Pre-existing commensal dysbiosis is a host-intrinsic regulator of tissue inflammation and tumor cell dissemination in hormone receptor-positive breast cancer

It is unknown why some patients with hormone receptor-positive (HR + ) breast cancer present with more aggressive and invasive disease. Metastatic dissemination occurs early in disease and is facilitated by crosstalk between the tumor and tissue environment, suggesting that undefined host-intrinsic...

Full description

Saved in:
Bibliographic Details
Published inCancer research (Chicago, Ill.) Vol. 79; no. 14; pp. 3662 - 3675
Main Authors Rosean, Claire Buchta, Bostic, Raegan R., Ferey, Joshua C. M., Feng, Tzu-Yu, Azar, Francesca N., Tung, Kenneth S., Dozmorov, Mikhail G., Smirnova, Ekaterina, Bos, Paula D., Rutkowski, Melanie R.
Format Journal Article
LanguageEnglish
Published 07.05.2019
Online AccessGet full text

Cover

Loading…
More Information
Summary:It is unknown why some patients with hormone receptor-positive (HR + ) breast cancer present with more aggressive and invasive disease. Metastatic dissemination occurs early in disease and is facilitated by crosstalk between the tumor and tissue environment, suggesting that undefined host-intrinsic factors enhance early dissemination and the probability of developing metastatic disease. Here, we have identified commensal dysbiosis as a host-intrinsic factor associated with metastatic dissemination. Using a mouse model of HR + mammary cancer, we demonstrate that a pre-established disruption of commensal homeostasis results in enhanced circulating tumor cells and subsequent dissemination to the tumor-draining lymph nodes and lungs. Commensal dysbiosis promoted early inflammation within the mammary gland that was sustained during HR + mammary tumor progression. Furthermore, dysbiosis enhanced fibrosis and collagen deposition both systemically and locally within the tumor microenvironment and induced significant myeloid infiltration into the mammary gland and breast tumor. These effects were recapitulated both by directly targeting gut microbes using non-absorbable antibiotics and by fecal microbiota transplantation of dysbiotic cecal contents, demonstrating the direct impact of gut dysbiosis on mammary tumor dissemination. This study identifies dysbiosis as a pre-existing, host-intrinsic regulator of tissue inflammation, myeloid recruitment, fibrosis, and dissemination of tumor cells in HR + breast cancer.
ISSN:0008-5472
1538-7445
DOI:10.1158/0008-5472.CAN-18-3464