Abstract

Telomere stability is important for cell viability, as cells with telomere DNA damage that is not repaired do not survive. We reported previously that androgen receptor (AR) antagonist induces telomere DNA damage in androgen-sensitive LNCaP prostate cancer cells; this triggers a DNA damage response (DDR) at telomeres that includes activation of ATM, and blocking ATM activation prevents telomere DNA repair and leads to cell death. Remarkably, AR antagonist induces telomere DNA damage and triggers ATM activation at telomeres also in 22Rv1 castration-resistant prostate cancer (CRPC) cells that are not growth inhibited by AR antagonist. Treatment with AR antagonist enzalutamide (ENZ) or ATM inhibitor (ATMi) by itself had no effect on growth in vitro or in vivo, but combined treatment with ENZ plus ATMi significantly inhibited cell survival in vitro and tumor growth in vivo. By inducing telomere DNA damage and activating a telomere DDR, an opportunity to inhibit DNA repair and promote cell death was created, even in CRPC cells. 22Rv1 cells express both full-length AR and AR splice variant AR-V7, but full-length AR was found to be the predominant form of AR associated with telomeres and required for telomere stability. Although 22Rv1 growth of untreated 22Rv1 cells appears to be driven by AR-V7, it is, ironically, expression of full-length AR that makes them sensitive to growth inhibition by combined treatment with ENZ plus ATMi. Notably, this combined treatment approach to induce telomere DNA damage and inhibit the DDR was effective in inducing cell death also in other CRPC cell lines (LNCaP/AR and C4-2B). Thus, the use of ENZ in combination with a DDR inhibitor, such as ATMi, may be effective in prolonging disease-free survival of patients with AR-positive metastatic CRPC, even those that co-express AR splice variant.

Highlights

  • The critical role of the androgen receptor (AR) in prostate cancer cell proliferation and survival is the enduring basis for treating advanced prostate cancer with drugs that block AR function or androgen biosynthesis [1, 2]

  • Because full-length AR is expressed in 80–100% of AR-positive castration-resistant prostate cancer (CRPC) metastases [18, 45], and because full-length AR, not AR-V7, is associated with telomeres and is critical for telomere stability, CRPC metastases are expected to be vulnerable to telomere DNA damage by AR antagonist, and activation of a telomere DNA damage response (DDR) that can be inhibited by an ATM inhibitor (ATMi) to promote cell death, as demonstrated in CRPC 22Rv1 cells and tumors, and in CRPC cell lines C4-2B and LNCaP/AR

  • Our data suggest that the use of ENZ in combination with a DDR inhibitor, such as an ATMi, may be effective in prolonging disease-free survival of patients with AR-positive mCRPC, even the 19–59% that co-express an AR splice variant [18, 45]

Read more

Summary

Introduction

The critical role of the androgen receptor (AR) in prostate cancer cell proliferation and survival is the enduring basis for treating advanced prostate cancer with drugs that block AR function or androgen biosynthesis [1, 2]. CRPC still relies on AR [4, 5], indicating a need to more fully understand the role of AR in cell survival. In this regard, we have discovered a role of AR in prostate cancer cell telomere stability [6, 7]. We have discovered a role of AR in prostate cancer cell telomere stability [6, 7] Inactivation of this role of AR creates a DNA damage response (DDR) target, inactivation of which blocks repair and promotes cell death [8]. Telomere stability is important for cell viability, and telomere DNA damage creates an opportunity to inhibit telomere DNA repair and activate cell death [8]

Methods
Results
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