Genome Medicine (Aug 2024)

Large-scale copy number alterations are enriched for synthetic viability in BRCA1/BRCA2 tumors

  • Yingjie Zhu,
  • Xin Pei,
  • Ardijana Novaj,
  • Jeremy Setton,
  • Daniel Bronder,
  • Fatemeh Derakhshan,
  • Pier Selenica,
  • Niamh McDermott,
  • Mehmet Orman,
  • Sarina Plum,
  • Shyamal Subramanyan,
  • Sara H. Braverman,
  • Biko McMillan,
  • Sonali Sinha,
  • Jennifer Ma,
  • Andrea Gazzo,
  • Atif Khan,
  • Samuel Bakhoum,
  • Simon N. Powell,
  • Jorge S. Reis-Filho,
  • Nadeem Riaz

DOI
https://doi.org/10.1186/s13073-024-01371-y
Journal volume & issue
Vol. 16, no. 1
pp. 1 – 17

Abstract

Read online

Abstract Background Pathogenic BRCA1 or BRCA2 germline mutations contribute to hereditary breast, ovarian, prostate, and pancreatic cancer. Paradoxically, bi-allelic inactivation of BRCA1 or BRCA2 (bBRCA1/2) is embryonically lethal and decreases cellular proliferation. The compensatory mechanisms that facilitate oncogenesis in bBRCA1/2 tumors remain unclear. Methods We identified recurrent genetic alterations enriched in human bBRCA1/2 tumors and experimentally validated if these improved proliferation in cellular models. We analyzed mutations and copy number alterations (CNAs) in bBRCA1/2 breast and ovarian cancer from the TCGA and ICGC. We used Fisher’s exact test to identify CNAs enriched in bBRCA1/2 tumors compared to control tumors that lacked evidence of homologous recombination deficiency. Genes located in CNA regions enriched in bBRCA1/2 tumors were further screened by gene expression and their effects on proliferation in genome-wide CRISPR/Cas9 screens. A set of candidate genes was functionally validated with in vitro clonogenic survival and functional assays to validate their influence on proliferation in the setting of bBRCA1/2 mutations. Results We found that bBRCA1/2 tumors harbor recurrent large-scale genomic deletions significantly more frequently than histologically matched controls (n = 238 cytobands in breast and ovarian cancers). Within the deleted regions, we identified 277 BRCA1-related genes and 218 BRCA2-related genes that had reduced expression and increased proliferation in bBRCA1/2 but not in wild-type cells in genome-wide CRISPR screens. In vitro validation of 20 candidate genes with clonogenic proliferation assays validated 9 genes, including RIC8A and ATMIN (ATM-Interacting protein). We identified loss of RIC8A, which occurs frequently in both bBRCA1/2 tumors and is synthetically viable with loss of both BRCA1 and BRCA2. Furthermore, we found that metastatic homologous recombination deficient cancers acquire loss-of-function mutations in RIC8A. Lastly, we identified that RIC8A does not rescue homologous recombination deficiency but may influence mitosis in bBRCA1/2 tumors, potentially leading to increased micronuclei formation. Conclusions This study provides a means to solve the tumor suppressor paradox by identifying synthetic viability interactions and causal driver genes affected by large-scale CNAs in human cancers.

Keywords