Login / Signup

Identification of a minority population of LMO2 + breast cancer cells that integrate into the vasculature and initiate metastasis.

Shaheen S SikandarGunsagar S GulatiJane AntonyIsobel J FetterAngera H KuoWilliam Hai Dang HoVeronica Haro-AcostaSoumyashree DasChloé Beate SteenThiago Almeida PereiraDalong QianPhilip A BeachyFrederick M DirbasKristy Red-HorseTerence Howard RabbittsJean Paul ThieryAaron M NewmanMichael F Clarke
Published in: Science advances (2022)
Metastasis is responsible for most breast cancer-related deaths; however, identifying the cellular determinants of metastasis has remained challenging. Here, we identified a minority population of immature THY1 + / VEGFA + tumor epithelial cells in human breast tumor biopsies that display angiogenic features and are marked by the expression of the oncogene, LMO2 . Higher abundance of LMO2 + basal cells correlated with tumor endothelial content and predicted poor distant recurrence-free survival in patients. Using MMTV-PyMT/Lmo2 CreERT2 mice, we demonstrated that Lmo2 lineage-traced cells integrate into the vasculature and have a higher propensity to metastasize. LMO2 knockdown in human breast tumors reduced lung metastasis by impairing intravasation, leading to a reduced frequency of circulating tumor cells. Mechanistically, we find that LMO2 binds to STAT3 and is required for STAT3 activation by tumor necrosis factor-α and interleukin-6. Collectively, our study identifies a population of metastasis-initiating cells with angiogenic features and establishes the LMO2-STAT3 signaling axis as a therapeutic target in breast cancer metastasis.
Keyphrases