Abstract

Triple negative breast cancer (TNBC) is an aggressive subset for which effective therapeutic approaches are needed. A significant proportion of TNBC patients harbor either germline or somatic mutations in BRCA1, or epigenetic silencing of BRCA1, which renders them deficient in DNA repair. Virtually all BRCA1 deficient breast cancers harbor mutations in TP53 suggesting that inactivation of p53 is a requirement for tumor progression in the setting of BRCA1 deficiency. Due to this dependency, we hypothesized that restoring wild type p53 function in BRCA1 deficient breast cancer would be therapeutic. The majority of TP53 mutations are missense, which generate a defective protein that potentially can be targeted with small molecules. Zinc metallochaperones (ZMCs) are a new class of anti-cancer drugs that specifically reactivate zinc-deficient mutant p53 by restoring zinc binding. Using ZMC1 in human breast cancer cell lines expressing the zinc deficient p53R175H, we demonstrate that loss of BRCA1 sensitizes cells to mutant p53 reactivation. Using murine breast cancer models with Brca1 deficiency, we demonstrate that ZMC1 significantly improves survival of mice bearing tumors harboring the zinc-deficient Trp53R172H allele but not the Trp53−/− allele. We synthesized a new formulation of ZMC1 (Zn-1), in which the drug is made in complex with zinc to improve zinc delivery, and demonstrate that Zn-1 has increased efficacy. Furthermore, we show that ZMC1 plus olaparib is a highly effective combination for p53R172H tumor growth inhibition. In conclusion, we have validated preclinically a new therapeutic approach for BRCA1 deficient breast cancer through reactivation of mutant p53.

Highlights

  • Triple negative breast cancers (TNBC) are an aggressive subset of human breast cancer for which chemotherapy remains the standard treatment approach

  • We have previously demonstrated that several structurally related members of the thiosemicarbazone family of metal ion chelators function to reactivate mutant p53 and as such we have classified them as Zinc metallochaperones (ZMCs) (ZMC1, 2, 3).[14,22]

  • The first part involves the binding of zinc extracellularly in a 2:1 ratio of compound to zinc and freely diffusing across the plasma membrane as a zinc ionophore to deliver it to mutant p53 (Fig. 1a)

Read more

Summary

INTRODUCTION

Triple negative breast cancers (TNBC) are an aggressive subset of human breast cancer for which chemotherapy remains the standard treatment approach. TP53 mutations include inhibition of complementary cell cycle checkpoints mediated by WEE1 and Chk[1], but these strategies only indirectly target p53 defect.[12,13] A more attractive approach would be to directly restore wild type p53 function in a BRCA1 deficient breast cancer. This approach remains untested due to the lack of clinically effective mutant p53 reactivators. That restoring wild type p53 function in BRCA1 mutant tumors could be a highly effective therapeutic strategy

RESULTS
Na et al 4
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