Triple-negative breast cancer (TNBC) lacks the expression of estrogen receptor (ER), progesterone receptor (PR) and Her2. TNBC can be classified into at least six subtypes including mesenchymal-like and basal-like with the majority of TNBC categorized as mesenchymal-like. TNBC makes up approximately 15-20% of all breast cancer cases and most commonly affects younger women and African-American women. Patients with TNBC generally have a poor prognosis and short-term survival. This aggressive disease does not respond to widely used targeted therapies such as trastuzumab or endocrine therapies, such as tamoxifen or aromatase inhibitors. Although women with metastatic TNBC can initially respond to conventional chemotherapeutic agents, relapse is inevitable, and oncologists have little to offer other than additional non-cross reactive systemic chemotherapy regimens. Therefore, the challenge is to develop a more effective treatment regimen for women with metastatic TNBC. We find that serine-threonine phosphatase PP2A is a rate-limiting factor (bottleneck) that regulates drug-resistance in TNBC cells. We show that PP2A inhibitors can sensitize both sensitive and resistant mesenchymal-like TNBC cells to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis. TRAIL-based drugs are currently being tested for the treatment of several cancer types in clinical trials. Therefore, these considerations suggest that mesenchymal-like TNBC may be likely to benefit from TRAIL therapy or in combination with PP2A inhibitor therapy. Although mesenchymal-like TNBC cells are highly sensitive to TRAIL-induced apoptosis, basal-like TNBC cells are resistant to TRAIL. We find that the PP2A inhibitor LB100, a small molecule that has recently been approved to enter phase I clinical trials for the treatment of several cancers, increases conventional chemotherapeutic agent-induced anticancer activity in basal-like TNBC cells in vitro and effectively inhibits tumor growth of basal-like TNBC in vivo. This consideration suggests that LB100 alone or in combination with clinically used chemotherapeutic agents could be an efficacious therapy for basal-like TNBC. In addition, our preliminary data show that TRAIL treatment causes the degradation of PP2A/C, a catalytic subunit of PP2A, in TNBC cells. Mechanistically, TRAIL treatment leads to the recruitment of PP2A/C to the death-inducing signaling complex (DISC) where a Cullin-3-based E3 ligase targets PP2A/C for ubiquitination and subsequent degradation. Importantly, in TNBC cells that have developed acquired TRAIL resistance, PP2A/C protein is resistant to TRAIL-induced degradation. In this proposal, we will test the hypotheses that TRAIL alone or in combination with inhibition of PP2A-mediated survival signals or clinically used chemotherapeutic agents sufficiently causes apoptosis in mesenchymal-like TNBC cells and that inhibition of PP2A survival signals alone or in combination with conventional chemotherapeutic agents effectively inhibits the growth of basal-like TNBC. We are proposing to test these hypotheses with the following three specific aims: 1) To evaluate the therapeutic efficacy of TRAIL as a single agent, in combination with LB100, or paclitaxel in mesenchymal-like TNBC cell lines and xenografts; 2) To define the molecular mechanism of PP2A/C ubiquitination in TRAIL-induced apoptosis; and 3) To determine the therapeutic efficacy of LB100 as a single agent or in combination with paclitaxel in basal-like TNBC cell lines and xenografts. This application will determine the efficacy of using TRAIL, a targeted therapy alone, in combination with LB100 (newly approved by the FDA to enter phase I clinical trials for the treatment of several types of cancer), or in combination with clinically usd chemotherapeutic agents to treat mesenchymal-like TNBC and LB100 alone or its combination with clinically used chemotherapeutic agents to treat basal-like TNBC.

Public Health Relevance

The application will evaluate the therapeutic potential of TRAIL alone or PP2A inhibitors alone as single agents, and in combination with each other, or with conventional chemotherapeutic agents to develop targeted therapies for triple-negative breast cancer, a type of breast cancers that does not respond to widely used targeted therapies such as trastuzumab or endocrine therapies, such as tamoxifen or aromatase inhibitors. The proposed research has relevance to public health and is highly translational with potential clinical impact.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
5R01CA174949-02
Application #
8884558
Study Section
Developmental Therapeutics Study Section (DT)
Program Officer
Forry, Suzanne L
Project Start
2014-08-01
Project End
2019-07-31
Budget Start
2015-08-01
Budget End
2016-07-31
Support Year
2
Fiscal Year
2015
Total Cost
Indirect Cost
Name
Wayne State University
Department
Internal Medicine/Medicine
Type
Schools of Medicine
DUNS #
001962224
City
Detroit
State
MI
Country
United States
Zip Code
48202
Obrist, Florine; Michels, Judith; Durand, Sylvere et al. (2018) Metabolic vulnerability of cisplatin-resistant cancers. EMBO J 37:
Yuan, Xun; Gajan, Ambikai; Chu, Qian et al. (2018) Developing TRAIL/TRAIL death receptor-based cancer therapies. Cancer Metastasis Rev :
Wang, J; Kho, D H; Zhou, J-Y et al. (2017) MKP-1 suppresses PARP-1 degradation to mediate cisplatin resistance. Oncogene 36:5939-5947
Yuan, Xun; Kho, Dhonghyo; Xu, Jing et al. (2017) ONC201 activates ER stress to inhibit the growth of triple-negative breast cancer cells. Oncotarget 8:21626-21638
Allen, Joshua E; Kline, C Leah B; Prabhu, Varun V et al. (2016) Discovery and clinical introduction of first-in-class imipridone ONC201. Oncotarget 7:74380-74392
Xu, Jing; Zhou, Jun-Ying; Xu, Zhengfan et al. (2014) The role of Cullin3-mediated ubiquitination of the catalytic subunit of PP2A in TRAIL signaling. Cell Cycle 13:3750-8