy in patients with esophageal squamous cell carcinoma. Cancer Res. 2012;72:3967–76. doi: 10.1158/0008-5472.CAN-12-0851.
    1. Martin OA, Anderson RL, Russell PA, Cox RA, Ivashkevich A, Swierczak A, et al. Mobilization of viable tumor cells into the circulation during radiation therapy. Int J Radiat Oncol Biol Phys. 2014;88:395–403. doi: 10.1016/j.ijrobp.2013.10.033.
    1. Marchesi M, Andersson E, Villabona L, Seliger B, Lundqvist A, Kiessling R, et al. HLA-dependent tumour development: a role for tumour associate macrophages? J Transl Med. 2013;11:247–5876. doi: 10.1186/1479-5876-11-247.
    1. Ma Y, Kepp O, Ghiringhelli F, Apetoh L, Aymeric L, Locher C, et al. Chemotherapy and radiotherapy: cryptic anticancer vaccines. Semin Immunol. 2010;22:113–24. doi: 10.1016/j.smim.2010.03.001.
    1. Fagnoni FF, Zerbini A, Pelosi G, Missale G. Combination of radiofrequency ablation and immunotherapy. Front Biosci. 2008;13:369–81. doi: 10.2741/2686.
    1. Dunn GP, Bruce AT, Ikeda H, Old LJ, Schreiber RD. Cancer immunoediting: from immunosurveillance to tumor escape. Nat Immunol. 2002;3:991–8. doi: 10.1038/ni1102-991.

Source: PubMed

3
S'abonner