Anticancer mechanisms of doxorubicin and zoledronic acid in breast cancer tumor growth in bone

Mol Cancer Ther. 2009 Oct;8(10):2821-32. doi: 10.1158/1535-7163.MCT-09-0462. Epub 2009 Sep 29.

Abstract

Patients with advanced breast cancer frequently develop bone metastases, and at this stage, the disease is considered incurable. Here, we show that a 6-week course of weekly administration of doxorubicin (2 mg/kg), followed 24 hours later by the bisphosphonate zoledronic acid (100microg/kg), causes substantial inhibition of MDA-MB-436 breast tumor burden in bone of immunocompromised mice, compared with administration of the single agents. Molecular analysis of tumors from animals treated sequentially with doxorubicin followed by zoledronic acid showed reduced numbers of proliferating tumor cells and decreased expression of cyclins E1, B, D1, and D3 as well as cdk2 and cdk4. Tumors from the sequential treatment group also displayed increased levels of apoptosis, increased expression of bcl2-associated X protein, decreased expression of B-cell chronic lymphocytic leukemia/lymphoma 2, and activation of caspase 3, 8, and 9. Zoledronic acid caused a small reduction in tumor volume, reduced tumor cell proliferation, and decreased expression of cyclins D1 and D3, compared with tumors from animals treated with saline or doxorubicin. Doxorubicin had no effect on tumor growth, cell cycle, or apoptosis in vivo, but did cause increased accumulation of a bisphosphonate in MDA-MB-436 cells in vitro, suggesting that doxorubicin may affect subsequent uptake of zoledronic acid. In support of this, accumulation of unprenylated Rap1A, a surrogate marker of zoledronic acid, was only detected in tumors following sequential treatment, and not following treatment with zoledronic acid alone. Our data are the first to show the specific molecular pathways by which sequential treatment with doxorubicin and zoledronic acid induce tumor cell apoptosis and inhibit proliferation in an in vivo model of breast tumor growth in bone.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Adult
  • Animals
  • Antineoplastic Agents / pharmacology
  • Antineoplastic Agents / therapeutic use*
  • Apoptosis / drug effects
  • Bone Neoplasms / drug therapy*
  • Bone Neoplasms / secondary*
  • Bone and Bones / drug effects
  • Bone and Bones / pathology
  • Breast Neoplasms / drug therapy
  • Breast Neoplasms / pathology*
  • Cell Cycle / drug effects
  • Cell Cycle / genetics
  • Cell Line, Tumor
  • Cell Proliferation / drug effects
  • Diphosphonates / administration & dosage
  • Diphosphonates / pharmacology
  • Diphosphonates / therapeutic use*
  • Doxorubicin / administration & dosage
  • Doxorubicin / pharmacology
  • Doxorubicin / therapeutic use*
  • Female
  • Gene Expression Regulation, Neoplastic / drug effects
  • Humans
  • Imidazoles / administration & dosage
  • Imidazoles / pharmacology
  • Imidazoles / therapeutic use*
  • Mice
  • Models, Biological
  • Osteoclasts / drug effects
  • Osteoclasts / pathology
  • Xenograft Model Antitumor Assays
  • Zoledronic Acid

Substances

  • Antineoplastic Agents
  • Diphosphonates
  • Imidazoles
  • Zoledronic Acid
  • Doxorubicin