4.7 Article

Extracellular vesicles and PD-L1 suppress macrophages, inducing therapy resistance in TP53-deficient B-cell malignancies

期刊

BLOOD
卷 139, 期 25, 页码 3617-3629

出版社

AMER SOC HEMATOLOGY
DOI: 10.1182/blood.2021014007

关键词

-

资金

  1. German research foundation (DFG) [KFO 286, CRC1530]
  2. 'Foerderprogramm Nachwuchsforschungsgruppen NRW 2015-2021
  3. CAP Program of the Center for Molecular Medicine Cologne
  4. Gilead Sciences
  5. German-Israeli Foundation for Research and Development [I-65-412.20-2016]
  6. Deutsche Forschungsgemeinschaft [KFO-286-RP2]
  7. Deutsche Jose Carreras Leukamie Stiftung [R12/08]
  8. Else Kroner-Fresenius Stiftung [EKFS-2014-A06, 2016_Kolleg.19]
  9. Deutsche Krebshilfe [1117240, 70113041]
  10. German Ministry of Education and Research (BMBF) [e:Med 01ZX1303A]
  11. Wilhelm-Sander Foundation
  12. German Research Foundation [DFG BR 4775/2-1]

向作者/读者索取更多资源

This study reveals that genetic alterations in the DNA damage response pathway are a common mechanism of resistance to chemoimmunotherapy in B-cell malignancies. Loss of TP53 inhibits macrophage phagocytic capacity and affects the release of extracellular vesicles (EVs) and PD-L1 expression, leading to resistance to chemoimmunotherapy. These findings highlight the importance of using checkpoint inhibition in the combination treatment of B-cell malignancies with TP53 loss.
Genetic alterations in the DNA damage response (DDR) pathway are a frequent mechanism of resistance to chemoimmunotherapy (CIT) in B-cell malignancies. We have previously shown that the synergy of CIT relies on secretory crosstalk elicited by chemotherapy between the tumor cells and macrophages. Here, we show that loss of multiple different members of the DDR pathway inhibits macrophage phagocytic capacity in vitro and in vivo. Particularly, loss of TP53 led to decreased phagocytic capacity ex vivo across multiple B-cell malignancies. We demonstrate via in vivo cyclophosphamide treatment using the Em-TCL1 mouse model that loss of macrophage phagocytic capacity in Tp53-deleted leukemia is driven by a significant downregulation of a phagocytic transcriptomic signature using small conditional RNA sequencing. By analyzing the tumor B-cell proteome, we identified a TP53-specific upregulation of proteins associated with extracellular vesicles (EVs). We abrogated EV biogenesis in tumor B-cells via clustered regularly interspaced short palindromic repeats (CRISPR)-knockout (KO) of RAB27A and confirmed that the EVs from TP53-deleted lymphoma cells were responsible for the reduced phagocytic capacity and the in vivo CIT resistance. Furthermore, we observed that TP53 loss led to an upregulation of both PD-L1 cell surface expression and secretion of EVs by lymphoma cells. Disruption of EV bound PD-L1 by anti-PD-L1 antibodies or PD-L1 CRISPR-KO improved macrophage phagocytic capacity and in vivo therapy response. Thus, we demonstrate enhanced EV release and increased PD-L1 expression in TP53-deficient B-cell lymphomas as novel mechanisms of macrophage function alteration in CIT resistance. This study indicates the use of checkpoint inhibition in the combination treatment of B-cell malignancies with TP53 loss.

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