Spliceosome mutations induce R loop-associated sensitivity to ATR inhibition in myelodysplastic syndromes

HD Nguyen, WY Leong, W Li, PNG Reddy, JD Sullivan… - Cancer research, 2018 - AACR
HD Nguyen, WY Leong, W Li, PNG Reddy, JD Sullivan, MJ Walter, L Zou, TA Graubert
Cancer research, 2018AACR
Heterozygous somatic mutations in spliceosome genes (U2AF1, SF3B1, ZRSR2, or SRSF2)
occur in> 50% of patients with myelodysplastic syndrome (MDS). These mutations occur
early in disease development, suggesting that they contribute to MDS pathogenesis and
may represent a unique genetic vulnerability for targeted therapy. Here, we show that RNA
splicing perturbation by expression of the U2AF1 (S34F) mutant causes accumulation of R
loops, a transcription intermediate containing RNA: DNA hybrids and displaced single …
Abstract
Heterozygous somatic mutations in spliceosome genes (U2AF1, SF3B1, ZRSR2, or SRSF2) occur in >50% of patients with myelodysplastic syndrome (MDS). These mutations occur early in disease development, suggesting that they contribute to MDS pathogenesis and may represent a unique genetic vulnerability for targeted therapy. Here, we show that RNA splicing perturbation by expression of the U2AF1(S34F) mutant causes accumulation of R loops, a transcription intermediate containing RNA:DNA hybrids and displaced single-stranded DNA, and elicits an ATR response. ATR inhibitors (ATRi) induced DNA damage and cell death in U2AF1(S34F)-expressing cells, and these effects of ATRi were enhanced by splicing modulating compounds. Moreover, ATRi-induced DNA damage was suppressed by overexpression of RNaseH1, an enzyme that specifically removes the RNA in RNA:DNA hybrids, suggesting that the ATRi sensitivity of U2AF1(S34F)-expressing cells arises from R loops. Taken together, our results demonstrate that ATR may represent a novel therapeutic target in patients with MDS carrying the U2AF1(S34F) mutation and potentially other malignancies harboring spliceosome mutations.
Significance: This study provides preclinical evidence that patients with MDS or other myeloid malignancies driven by spliceosome mutations may benefit from ATR inhibition to exploit the R loop–associated vulnerability induced by perturbations in splicing. Cancer Res; 78(18); 5363–74. ©2018 AACR.
AACR