Tumor-selective activity of XTX202, a protein-engineered IL-2, in mice without peripheral toxicities in nonhuman primates

Abstract only 2563 Background: High-dose recombinant human interleukin-2 (aldesleukin) elicits anti-tumor immunity and is approved for the treatment of renal cell carcinoma and melanoma based on durable complete remissions. However, use of aldesleukin is limited due to treatment-related life-threate...

Full description

Saved in:
Bibliographic Details
Published inJournal of clinical oncology Vol. 39; no. 15_suppl; p. 2563
Main Authors O'Neil, Jennifer, Guzman, Wilson, Yerov, Oleg, Johnson, Parker, Fanny, Manoussa, Greene, Justin, McLaughlin, Megan, Jenkins, Kurt, O'Donnell, Rebekah, Qiu, Huawei, Nicholson, Benjamin, Avery, William, O'Hagan, Ronan C.
Format Journal Article
LanguageEnglish
Published 20.05.2021
Online AccessGet full text

Cover

Loading…
More Information
Summary:Abstract only 2563 Background: High-dose recombinant human interleukin-2 (aldesleukin) elicits anti-tumor immunity and is approved for the treatment of renal cell carcinoma and melanoma based on durable complete remissions. However, use of aldesleukin is limited due to treatment-related life-threatening toxicities. Recent second-generation efforts to alleviate toxicities have largely focused on eliminating binding to IL-2Rα, often with half-life extension. We have determined that mice and non-human primates (NHPs) treated with a second generation IL-2 surrogate still experience characteristic dose-limiting toxicities, including vascular leak syndrome. To overcome these toxicities and improve the therapeutic index (TI) of IL-2 as an anti-tumor immunotherapy, we employed protein engineering to generate XTX202, a highly potent third generation masked IL-2. XTX202 is unmasked in the tumor microenvironment by proteolytic activation resulting in full restoration of binding to IL-2Rβ without binding to IL-2Rα. The current study characterizes the therapeutic index of XTX202 versus aldesleukin and a second generation IL-2 surrogate. Methods: XTX202 bioactivity was measured using STAT-5 phosphorylation in human PBMCs and reporter cell lines. Anti-tumor efficacy and peripheral immune activation were evaluated in mice bearing syngeneic tumor models. Safety was evaluated in rodents and Cynomolgus monkeys. XTX200, an unmasked half-life extended IL-2 that does not bind to IL-2Rα, was used as a surrogate second generation IL-2. Results: Masked XTX202 showed limited IL-2R-dependent STAT-5 signaling in vitro. Proteolytic activation of XTX202 resulted in CD8 + T and NK cell activation and over 1000-fold reduction in Treg activation as compared to WT IL-2. XTX202 achieved potent tumor growth inhibition in syngeneic mouse models as a single agent with no evidence of toxicity or peripheral immune activation, thus demonstrating tumor selective activity. XTX202 efficacy in mice at 2 mg/kg dose was equivalent to that achieved with the MTD dose of 0.5 mg/kg of a second generation IL-2 surrogate. XTX202 was well tolerated in NHPs in a 4-week repeat dose study at doses up to 30 mg/kg QW whereas a second generation IL-2 surrogate was not tolerated beyond 0.7 mg/kg QW. Based on these data, XTX202 has a 10 fold improvement in TI vs second generation IL-2. Based on comparative efficacy studies with aldesleukin and literature NHP tolerability data, XTX202 is projected to have a ≥150 fold greater TI than aldesleukin. Conclusions: XTX202, a third generation, tumor-selective IL-2, inhibits tumor growth and is well tolerated in repeat dose studies in NHPs at high doses. GLP toxicity studies with XTX202 are underway and first-in-human studies are expected to initiate this year. XTX202 has the potential to be a best-in-class IL-2 immunotherapy by expanding the curative anti-tumor activity of IL-2 while minimizing dose-limiting toxicities.
ISSN:0732-183X
1527-7755
DOI:10.1200/JCO.2021.39.15_suppl.2563