Abstract

IntroductionInhibition of phosphatidylinositol-3-kinase (PI3K) induces apoptosis when combined with estrogen deprivation in estrogen receptor (ER)-positive breast cancer. The aims of the present study were to identify effective PI3K pathway inhibitor and endocrine therapy combinations, to evaluate the effect of PI3K pathway mutations and estrogen dependency on tumor response, and to determine the relevance of PIK3CA mutation in recurrent disease.MethodsThe PI3K catalytic subunit inhibitor BKM120, the mammalian target of rapamycin (mTOR) inhibitor RAD001 and the dual PI3K/mTOR inhibitor BGT226 were tested against ER-positive breast cancer cell lines before and after long-term estrogen deprivation (LTED). The impact of estradiol deprivation and the ER downregulator fulvestrant on PI3K pathway inhibitor-induced apoptosis was assessed. PIK3CA hotspot mutation analysis was performed in 51 recurrent or metastatic breast cancers and correlated with ER status and survival.ResultsDrug-induced apoptosis was most marked in short-term estrogen-deprived cells with PIK3CA mutation and phosphatase and tensin homolog loss. Apoptosis was most highly induced by BGT226, followed by BKM120, and then RAD001. Estradiol antagonized PI3K inhibitor-induced apoptosis following short-term estrogen deprivation, emphasizing a role for estrogen-deprivation therapy in promoting PI3K inhibitor activity in the first-line setting. ER-positive MCF7 LTED cells exhibited relative resistance to PI3K pathway inhibition that was reversed by fulvestrant. In contrast, T47D LTED cells exhibited ER loss and ER-independent PI3K agent sensitivity. PIK3CA mutation was prevalent in relapsed ER-positive disease (48%) and was associated with persistent ER positivity and a late relapse pattern.ConclusionsEstrogen deprivation increased the apoptotic effects of PI3K and dual PI3K/mTOR inhibitors in ER-positive disease, providing a rationale for PI3K/aromatase inhibitor combinations as first-line therapy. In LTED cells, differential effects on ER expression may be a relevant consideration. When ER was persistently expressed, fulvestrant strongly promoted PI3K drug activity. When ER was lost, PI3K inhibitor monotherapy was sufficient to induce high-level apoptosis. Although tumors with PIK3CA mutation had a late recurrence pattern, these mutations were common in metastatic disease and were most often associated with persistent ER expression. Targeting PIK3CA mutant tumors with a PI3K pathway inhibitor and fulvestrant is therefore a feasible strategy for aromatase-inhibitor-resistant ER-positive relapsed breast cancer.

Highlights

  • Inhibition of phosphatidylinositol-3-kinase (PI3K) induces apoptosis when combined with estrogen deprivation in estrogen receptor (ER)-positive breast cancer

  • Expression and activation of PI3K pathway proteins in breast cancer cells To assess PI3K signaling activity in the panel of breast cancer cells used for the present investigation, the levels of phosphorylated forms of AKT, S6 protein kinase 1 and S6, and the expression of PI3K catalytic subunit isoforms, phosphatase and tensin homolog (PTEN), AKT isoforms and mammalian target of rapamycin (mTOR) were examined (Figure 1)

  • Consistent with previous studies, high levels of phospho-Ser473 Akt (p-Akt) were present in cells with PIK3CA kinase domain mutation (T47D), PTEN mutation (MDA-MB-415, ZR75-1 and CAMA-1), human epidermal growth factor receptor 2 (HER2) amplification (HCC1419, SK-BR-3) [9,10,11] and the heregulin-dependent MDA-MB-175 cell line

Read more

Summary

Introduction

Inhibition of phosphatidylinositol-3-kinase (PI3K) induces apoptosis when combined with estrogen deprivation in estrogen receptor (ER)-positive breast cancer. Genes in the PI3K pathway are frequently mutated or amplified in ER-positive breast cancer, suggesting that hyperactivation of PI3K signaling is a key target that, if effectively inhibited, could improve outcomes [4]. We have already shown that estrogen deprivation in combination with PI3K inhibition by RNA interference induces synthetic lethality and promotes cell death in ER-positive breast cancer cell lines [5], providing a rational for combination approaches that target the ER and PI3K pathways simultaneously. ER-positive breast cancers are genetically heterogeneous, and cell-intrinsic factors may modulate sensitivity to this approach It is unclear whether mutations in PI3K pathway proteins - especially in PIK3CA, the gene that encodes the PI3Ka catalytic subunit - sensitize tumors to this strategy. If this is the case, PIK3CA mutations would be expected to be rare in advanced disease and less relevant as a therapeutic target in this setting

Objectives
Methods
Results
Discussion
Conclusion
Full Text
Paper version not known

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

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.