학술논문

LGR5 in breast cancer and ductal carcinoma in situ: a diagnostic and prognostic biomarker and a therapeutic target
Document Type
article
Source
BMC Cancer. 20(1)
Subject
Biomedical and Clinical Sciences
Oncology and Carcinogenesis
Immunology
Breast Cancer
Biotechnology
Prevention
Cancer
Adult
Aged
Aged
80 and over
Animals
Biomarkers
Tumor
Breast Neoplasms
Carcinoma
Intraductal
Noninfiltrating
Cell Line
Tumor
Female
Heterografts
Humans
Mice
Middle Aged
Prognosis
RNA
Neoplasm
Real-Time Polymerase Chain Reaction
Receptor
ErbB-2
Receptors
G-Protein-Coupled
Tissue Array Analysis
LGR5
Breast cancer
DCIS
Estrogen receptor
Targeted therapy
Receptor
erbB-2
Public Health and Health Services
Oncology & Carcinogenesis
Oncology and carcinogenesis
Epidemiology
Language
Abstract
BackgroundNovel biomarkers are required to discern between breast tumors that should be targeted for treatment from those that would never become clinically apparent and/or life threatening for patients. Moreover, therapeutics that specifically target breast cancer (BC) cells with tumor-initiating capacity to prevent recurrence are an unmet need. We investigated the clinical importance of LGR5 in BC and ductal carcinoma in situ (DCIS) to explore LGR5 as a biomarker and a therapeutic target.MethodsWe stained BC (n = 401) and DCIS (n = 119) tissue microarrays with an antibody against LGR5. We examined an LGR5 knockdown ER- cell line that was orthotopically transplanted and used for in vitro colony assays. We also determined the tumor-initiating role of Lgr5 in lineage-tracing experiments. Lastly, we transplanted ER- patient-derived xenografts into mice that were subsequently treated with a LGR5 antibody drug conjugate (anti-LGR5-ADC).ResultsLGR5 expression correlated with small tumor size, lower grade, lymph node negativity, and ER-positivity. ER+ patients with LGR5high tumors rarely had recurrence, while high-grade ER- patients with LGR5high expression recurred and died due to BC more often. Intriguingly, all the DCIS patients who later died of BC had LGR5-positive tumors. Colony assays and xenograft experiments substantiated a role for LGR5 in ER- tumor initiation and subsequent growth, which was further validated by lineage-tracing experiments in ER- /triple-negative BC mouse models. Importantly, by utilizing LGR5high patient-derived xenografts, we showed that anti-LGR5-ADC should be considered as a therapeutic for high-grade ER- BC.ConclusionLGR5 has distinct roles in ER- vs. ER+ BC with potential clinical applicability as a biomarker to identify patients in need of therapy and could serve as a therapeutic target for high-grade ER- BC.