Abstract

PurposePhosphate and tensin homolog (PTEN), a negative regulator of PI3K signaling, is involved in DNA repair. ATR is a key sensor of DNA damage and replication stress. We evaluated whether ATR signaling has clinical significance and could be targeted by synthetic lethality in PTEN-deficient triple-negative breast cancer (TNBC).MethodsPTEN, ATR and pCHK1Ser345 protein level was evaluated in 1650 human breast cancers. ATR blockade by VE-821 was investigated in PTEN-proficient- (MDA-MB-231) and PTEN-deficient (BT-549, MDA-MB-468) TNBC cell lines. Functional studies included DNA repair expression profiling, MTS cell-proliferation assay, FACS (cell cycle progression & γH2AX accumulation) and FITC-annexin V flow cytometry analysis.ResultsLow nuclear PTEN was associated with higher grade, pleomorphism, de-differentiation, higher mitotic index, larger tumour size, ER negativity, and shorter survival (p values < 0.05). In tumours with low nuclear PTEN, high ATR and/or high pCHK1ser345 level was also linked to higher grade, larger tumour size and poor survival (all p values < 0.05). VE-821 was selectively toxic in PTEN-deficient TNBC cells and resulted in accumulation of double-strand DNA breaks, cell cycle arrest, and increased apoptosis.ConclusionATR signalling adversely impact survival in PTEN-deficient breast cancers. ATR inhibition is synthetically lethal in PTEN-deficient TNBC cells.

Highlights

  • Phosphate and tensin homolog (PTEN) is a key tumor suppressor [5, 7, 14, 22]

  • The loss of nuclear PTEN expression was observed in 508/811 (62.6%) tumours compared with 303/811 (37.4%)

  • We provide the first clinical evidence that Ataxia telangiectasia-mutatedand Rad3-related protein (ATR) signaling could have adverse impact on survival in PTEN-deficient breast cancers

Read more

Summary

Introduction

Phosphate and tensin homolog (PTEN) is a key tumor suppressor [5, 7, 14, 22]. Mutations in PTEN have been reported in several tumours including brain, prostrate, melanoma, endometrial and breast cancers. Nuclear PTEN has been shown to have roles in DNA repair [5, 7, 14, 22]. Activated ATR in turn phosphorylates Chk at S­ er345 and S­ er317, as well as several other target proteins involved in homologous recombination repair and. Phosphorylation of Chk at ­Ser345 (pChk1) leads to its activation which further coordinates cell cycle progression and DNA repair [9, 10, 19, 20]. We provide evidence that ATR and pChk expression in of PTEN-deficient breast cancer adversely impact on survival. ATR inhibition is synthetically lethal in PTEN-deficient TNBC cells implying a promising personalized therapy approach in breast cancers

Patients and methods
Evaluation of immunohistochemical staining
Results
Discussion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call