Phase I study of single agent NIZ985, a recombinant heterodimeric IL-15 agonist, in adult patients with metastatic or unresectable solid tumors

Kevin Conlon, Dionysios C Watson, Thomas A Waldmann, Antonio Valentin, Cristina Bergamaschi, Barbara K Felber, Cody J Peer, William D Figg, E Lake Potter, Mario Roederer, Douglas G McNeel, John A Thompson, Sumati Gupta, Rom Leidner, Andrea Wang-Gillam, Nehal S Parikh, Debby Long, Sema Kurtulus, Lang Ho Lee, Niladri Roy Chowdhury, Florent Bender, George N Pavlakis, Kevin Conlon, Dionysios C Watson, Thomas A Waldmann, Antonio Valentin, Cristina Bergamaschi, Barbara K Felber, Cody J Peer, William D Figg, E Lake Potter, Mario Roederer, Douglas G McNeel, John A Thompson, Sumati Gupta, Rom Leidner, Andrea Wang-Gillam, Nehal S Parikh, Debby Long, Sema Kurtulus, Lang Ho Lee, Niladri Roy Chowdhury, Florent Bender, George N Pavlakis

Abstract

Background: NIZ985 is a recombinant heterodimer of physiologically active interleukin (IL-)15 and IL-15 receptor alpha. In preclinical models, NIZ985 promotes cytotoxic lymphocyte proliferation, killing function, and organ/tumor infiltration, with resultant anticancer effects. In this first-in-human study, we assessed the safety, pharmacokinetics, and immune effects of NIZ985 in patients with metastatic or unresectable solid tumors.

Methods: Single agent NIZ985 dose escalation data are reported from a phase I dose escalation/expansion study of NIZ985 as monotherapy. Adult patients (N=14) received 0.25, 0.5, 1, 2 or 4 µg/kg subcutaneous NIZ985 three times weekly (TIW) for the first 2 weeks of each 28-day cycle, in an accelerated 3+3 dose escalation trial design. IL-15 and endogenous cytokines were monitored by ELISA and multiplexed electrochemiluminescent assays. Multiparameter flow cytometry assessed the frequency, phenotype and proliferation of peripheral blood mononuclear cells. Preliminary antitumor activity was assessed by overall response rate (Response Evaluation Criteria in Solid Tumors V.1.1).

Results: As of March 2, 2020, median treatment duration was 7.5 weeks (range 1.1-77.1). Thirteen patients had discontinued and one (uveal melanoma) remains on treatment with stable disease. Best clinical response was stable disease (3 of 14 patients; 21%). The most frequent adverse events (AEs) were circular erythematous injection site reactions (100%), chills (71%), fatigue (57%), and fever (50%). Treatment-related grade 3/4 AEs occurred in six participants (43%); treatment-related serious AEs (SAEs) in three (21%). The per-protocol maximum tolerated dose was not reached. Pharmacokinetic accumulation of serum IL-15 in the first week was followed by significantly lower levels in week 2, likely due to more rapid cytokine consumption by an expanding lymphocyte pool. NIZ985 treatment was associated with increases in several cytokines, including interferon (IFN)-γ, IL-18, C-X-C motif chemokine ligand 10, and tumor necrosis factor-β, plus significant induction of cytotoxic lymphocyte proliferation (including natural killer and CD8+ T cells), increased CD16+ monocytes, and increased CD163+ macrophages at injection sites.

Conclusions: Subcutaneous NIZ985 TIW was generally well tolerated in patients with advanced cancer and produced immune activation paralleling preclinical observations, with induction of IFN-γ and proliferation of cytotoxic lymphocytes. Due to delayed SAEs at the two highest dose levels, administration is being changed to once-weekly in a revised protocol, as monotherapy and combined with checkpoint inhibitor spartalizumab. These alterations are expected to maximize the potential of NIZ985 as a novel immunotherapy.

Trial registration number: NCT02452268.

Keywords: clinical trials as topic; cytokines; immunomodulation; immunotherapy; investigational; therapies.

Conflict of interest statement

Competing interests: AV, CB, BKF, and GNP report inventorship in issued patents of relevance to this work (licensed to Novartis; managed by the National Cancer Institute) and other support by Novartis during the conduct of this study. DGM and TAW report other support from Novartis during the conduct of the study. JT reports grants from Alpine Biosciences, other support from Novartis during the conduct of the study, other support from Pfizer, Five Prime, Merck, Trillium, Incyte and Xencor outside of the current work, and personal fees from Regeneron, Aveo, Neoleukin, Seattle Genetics, BJ Biosciences, and Calithera outside of the current work. SG reports other support from Novartis during the conduct of the study, other support from Bristol Myers Squibb, Rexahn, Incyte, LSK, Five Prime, Mirati, QED, Debiopharm, Merck, Pfizer, AstraZeneca, Medimmune, Clovis, and Seattle Genetics outside of the current work, and ownership of stock in Salerius Pharmaceuticals by spouse. RL reports grants and personal fees from Bristol Myers Squibb, personal fees from Merck, Oncolys, and Sanofi, and non-financial support from Clinigen outside of the current work. NSP, DL, SK, LHL, NRC and FB are, or were at the time of the work described, employees of Novartis Pharmaceuticals. KC, DCW, CJP, WF, ELP, MR and AW-G report nothing to disclose.

© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY. Published by BMJ.

Figures

Figure 1
Figure 1
NIZ985 injection site reaction is characterized by local immune cell infiltration. Micrographs from skin biopsy of patient treated with 2 μg/kg NIZ985. (A) H&E stain demonstrated subacute spongiotic dermatitis with superficial perivascular inflammation. (B–E) Immunohistochemistry micrographs for immune cell markers. (B) CD3+ cells (T cells) and (C) CD8+ cells were primarily found near dermal blood vessels (black arrows). (D) CD56+ cells were also found near the dermal/epidermal junction, while (E) CD163+ cells (monocyte lineage) were prominent throughout the affected dermis.
Figure 2
Figure 2
Pharmacokinetics and circulating serum IL-15 concentrations during cycle 1 of NIZ985 treatment in a subset of participants. (A) Serum IL-15 concentration–time profile (mean±SEM) after subcutaneous injection of NIZ985 on cycle 1, day 1. (B) Pooled analysis of serum IL-15 levels immediately before each injection of NIZ985 (1 and 2 μg/kg groups). Violin plots depict median, quartiles and range of data. Each data point represents an individual patient sample. Comparison between indicated time points was by mixed-effects ANOVA. ANOVA, analysis of variance; IL, interleukin.
Figure 3
Figure 3
Lymphocyte proliferation is consistently induced during NIZ985 treatment cycles. (A–D) Individual proliferative responses (percentage Ki67+ cells) induced during each cycle of NIZ985 in circulating CD4+ T cells, CD8+ T cells, γδ T cells, and NK cells. Insets show mean peak percentage Ki67+ cells by dosing group; (E) Individual percentage CD16+ monocytes (from total CD14+ monocytes) during each cycle of NIZ985. NK, natural killer.
Figure 4
Figure 4
(A–E) Mean peak induction (log2-transformed mean ratios of peak-to-baseline levels) of selected plasma cytokines during cycle 1 dosing; (F) Mean cytokine induction (24–240 hours post-dose) in the pooled 1.0 and 2.0 µg/kg dosing groups. X-axis: Log2-transformed mean ratios of peak to baseline cytokine levels in both groups; Y-axis: Correlation coefficients of the mean ratio across all dose groups and NIZ985 dose level. Bars identify regions of high correlation (Pearson’s r ≥0.8 or ≤–0.8) and high response (≥2-fold). C, cycle; CRP, C-reactive protein; CXCL10, C-X-C motif chemokine ligand 10; D, day; FC, fold-change; IFN, interferon; IL, interleukin; TNF, tumor necrosis factor.

References

    1. Ma A, Koka R, Burkett P. Diverse functions of IL-2, IL-15, and IL-7 in lymphoid homeostasis. Annu Rev Immunol 2006;24:657–79. 10.1146/annurev.immunol.24.021605.090727
    1. Waldmann TA. The shared and contrasting roles of IL2 and IL15 in the life and death of normal and neoplastic lymphocytes: implications for cancer therapy. Cancer Immunol Res 2015;3:219–27. 10.1158/2326-6066.CIR-15-0009
    1. Ahmadzadeh M, Rosenberg SA. IL-2 administration increases CD4+ CD25(hi) Foxp3+ regulatory T cells in cancer patients. Blood 2006;107:2409–14. 10.1182/blood-2005-06-2399
    1. Long SA, Rieck M, Sanda S, et al. . Rapamycin/IL-2 combination therapy in patients with type 1 diabetes augments tregs yet transiently impairs β-cell function. Diabetes 2012;61:2340–8. 10.2337/db12-0049
    1. Pachella LA, Madsen LT, Dains JE. The toxicity and benefit of various dosing strategies for interleukin-2 in metastatic melanoma and renal cell carcinoma. J Adv Pract Oncol 2015;6:212–21.
    1. Marks-Konczalik J, Dubois S, Losi JM, et al. . Il-2-induced activation-induced cell death is inhibited in IL-15 transgenic mice. Proc Natl Acad Sci U S A 2000;97:11445–50. 10.1073/pnas.200363097
    1. Wuest TY, Willette-Brown J, Durum SK, et al. . The influence of IL-2 family cytokines on activation and function of naturally occurring regulatory T cells. J Leukoc Biol 2008;84:973–80. 10.1189/jlb.1107778
    1. Bergamaschi C, Stravokefalou V, Stellas D, et al. . Heterodimeric IL-15 in cancer immunotherapy. Cancers 2021;13:837. 10.3390/cancers13040837
    1. Burkett PR, Koka R, Chien M, et al. . Coordinate expression and trans presentation of interleukin (IL)-15Ralpha and IL-15 supports natural killer cell and memory CD8+ T cell homeostasis. J Exp Med 2004;200:825–34. 10.1084/jem.20041389
    1. Sandau MM, Schluns KS, Lefrancois L, et al. . Cutting edge: transpresentation of IL-15 by bone marrow-derived cells necessitates expression of IL-15 and IL-15R alpha by the same cells. J Immunol 2004;173:6537–41. 10.4049/jimmunol.173.11.6537
    1. Schluns KS, Klonowski KD, Lefrançois L. Transregulation of memory CD8 T-cell proliferation by IL-15Ralpha+ bone marrow-derived cells. Blood 2004;103:988–94. 10.1182/blood-2003-08-2814
    1. Anderson DM, Kumaki S, Ahdieh M, et al. . Functional characterization of the human interleukin-15 receptor alpha chain and close linkage of IL15RA and IL2RA genes. J Biol Chem 1995;270:29862–9. 10.1074/jbc.270.50.29862
    1. Bergamaschi C, Rosati M, Jalah R, et al. . Intracellular interaction of interleukin-15 with its receptor alpha during production leads to mutual stabilization and increased bioactivity. J Biol Chem 2008;283:4189–99. 10.1074/jbc.M705725200
    1. Bergamaschi C, Jalah R, Kulkarni V, et al. . Secretion and biological activity of short signal peptide IL-15 is chaperoned by IL-15 receptor alpha in vivo. J Immunol 2009;183:3064–72. 10.4049/jimmunol.0900693
    1. Mortier E, Woo T, Advincula R, et al. . Il-15Ralpha chaperones IL-15 to stable dendritic cell membrane complexes that activate NK cells via trans presentation. J Exp Med 2008;205:1213–25. 10.1084/jem.20071913
    1. Dubois S, Mariner J, Waldmann TA, et al. . Il-15Ralpha recycles and presents IL-15 in trans to neighboring cells. Immunity 2002;17:537–47. 10.1016/S1074-7613(02)00429-6
    1. Chertova E, Bergamaschi C, Chertov O, et al. . Characterization and favorable in vivo properties of heterodimeric soluble IL-15·IL-15Rα cytokine compared to IL-15 monomer. J Biol Chem 2013;288:18093–103. 10.1074/jbc.M113.461756
    1. Bergamaschi C, Bear J, Rosati M, et al. . Circulating IL-15 exists as heterodimeric complex with soluble IL-15Rα in human and mouse serum. Blood 2012;120:e1–8. 10.1182/blood-2011-10-384362
    1. Bergamaschi C, Kulkarni V, Rosati M, et al. . Intramuscular delivery of heterodimeric IL-15 DNA in macaques produces systemic levels of bioactive cytokine inducing proliferation of NK and T cells. Gene Ther 2015;22:76–86. 10.1038/gt.2014.84
    1. Bergamaschi C, Watson DC, Valentin A, et al. . Optimized administration of hetIL-15 expands lymphocytes and minimizes toxicity in rhesus macaques. Cytokine 2018;108:213–24. 10.1016/j.cyto.2018.01.011
    1. Watson DC, Moysi E, Valentin A, et al. . Treatment with native heterodimeric IL-15 increases cytotoxic lymphocytes and reduces SHIV RNA in lymph nodes. PLoS Pathog 2018;14:e1006902. 10.1371/journal.ppat.1006902
    1. Dubois S, Patel HJ, Zhang M, et al. . Preassociation of IL-15 with IL-15R alpha-IgG1-Fc enhances its activity on proliferation of NK and CD8+/CD44high T cells and its antitumor action. J Immunol 2008;180:2099–106. 10.4049/jimmunol.180.4.2099
    1. Rubinstein MP, Kovar M, Purton JF, et al. . Converting IL-15 to a superagonist by binding to soluble IL-15R{alpha}. Proc Natl Acad Sci U S A 2006;103:9166–71. 10.1073/pnas.0600240103
    1. Stoklasek TA, Schluns KS, Lefrançois L. Combined IL-15/IL-15Ralpha immunotherapy maximizes IL-15 activity in vivo. J Immunol 2006;177:6072–80. 10.4049/jimmunol.177.9.6072
    1. Klebanoff CA, Finkelstein SE, Surman DR, et al. . Il-15 enhances the in vivo antitumor activity of tumor-reactive CD8+ T cells. Proc Natl Acad Sci U S A 2004;101:1969–74. 10.1073/pnas.0307298101
    1. Ng SSM, Nagy BA, Jensen SM, et al. . Heterodimeric IL15 treatment enhances tumor infiltration, persistence, and effector functions of adoptively transferred tumor-specific T cells in the absence of Lymphodepletion. Clin Cancer Res 2017;23:2817–30. 10.1158/1078-0432.CCR-16-1808
    1. Bergamaschi C, Pandit H, Nagy BA, et al. . Heterodimeric IL-15 delays tumor growth and promotes intratumoral CTL and dendritic cell accumulation by a cytokine network involving XCL1, IFN-γ, CXCL9 and CXCL10. J Immunother Cancer 2020;8:e000599. 10.1136/jitc-2020-000599
    1. Epardaud M, Elpek KG, Rubinstein MP, et al. . Interleukin-15/interleukin-15R alpha complexes promote destruction of established tumors by reviving tumor-resident CD8+ T cells. Cancer Res 2008;68:2972–83. 10.1158/0008-5472.CAN-08-0045
    1. Kim PS, Kwilas AR, Xu W, et al. . Il-15 superagonist/IL-15RαSushi-Fc fusion complex (IL-15SA/IL-15RαSu-Fc; ALT-803) markedly enhances specific subpopulations of NK and memory CD8+ T cells, and mediates potent anti-tumor activity against murine breast and colon carcinomas. Oncotarget 2016;7:16130–45. 10.18632/oncotarget.7470
    1. Munger W, DeJoy SQ, Jeyaseelan R, et al. . Studies evaluating the antitumor activity and toxicity of interleukin-15, a new T cell growth factor: comparison with interleukin-2. Cell Immunol 1995;165:289–93. 10.1006/cimm.1995.1216
    1. Rosario M, Liu B, Kong L, et al. . The IL-15-based ALT-803 complex enhances FcγRIIIa-triggered NK cell responses and in vivo clearance of B cell lymphomas. Clin Cancer Res 2016;22:596–608. 10.1158/1078-0432.CCR-15-1419
    1. Zhang M, Ju W, Yao Z, et al. . Augmented IL-15Rα expression by CD40 activation is critical in synergistic CD8 T cell-mediated antitumor activity of anti-CD40 antibody with IL-15 in TRAMP-C2 tumors in mice. J Immunol 2012;188:6156–64. 10.4049/jimmunol.1102604
    1. Zhang M, Yao Z, Dubois S, et al. . Interleukin-15 combined with an anti-CD40 antibody provides enhanced therapeutic efficacy for murine models of colon cancer. Proc Natl Acad Sci U S A 2009;106:7513–8. 10.1073/pnas.0902637106
    1. Conlon KC, Lugli E, Welles HC, et al. . Redistribution, hyperproliferation, activation of natural killer cells and CD8 T cells, and cytokine production during first-in-human clinical trial of recombinant human interleukin-15 in patients with cancer. J Clin Oncol 2015;33:10.1200/JCO.2014.57.3329:74–82. 10.1200/JCO.2014.57.3329
    1. Cooley S, He F, Bachanova V, et al. . First-in-human trial of rhIL-15 and haploidentical natural killer cell therapy for advanced acute myeloid leukemia. Blood Adv 2019;3:1970–80. 10.1182/bloodadvances.2018028332
    1. Miller JS, Morishima C, McNeel DG, et al. . A first-in-human phase I study of subcutaneous outpatient recombinant human IL15 (rhIL15) in adults with advanced solid tumors. Clin Cancer Res 2018;24:1525–35. 10.1158/1078-0432.CCR-17-2451
    1. Conlon KC, Potter EL, Pittaluga S, et al. . Il15 by continuous intravenous infusion to adult patients with solid tumors in a phase I trial induced dramatic NK-cell subset expansion. Clin Cancer Res 2019;25:4945–54. 10.1158/1078-0432.CCR-18-3468
    1. Margolin K, Morishima C, Velcheti V, et al. . Phase I trial of ALT-803, a novel recombinant IL15 complex, in patients with advanced solid tumors. Clin Cancer Res 2018;24:5552–61. 10.1158/1078-0432.CCR-18-0945
    1. Romee R, Cooley S, Berrien-Elliott MM, et al. . First-in-human phase 1 clinical study of the IL-15 superagonist complex ALT-803 to treat relapse after transplantation. Blood 2018;131:10.1182/blood-2017-12-823757:2515–27. 10.1182/blood-2017-12-823757
    1. Beddall M, Chattopadhyay PK, Kao S-F, et al. . A simple tube adapter to expedite and automate thawing of viably frozen cells. J Immunol Methods 2016;439:74–8. 10.1016/j.jim.2016.08.009
    1. Naidoo J, Schindler K, Querfeld C, et al. . Autoimmune bullous skin disorders with immune checkpoint inhibitors targeting PD-1 and PD-L1. Cancer Immunol Res 2016;4:383–9. 10.1158/2326-6066.CIR-15-0123
    1. Wang R, Das T, Takou A. IgA nephropathy after pembrolizumab therapy for mesothelioma. BMJ Case Rep 2020;13:e237008. 10.1136/bcr-2020-237008
    1. Tanabe K, Kanzaki H, Wada T, et al. . Nivolumab-induced IgA nephropathy in a patient with advanced gastric cancer: a case report. Medicine 2020;99:e20464. 10.1097/MD.0000000000020464
    1. Gattinoni L, Finkelstein SE, Klebanoff CA, et al. . Removal of homeostatic cytokine sinks by lymphodepletion enhances the efficacy of adoptively transferred tumor-specific CD8+ T cells. J Exp Med 2005;202:907–12. 10.1084/jem.20050732
    1. Barrenäs F, Hansen SG, Law L, et al. . Interleukin-15 response signature predicts RhCMV/SIV vaccine efficacy. PLoS Pathog 2021;17:e1009278. 10.1371/journal.ppat.1009278
    1. Robinson TO, Schluns KS. The potential and promise of IL-15 in immuno-oncogenic therapies. Immunol Lett 2017;190:159–68. 10.1016/j.imlet.2017.08.010
    1. Knudson KM, Hodge JW, Schlom J, et al. . Rationale for IL-15 superagonists in cancer immunotherapy. Expert Opin Biol Ther 2020;20:705–9. 10.1080/14712598.2020.1738379
    1. Waldmann TA, Miljkovic MD, Conlon KC. Interleukin-15 (dys)regulation of lymphoid homeostasis: Implications for therapy of autoimmunity and cancer. J Exp Med 2020;217:e20191062. 10.1084/jem.20191062
    1. Waldmann TA, Dubois S, Miljkovic MD, et al. . Il-15 in the combination immunotherapy of cancer. Front Immunol 2020;11:868. 10.3389/fimmu.2020.00868
    1. Wrangle JM, Patterson A, Johnson CB, et al. . Il-2 and beyond in cancer immunotherapy. J Interferon Cytokine Res 2018;38:45–68. 10.1089/jir.2017.0101
    1. Wrangle JM, Velcheti V, Patel MR, et al. . ALT-803, an IL-15 superagonist, in combination with nivolumab in patients with metastatic non-small cell lung cancer: a non-randomised, open-label, phase 1B trial. Lancet Oncol 2018;19:694–704. 10.1016/S1470-2045(18)30148-7
    1. Felices M, Kodal B, Hinderlie P, et al. . Novel CD19-targeted TriKE restores NK cell function and proliferative capacity in CLL. Blood Adv 2019;3:897–907. 10.1182/bloodadvances.2018029371
    1. Vallera DA, Felices M, McElmurry R, et al. . Il15 trispecific killer Engagers (TriKE) make natural killer cells specific to CD33+ targets while also inducing persistence, in vivo expansion, and enhanced function. Clin Cancer Res 2016;22:3440–50. 10.1158/1078-0432.CCR-15-2710

Source: PubMed

3
Předplatit