[HTML][HTML] Tumor-expressed adrenomedullin accelerates breast cancer bone metastasis

VA Siclari, KS Mohammad, DR Tompkins… - Breast Cancer …, 2014 - Springer
VA Siclari, KS Mohammad, DR Tompkins, H Davis, CR McKenna, X Peng, LL Wessner…
Breast Cancer Research, 2014Springer
Introduction Adrenomedullin (AM) is secreted by breast cancer cells and increased by
hypoxia. It is a multifunctional peptide that stimulates angiogenesis and proliferation. The
peptide is also a potent paracrine stimulator of osteoblasts and bone formation, suggesting a
role in skeletal metastases—a major site of treatment-refractory tumor growth in patients with
advanced disease. Methods The role of adrenomedullin in bone metastases was tested by
stable overexpression in MDA-MB-231 breast cancer cells, which cause osteolytic bone …
Introduction
Adrenomedullin (AM) is secreted by breast cancer cells and increased by hypoxia. It is a multifunctional peptide that stimulates angiogenesis and proliferation. The peptide is also a potent paracrine stimulator of osteoblasts and bone formation, suggesting a role in skeletal metastases—a major site of treatment-refractory tumor growth in patients with advanced disease.
Methods
The role of adrenomedullin in bone metastases was tested by stable overexpression in MDA-MB-231 breast cancer cells, which cause osteolytic bone metastases in a standard animal model. Cells with fivefold increased expression of AM were characterized in vitro, inoculated into immunodeficient mice and compared for their ability to form bone metastases versus control subclones. Bone destruction was monitored by X-ray, and tumor burden and osteoclast numbers were determined by quantitative histomorphometry. The effects of AM overexpression on tumor growth and angiogenesis in the mammary fat pad were determined. The effects of AM peptide on osteoclast-like multinucleated cell formation were tested in vitro. A small-molecule AM antagonist was tested for its effects on AM-stimulated ex vivo bone cell cultures and co-cultures with tumor cells, where responses of tumor and bone were distinguished by species-specific real-time PCR.
Results
Overexpression of AM mRNA did not alter cell proliferation in vitro, expression of tumor-secreted factors or cell cycle progression. AM-overexpressing cells caused osteolytic bone metastases to develop more rapidly, which was accompanied by decreased survival. In the mammary fat pad, tumors grew more rapidly with unchanged blood vessel formation. Tumor growth in the bone was also more rapid, and osteoclasts were increased. AM peptide potently stimulated bone cultures ex vivo; responses that were blocked by small-molecule adrenomedullin antagonists in the absence of cellular toxicity. Antagonist treatment dramatically suppressed tumor growth in bone and decreased markers of osteoclast activity.
Conclusions
The results identify AM as a target for therapeutic intervention against bone metastases. Adrenomedullin potentiates osteolytic responses in bone to metastatic breast cancer cells. Small-molecule antagonists can effectively block bone-mediated responses to tumor-secreted adrenomedullin, and such agents warrant development for testing in vivo.
Springer