Pigment Epithelium-Derived Factor Alleviates Tamoxifen-Induced Endometrial Hyperplasia

Mol Cancer Ther. 2015 Dec;14(12):2840-9. doi: 10.1158/1535-7163.MCT-15-0523. Epub 2015 Oct 8.

Abstract

Tamoxifen is a cornerstone component of adjuvant endocrine therapy for patients with hormone-receptor-positive breast cancer. Its significant adverse effects include uterine hyperplasia, polyps, and increased risk of endometrial cancer. However, the underlying molecular mechanism remains unclear. Excessive angiogenesis, a hallmark of tumorigenesis, is a result of disrupted balance between pro- and anti-angiogenic factors. VEGF is a pro-angiogenic factor shown to be elevated by tamoxifen in the uterus. Pigment epithelium-derived factor (PEDF) is a potent anti-angiogenic factor that suppresses strong pro-angiogenic factors, such as VEGF. Our aim was to investigate whether angiogenic balance plays a role in tamoxifen-induced uterine pathologies, elucidate the molecular impairment in that network, and explore potential intervention to offset the proposed imbalance elicited by tamoxifen. Using in vivo mouse models, we demonstrated that tamoxifen induced a dose-dependent shift in endogenous uterine angiogenic balance favoring VEGF over PEDF. Treatment with recombinant PEDF (rPEDF) abrogated tamoxifen-induced uterine hyperplasia and VEGF elevation, resulting in reduction of blood vessels density. Exploring the molecular mechanism revealed that tamoxifen promoted survival and malignant transformation pathways, whereas rPEDF treatment prevents these changes. Activation of survival pathways was decreased, demonstrated by reduction in AKT phosphorylation concomitant with elevation in JNK phosphorylation. Estrogen receptor-α and c-Myc oncoprotein levels were reduced. Our findings provide novel insight into the molecular mechanisms tamoxifen induces in the uterus, which may become the precursor events of subsequent endometrial hyperplasia and cancer. We demonstrate that rPEDF may serve as a useful intervention to alleviate the risk of tamoxifen-induced endometrial pathologies.

MeSH terms

  • Animals
  • Blood Vessels / drug effects
  • Blood Vessels / pathology
  • Breast Neoplasms / complications
  • Breast Neoplasms / drug therapy*
  • Breast Neoplasms / genetics
  • Breast Neoplasms / pathology
  • Endometrial Hyperplasia / chemically induced
  • Endometrial Hyperplasia / genetics*
  • Endometrial Hyperplasia / therapy
  • Estrogen Receptor alpha / biosynthesis
  • Eye Proteins / administration & dosage
  • Eye Proteins / genetics*
  • Eye Proteins / metabolism
  • Female
  • Gene Expression Regulation, Neoplastic / drug effects
  • Humans
  • Mice
  • Neovascularization, Pathologic / drug therapy*
  • Neovascularization, Pathologic / pathology
  • Nerve Growth Factors / administration & dosage
  • Nerve Growth Factors / genetics*
  • Nerve Growth Factors / metabolism
  • Proto-Oncogene Proteins c-myc / biosynthesis
  • Recombinant Proteins / administration & dosage
  • Recombinant Proteins / metabolism
  • Serpins / administration & dosage
  • Serpins / genetics*
  • Serpins / metabolism
  • Tamoxifen / adverse effects
  • Vascular Endothelial Growth Factor A / genetics
  • Vascular Endothelial Growth Factor A / metabolism*
  • Xenograft Model Antitumor Assays

Substances

  • Estrogen Receptor alpha
  • Eye Proteins
  • MYC protein, human
  • Nerve Growth Factors
  • Proto-Oncogene Proteins c-myc
  • Recombinant Proteins
  • Serpins
  • VEGFA protein, human
  • Vascular Endothelial Growth Factor A
  • pigment epithelium-derived factor
  • Tamoxifen