Blocking CXCR4 alleviates desmoplasia, increases T-lymphocyte infiltration, and improves immunotherapy in metastatic breast cancer

Metastatic breast cancers (mBCs) are largely resistant to immune checkpoint blockade, but the mechanisms remain unclear. Primary breast cancers are characterized by a dense fibrotic stroma, which is considered immunosuppressive in multiple malignancies, but the stromal composition of breast cancer m...

Full description

Bibliographic Details
Main Authors: Chen, Ivy Xiaoyu (Author), Chauhan, Vikash P. (Author), Posada, Jessica (Author), Ng, Mei R. (Author), Wu, Michelle W. (Author), Adstamongkonkul, Pichet (Author), Huang, Peigen (Author), Lindeman, Neal (Author), Langer, Robert S (Author), Jain, Rakesh K. (Author)
Other Authors: Massachusetts Institute of Technology. Department of Chemical Engineering (Contributor), Massachusetts Institute of Technology. Department of Biological Engineering (Contributor), Koch Institute for Integrative Cancer Research at MIT (Contributor)
Format: Article
Language:English
Published: National Academy of Sciences, 2020-05-06T14:06:26Z.
Subjects:
Online Access:Get fulltext
Description
Summary:Metastatic breast cancers (mBCs) are largely resistant to immune checkpoint blockade, but the mechanisms remain unclear. Primary breast cancers are characterized by a dense fibrotic stroma, which is considered immunosuppressive in multiple malignancies, but the stromal composition of breast cancer metastases and its role in immunosuppression are largely unknown. Here we show that liver and lung metastases of human breast cancers tend to be highly fibrotic, and unlike primary breast tumors, they exclude cytotoxic T lymphocytes (CTLs). Unbiased analysis of the The Cancer Genome Atlas database of human breast tumors revealed a set of genes that are associated with stromal T-lymphocyte exclusion. Among these, we focused on CXCL12 as a relevant target based on its known roles in immunosuppression in other cancer types. We found that the CXCL12 receptor CXCR4 is highly expressed in both human primary tumors and metastases. To gain insight into the role of the CXCL12/CXCR4 axis, we inhibited CXCR4 signaling pharmacologically and found that plerixafor decreases fibrosis, alleviates solid stress, decompresses blood vessels, increases CTL infiltration, and decreases immunosuppression in murine mBC models. By deleting CXCR4 in αSMA+ cells, we confirmed that these immunosuppressive effects are dependent on CXCR4 signaling in αSMA+ cells, which include cancer-associated fibroblasts as well as other cells such as pericytes. Accordingly, CXCR4 inhibition more than doubles the response to immune checkpoint blockers in mice bearing mBCs. These findings demonstrate that CXCL12/CXCR4- mediated desmoplasia in mBC promotes immunosuppression and is a potential target for overcoming therapeutic resistance to immune checkpoint blockade in mBC patients. Keywords: tumor microenvironment; mnetastatic breast cancer; immune checkpoint blockade; tumor desmoplasia; carcinoma-associated fibroblasts
United States. National Cancer Institute (Grant P01-CA080124)
United States. National Cancer Institute (Grant R01-CA098706)
United States. National Cancer Institute (Grant R01-CA208205)
United States. National Cancer Institute (Grant U01- CA224348)
National Institutes of Health (U.S.) (Ruth L. Kirschstein National Research Service Award Postdoctoral Fellowship F32- CA073479)
S. Leslie Misrock Frontier Research Fund for Cancer Nanotechnology (Misrock Fellowship)