Abstract

Abstract Background Tumor phenotype across various cancers, including breast cancer, is predominantly shaped by a synergy between genomic alterations and the cell-of-origin from which the tumor emerges. These elements collectively influence crucial aspects like tumor aggressiveness, treatment response, and patient prognosis. While existing research highlights that the cell-of-origin often leaves a lasting imprint on the molecular architecture of tumors, leveraging this information for actionable clinical insights has proven challenging. Specifically, in the realm of breast cancer, utilizing cellular ancestry signatures could be a key factor in tailoring effective therapeutic strategies and improving patient outcomes. Methods In this study, we developed a unique signature anchored in the expression levels of triple hormone receptors (THR)—namely androgen (AR), estrogen (ER), and vitamin D (VDR)—in normal breast cells. Building on this, we formulated two distinct mRNA markers, THR-50 and THR-70, aimed at categorizing breast tumors based on their THR expression profiles. These markers were rigorously validated across 65 independent breast cancer studies, involving a total of 6,679 patients, utilizing Kaplan-Meier survival curves, Cox proportional hazards models, and unsupervised clustering analyses. Results Our findings indicate that both THR-50 and THR-70 are robustly associated with overall survival and recurrence-free survival in breast cancer patients across all datasets evaluated. Importantly, these THR signatures demonstrate broad applicability across various breast cancer subtypes, grades, and treatment phases—unlike conventional prognostic markers that tend to be subtype-specific. Additionally, the THR signatures have revealed four unique patient clusters with divergent survival outcomes, three of which are ER-positive and one ER-negative. When augmented with an immune-based signature (i20), THR-70 identifies a specific ER-negative breast cancer subgroup with a highly favorable prognosis, comparable to ER-positive subtypes, as well as another ER-negative subgroup characterized by exceedingly poor survival outcomes. Conclusion The THR-based cellular ancestry signatures open a new avenue in understanding the intricacies of breast cancer biology. These signatures offer a robust and stable framework for developing other prognostic markers, thus providing an enhanced stratification of existing breast cancer categories as well as creating new classifications. Citation Format: Mohamed Omar, Chuck Harrell, Rulla Tamimi, Luigi Marchionni, Tan Ince. Triple Hormone Receptor Signatures as Novel Prognostic Markers in Breast Cancer [abstract]. In: Proceedings of the 2023 San Antonio Breast Cancer Symposium; 2023 Dec 5-9; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2024;84(9 Suppl):Abstract nr PO3-28-08.

Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call