4.7 Article

Synergy of nanodiamond-doxorubicin conjugates and PD-L1 blockade effectively turns tumor-associated macrophages against tumor cells

期刊

JOURNAL OF NANOBIOTECHNOLOGY
卷 19, 期 1, 页码 -

出版社

BMC
DOI: 10.1186/s12951-021-01017-w

关键词

PD-L1/PD-1; Non-small cell lung cancer; Tumor-associated macrophages; HMGB1; RAGE/NF-kappa B signaling; Nanodiamond-doxorubicin conjugates

资金

  1. National Natural Science Foundation of China [81671818]
  2. Hubei Province health and family planning scientific research project, China [WJ2019Z010]
  3. Science and Technology Program of Wuhan, China [2017060201010148]
  4. Medical Science Advancement Program (Basic Medical Science) of Wuhan University, China [TFJC2018003]
  5. Hubei Provincial Natural Science Foundation [2020CFB152]
  6. Cultivating Project for Young Scholar at Hubei University of Medicine [2020QDJZR002]
  7. National Training Program of Innovation and Entrepreneurship for Undergraduates [202010929005, S202013249001]
  8. Innovative Research Program for Graduates of Hubei University of Medicine [YC2021016]

向作者/读者索取更多资源

The combination of Nano-DOX and BMS-1 can activate TAMs' M1-type response, inhibit tumor cell growth, and achieve synergistic therapeutic efficacy against tumors.
Background: Tumor-associated macrophages (TAMs) are the most abundant stromal cells in the tumor microenvironment. Turning the TAMs against their host tumor cells is an intriguing therapeutic strategy particularly attractive for patients with immunologically cold tumors. This concept was mechanistically demonstrated on in vitro human and murine lung cancer cells and their corresponding TAM models through combinatorial use of nanodiamond-doxorubicin conjugates (Nano-DOX) and a PD-L1 blocking agent BMS-1. Nano-DOX are an agent previously proved to be able to stimulate tumor cells' immunogenicity and thereby reactivate the TAMs into the anti-tumor M1 phenotype. Results: Nano-DOX were first shown to stimulate the tumor cells and the TAMs to release the cytokine HMGB1 which, regardless of its source, acted through the RAGE/NF-kappa B pathway to induce PD-L1 in the tumor cells and PD-L1/PD-1 in the TAMs. Interestingly, Nano-DOX also induced NF-kappa B-dependent RAGE expression in the tumor cells and thus reinforced HMGB1's action thereon. Then, BMS-1 was shown to enhance Nano-DOX-stimulated M1-type activation of TAMs both by blocking Nano-DOX-induced PD-L1 in the TAMs and by blocking tumor cell PD-L1 ligation with TAM PD-1. The TAMs with enhanced M1-type repolarization both killed the tumor cells and suppressed their growth. BMS-1 could also potentiate Nano-DOX's action to suppress tumor cell growth via blocking of Nano-DOX-induced PD-L1 therein. Finally, Nano-DOX and BMS-1 achieved synergistic therapeutic efficacy against in vivo tumor grafts in a TAM-dependent manner. Conclusions: PD-L1/PD-1 upregulation mediated by autocrine and paracrine activation of the HMGB1/RAGE/NF-kappa B signaling is a key response of lung cancer cells and their TAMs to stress, which can be induced by Nano-DOX. Blockade of Nano-DOX-induced PD-L1, both in the cancer cells and the TAMs, achieves enhanced activation of TAM-mediated anti-tumor response. Graphic abstract

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

4.7
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