Breast cancer (BC) has several distinct molecular subtypes, including estrogen receptor (ESR1) positive and triple negative BC (TNBC). A significant proportion of ESR1-positive therapy sensitive-BCs (TS-BC) initially respond to antiestrogens or aromatase inhibitors, but become therapy resistant-BCs (TR-BC) and progress to incurable metastases. Further, TNBC subtype has a more aggressive clinical course and lack targeted therapies. Development of effective therapies for women with TR-BC and TNBCs represents the highest unmet need. Recent studies revealed the potential role of several members of the Nuclear Receptor (NR) superfamily as molecular drivers in TR-BC and TNBC, including the androgen receptor (AR), glucocorticoid receptor (GR) and the orphan NR tailless (TLX, NR2E1). The specificity and magnitude of NR signaling is mediated by the interaction between NR and critical coregulators and depending on the molecular context in which NRs and NR coregulators are altered, they may contribute to BC progression. The variability of the contribution of specific NRs and NR coregulators to disease progression in TR-BCs and TNBCs poses a therapeutic challenge but also an opportunity for agents that can target multiple NRs and NR coregulators. We have developed a first-in-class polyfunctional small molecules, ERXs that have activity in the TR-BCs and TNBCs. ERXs block NR and coregulator interactions. Uniquely, our preliminary studies identified three lead compounds with differential activity to distinct BC molecular subtypes: ERX-11 (activity against TS-BC, TR-BCs), ERX-41 (activity against TS-BCs, TR-BCs and TNBCs) and ERX-1113 (activity against TNBC only). The objective of this proposal is to further develop lead ERXs to treat TR-BC and TNBC. Our overarching hypothesis is that targeting the NR- coregulator interactome will have therapeutic utility in treating TR-BC and TNBC. Our initial preliminary studies indicated that ERX-11 targets ESR1, while the molecular targets of ERX-41 and ERX-1113 are not yet defined.
In Aim1, we will determine the mechanism of action of ERX derivatives using a novel forward genetics approach to identify the molecular target(s) of ERX compounds in ESR1+ BC and TNBCs and establish the molecular interactome using unbiased mass spectroscopy-based approaches and whole genome sequencing approaches.
In Aim2 we will optimize the translatability of ERX derivatives and conduct detailed PK, PD, tolerability and toxicity studies on lead ERX compounds.
In Aim3, we will test of the efficacy of ERX compounds in using patient derived explant tissues, endocrine resistant models, and by using syngeneic, orthotopic xenografts and in patient derived xenografts (PDX). This proposal is innovative as ERXs block multiple critical protein-protein interactions, and uniquely have activity against a large number of TR-BC and TNBC cell lines. Successful completion of the proposed studies will lead to the development of first-in-class cancer therapy drugs that addresses the critical need of targeting TR-BC and TNBC.

Public Health Relevance

Breast cancer (BC) is the most common malignancy in women, the second leading cause of cancer-related death after lung cancer. Most BC patients develop resistance to current endocrine therapy drugs over a period of time, and disease progression is common. Further, a significant number of BCs are categorized as triple negative BC (TNBC) and they lack targeted therapies. In this study, we have rationally designed and synthesized first-in-class poly-functional small molecules (ERXs) that target nuclear receptor interactome. Our preliminary studies showed that ERXs have unique activities against different molecular BC subtypes in vitro and in vivo. Successful completion of aims will establish ERXs as novel drugs for treating advanced BC and ERXs represent a quantum leap in the development of drugs with potent activity against therapy resistant BC and TNBCs.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
5R01CA223828-02
Application #
9613810
Study Section
Special Emphasis Panel (ZRG1)
Program Officer
Alley, Michael C
Project Start
2017-12-14
Project End
2022-11-30
Budget Start
2018-12-01
Budget End
2019-11-30
Support Year
2
Fiscal Year
2019
Total Cost
Indirect Cost
Name
University of Texas Health Science Center
Department
Obstetrics & Gynecology
Type
Schools of Medicine
DUNS #
800772162
City
San Antonio
State
TX
Country
United States
Zip Code
78229