体内
阻塞(统计)
阻断抗体
抗体
癌症研究
体外
化学
药理学
医学
免疫学
计算机科学
生物
生物化学
计算机网络
生物技术
作者
Ouri Cohen,Safra Rudnick‐Glick,Ivana Savic Azoulay,Tania Fine,Ela Elyada,Lihi Radomir,Kiryu K. Yap,Debasish Sen,Michele L. Kus,Nicole Otoc,Tetsuya Taura,Trang Vuong,Nurit Ashkenazi,Jason H. Gill,Anthony Doyle,David S. Wilson,Inbal Ben‐Eliezer,Paul M. Ayton,Ianai Fishbein
标识
DOI:10.1136/jitc-2024-sitc2024.0800
摘要
Background
IL-2 plays a critical role in regulating the immune system by stimulating the proliferation and activation of T cells, and thus has the potential to enhance anti-tumor immunity. Nevertheless, the clinical use of IL-2 is severely limited by significant systemic toxicity and a narrow therapeutic window. To overcome these limitations, we developed an antibody-cytokine fusion protein, termed anti-PD1-IL2Att, that is composed of an anti-PD1 antibody fused to a highly attenuated variant of IL-2. This construct was designed to deliver attenuated IL-2 specifically to PD-1-expressing cells. Anti-PD1-IL2Att selectively stimulates PD-1+ effector T cells, while not inhibiting PD-1 receptor function. Since PD-1 expression is significantly higher in tumor-infiltrating T cells compared to circulating and tissue-resident T cells, targeting IL-2 to PD-1+ T cells may boost anti-tumor immunity, while avoiding systemic IL-2-related adverse effects. Here we describe the impact of anti-PD1-IL2Att activity on tumor progression and tumor-infiltrating lymphocytes in pre-clinical tumor models. Methods
The anti-tumor effect of anti-PD1-IL2Att was evaluated using a mouse surrogate antibody-cytokine fusion protein (mAPD1-IL2Att) in a mouse model of MC38 colorectal adenocarcinoma in a dose escalation study, a repeated-dose administration study and a long-term immunity rechallenge study. Additionally, the ability of a human anti-PD1-IL2Att, TEV-56278, to induce anti-tumor immune responses was evaluated in humanized immune-system mouse models engrafted with human melanoma tumor cells. All models employed flow cytometry to characterize T cell population variations within the tumor microenvironment after treatment. Results
Administration of mAPD1-IL2Att in the MC38 mouse model resulted in dose-dependent reduction in tumor growth following single dose administration and complete tumor regression following repeated administration. Furthermore, mice responsive to mAPD1-IL2Att treatment were fully protected from secondary tumor re-challenge, indicating induction of long-term anti-tumor immunity. Additionally, TEV-56278 administration to humanized immune system mice engrafted with human melanoma tumors led to tumor growth inhibition similar to that observed in the MC38 model. Flow cytometry analysis of tumor-infiltrating lymphocytes revealed a shift in the immune response in both models following treatment. Specifically, we observed a significant increase in the proportion of effector T cells, accompanied by a decrease in regulatory T cells. Moreover, in the MC38 model terminally exhausted CD8+ T cells (CD8+CD44+CD62L-CD69+ki67+PD-1+Tim3+) were decreased following treatment. These findings collectively suggest a favorable immune-activated anti-tumor response. Conclusions
Anti-PD1-IL2Att treatment led to tumor regression, increased infiltration of T cells into the tumor, and establishment of durable immune memory. Our preclinical models highlight the potential of anti-PD1-IL2Att as an effective anti-tumor immunotherapy. Ethics Approval
The in vivo animal studies described in the abstract were approved by the following ethics review boards: IACUC #: NPC-TE-IL-2307-447 (Israel Ministry of Health); IACUC ASP #: 980701 (Charles River) and IACUC ASP #: 980702 (Charles River).
科研通智能强力驱动
Strongly Powered by AbleSci AI