Signal Transduction and Targeted Therapy (Apr 2022)

Identification of the receptor of oncolytic virus M1 as a therapeutic predictor for multiple solid tumors

  • Deli Song,
  • Xudong Jia,
  • Xincheng Liu,
  • Linyi Hu,
  • Kaiying Lin,
  • Tong Xiao,
  • Yangyang Qiao,
  • Jiayu Zhang,
  • Jia Dan,
  • Chunwa Wong,
  • Cheng Hu,
  • Ke Sai,
  • Shoufang Gong,
  • Max Sander,
  • Runling Shen,
  • Xiaoyu Chen,
  • Xiaoting Xiao,
  • Jiehong Chen,
  • Yanming Zhang,
  • Cailv Wei,
  • Xiao Xiao,
  • Jiankai Liang,
  • Qinfen Zhang,
  • Jun Hu,
  • Wenbo Zhu,
  • Guangmei Yan,
  • Yuan Lin,
  • Jing Cai

DOI
https://doi.org/10.1038/s41392-022-00921-3
Journal volume & issue
Vol. 7, no. 1
pp. 1 – 11

Abstract

Read online

Abstract Over the last decade, oncolytic virus (OV) therapy has shown its promising potential in tumor treatment. The fact that not every patient can benefit from it highlights the importance for defining biomarkers that help predict patients’ responses. As particular self-amplifying biotherapeutics, the anti-tumor effects of OVs are highly dependent on the host factors for viral infection and replication. By using weighted gene co-expression network analysis (WGCNA), we found matrix remodeling associated 8 (MXRA8) is positively correlated with the oncolysis induced by oncolytic virus M1 (OVM). Consistently, MXRA8 promotes the oncolytic efficacy of OVM in vitro and in vivo. Moreover, the interaction of MXRA8 and OVM studied by single-particle cryo-electron microscopy (cryo-EM) showed that MXRA8 directly binds to this virus. Therefore, MXRA8 acts as the entry receptor of OVM. Pan-cancer analysis showed that MXRA8 is abundant in most solid tumors and is highly expressed in tumor tissues compared with adjacent normal ones. Further study in cancer cell lines and patient-derived tumor tissues revealed that the tumor selectivity of OVM is predominantly determined by a combinational effect of the cell membrane receptor MXRA8 and the intracellular factor, zinc-finger antiviral protein (ZAP). Taken together, our study may provide a novel dual-biomarker for precision medicine in OVM therapy.