Genome Medicine (Sep 2021)

The genomic architecture of EBV and infected gastric tissue from precursor lesions to carcinoma

  • Zhang-Hua Chen,
  • Shu-Mei Yan,
  • Xi-Xi Chen,
  • Qi Zhang,
  • Shang-Xin Liu,
  • Yang Liu,
  • Yi-Ling Luo,
  • Chao Zhang,
  • Miao Xu,
  • Yi-Fan Zhao,
  • Li-Yun Huang,
  • Bin-Liu Liu,
  • Tian-Liang Xia,
  • Da-Zhi Xu,
  • Yao Liang,
  • Yong-Ming Chen,
  • Wei Wang,
  • Shu-Qiang Yuan,
  • Hui-Zhong Zhang,
  • Jing-Ping Yun,
  • Wei-Wei Zhai,
  • Mu-Sheng Zeng,
  • Fan Bai,
  • Qian Zhong

DOI
https://doi.org/10.1186/s13073-021-00963-2
Journal volume & issue
Vol. 13, no. 1
pp. 1 – 22

Abstract

Read online

Abstract Background Epstein-Barr virus (EBV)-associated gastric carcinomas (EBVaGCs) present unique molecular signatures, but the tumorigenesis of EBVaGCs and the role EBV plays during this process remain poorly understood. Methods We applied whole-exome sequencing, EBV genome sequencing, and whole-genome bisulfite sequencing to multiple samples (n = 123) derived from the same patients (n = 25), which covered saliva samples and different histological stages from morphologically normal epithelial tissues to dysplasia and EBVaGCs. We compared the genomic landscape between EBVaGCs and their precursor lesions and traced the clonal evolution for each patient. We also analyzed genome sequences of EBV from samples of different histological types. Finally, the key molecular events promoting the tumor evolution were demonstrated by MTT, IC50, and colony formation assay in vitro experiments and in vivo xenograft experiments. Results Our analysis revealed increasing mutational burden and EBV load from normal tissues and low-grade dysplasia (LD) to high-grade dysplasia (HD) and EBVaGCs, and oncogenic amplifications occurred late in EBVaGCs. Interestingly, within each patient, EBVaGCs and HDs were monoclonal and harbored single-strain-originated EBV, but saliva or normal tissues/LDs had different EBV strains from that in EBVaGCs. Compared with precursor lesions, tumor cells showed incremental methylation in promotor regions, whereas EBV presented consistent hypermethylation. Dominant alterations targeting the PI3K-Akt and Wnt pathways were found in EBV-infected cells. The combinational inhibition of these two pathways in EBV-positive tumor cells confirmed their synergistic function. Conclusions We portrayed the (epi) genomic evolution process of EBVaGCs, revealed the extensive genomic diversity of EBV between tumors and normal tissue sites, and demonstrated the synergistic activation of the PI3K and Wnt pathways in EBVaGCs, offering a new potential treatment strategy for this disease.