Published in

American Association for the Advancement of Science, Science Advances, 45(8), 2022

DOI: 10.1126/sciadv.abm3548

Links

Tools

Export citation

Search in Google Scholar

Identification of a minority population of LMO2 <sup>+</sup> breast cancer cells that integrate into the vasculature and initiate metastasis

This paper is made freely available by the publisher.
This paper is made freely available by the publisher.

Full text: Download

Green circle
Preprint: archiving allowed
Red circle
Postprint: archiving forbidden
Green circle
Published version: archiving allowed
Data provided by SHERPA/RoMEO

Abstract

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 immatureTHY1+/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 ofLMO2+basal cells correlated with tumor endothelial content and predicted poor distant recurrence–free survival in patients. UsingMMTV-PyMT/Lmo2CreERT2mice, we demonstrated thatLmo2lineage–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.