Abstract

Abstract Although recombinant human IL-2 (rhIL-2, aldesleukin) produced approximately a 15% objective response rate in patients with renal cell carcinoma (RCC) and metastatic melanoma, its IL-2Rα (CD25)-biased binding and short half-life (t1/2 in human: 13-85 min) require a high dose and frequent dosing interval, leading to systemic toxicity such as vascular leak syndrome (VLS) and cytokine release syndrome (CRS). To overcome this limitation, various research groups are developing engineered IL-2 muteins with abolished CD25 binding affinity. While such IL-2 muteins may reduce CD25-mediated toxicity, there is a risk of a lopsided immune response. Furthermore, concomitantly reduced affinity to IL-2Rβ (CD122) may cause insufficient anti-tumor activity. We are developing HM16390, a long-acting IL-2 analog with a significantly increased binding affinity to CD122 for potent tumor killing effect. Simultaneously, optimal binding property to CD25 was introduced for a suppression of uncontrolled systemic immune response via peripheral Treg expansion. From these novel sequence modifications, HM16390 exhibited approximately 14-fold potent activity in human NK and CD8+ T cells compared to rhIL-2 and comparable activity with rhIL-2 in human Tregs (EC50 of STAT5 phosphorylation) without undesired release of cytokines associated with CRS. In B16F10 mouse model, HM16390 has a longer-lasting PK property (t1/2: 14.2~26.0 hr) after single subcutaneous (SC) administration. CD8+ T cells were significantly increased in peripheral blood along with a transient increase of peripheral Tregs, suggesting that HM16390 not only has a significant effect on expansion of CD8+ T cells but can simultaneously modulate exaggerated peripheral immune response. It is also noteworthy that, unlike in the peripheral, Tregs were drastically reduced in the tumor immune microenvironment, and CD8+ T cells were significantly increased. Next, in orthotopic model of mouse RCC (RENCA-luc cell implantation), across treatment groups, 40%(n=4/10) to 80%(n=8/10) of animals achieved complete tumor regression in dose-dependent manner after once weekly SC injection of HM16390 for 4 weeks. Furthermore, tumor nodules were not observed in lung and kidney after intravenous (IV) rechallenge of RENCA cells in the mice cured by HM16390 for at least 4 months after drug discontinuation. This indicates a long-term immunological memory response of generated memory cells by treatment of HM16390. In conclusion, HM16390 demonstrates potent and durable anti-tumor activity in various murine tumor models via its enhanced CD122 binding affinity, and incorporated optimal CD25 binding affinity may alleviate systemic toxicity by immunomodulatory role of peripheral Tregs. Citation Format: Sol-Bi Shin, Jinyoung Kim, Jaehyuk Choi, Seongju Jeong, Yunjae Kim, Jooyun Byun, Sungmin Bae, Daejin Kim, In Young Choi. HM16390, a novel long-acting IL-2 analog with fine-tuned binding affinities to IL-2 receptor subunits for favorable safety profile, exhibits potent tumor killing effect in the various tumor syngeneic models [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2024; Part 2 (Late-Breaking, Clinical Trial, and Invited Abstracts); 2024 Apr 5-10; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2024;84(7_Suppl):Abstract nr LB118.

Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call