4.8 Article

Improved intratumoral penetration of IL12 immunocytokine enhances the antitumor efficacy

期刊

FRONTIERS IN IMMUNOLOGY
卷 13, 期 -, 页码 -

出版社

FRONTIERS MEDIA SA
DOI: 10.3389/fimmu.2022.1034774

关键词

immunocytokine; IL12; solid tumor; tumor penetration; binding kinetics; T cell activation

资金

  1. Dragonfly Therapeutics, Inc.
  2. Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI) - Ministry of Health Welfare [HR16C0001]
  3. National Research Foundation of Korea (NRF) - Ministry of Science, ICT & Future Planning, Republic of Korea [2019R1A6A1A11051471]

向作者/读者索取更多资源

Tumor-targeting antibody-fused cytokines can enhance antitumor efficacy and reduce toxicity by delivering cytokines to tumors. However, the localization and penetration of these immunocytokines within solid tumors pose challenges. This study found that optimizing the tumor antigen-binding kinetics and affinity of the antibody moiety is crucial for achieving maximal antitumor efficacy.
Tumor-targeting antibody (Ab)-fused cytokines, referred to as immunocytokines, are designed to increase antitumor efficacy and reduce toxicity through the tumor-directed delivery of cytokines. However, the poor localization and intratumoral penetration of immunocytokines, especially in solid tumors, pose a challenge to effectively stimulate antitumor immune cells to kill tumor cells within the tumor microenvironment. Here, we investigated the influence of the tumor antigen-binding kinetics of a murine interleukin 12 (mIL12)-based immunocytokine on tumor localization and diffusive intratumoral penetration, and hence the consequent antitumor activity, by activating effector T cells in immunocompetent mice bearing syngeneic colon tumors. Based on tumor-associated antigen HER2-specific Ab Herceptin (HCT)-fused mIL12 carrying one molecule of mIL12 (HCT-mono-mIL12 immunocytokine), we generated a panel of HCT-mono-mIL12 variants with different affinities (K-D) mainly varying in their dissociation rates (k(off)) for HER2. Systemic administration of HCT-mono-mIL12 required an anti-HER2 affinity above a threshold (K-D = 130 nM) for selective localization and antitumor activity to HER2-expressing tumors versus HER2-negative tumors. However, the high affinity (K-D = 0.54 or 46 nM) due to the slow k(off) from HER2 antigen limited the depth of intratumoral penetration of HCT-mono-mIL12 and the consequent tumor infiltration of T cells, resulting in inferior antitumor activity compared with that of HCT-mono-mIL12 with moderate affinity of (K-D = 130 nM) and a faster k(off). The extent of intratumoral penetration of HCT-mono-mIL12 variants was strongly correlated with their tumor infiltration and intratumoral activation of CD4(+) and CD8(+) T cells to kill tumor cells. Collectively, our results demonstrate that when developing antitumor immunocytokines, tumor antigen-binding kinetics and affinity of the Ab moiety should be optimized to achieve maximal antitumor efficacy.

作者

我是这篇论文的作者
点击您的名字以认领此论文并将其添加到您的个人资料中。

评论

主要评分

4.8
评分不足

次要评分

新颖性
-
重要性
-
科学严谨性
-
评价这篇论文

推荐

暂无数据
暂无数据