Epithelial ovarian cancer (EOC) is the leading cause of death among gynecological malignancies in the United States. Among all EOC subtypes, ovarian clear cell carcinoma (OCCC) carries the worst prognosis when diagnosed at an advanced stage and there is currently no effective therapy for this disease. ARID1A, a subunit of the epigenetic SWI/SNF chromatin remodeler, is among the genes that show the highest mutation rates across many cancer types. In fact, the ARID1A gene is the highest mutated gene in OCCC that occurs in over 50% of the cases. The ultimate goal of the current proposal is to develop novel approaches to treat and eradicate this devastating disease. Since ARID1A is the highest mutated gene in OCCC, we performed an unbiased screen and demonstrated that in ARID1A mutated OCCC the inhibition of EZH2, another epigenetic regulator, is synthetically lethal. The observed synthetic lethality is due to antagonistic roles played by ARID1A and EZH2 in regulating the expression of the same set of target genes. Inhibition of PI3K/AKT signaling contributes to the observed synthetic lethality. Interestingly, ARID1A mutation typically co-exists with PIK3CA mutation in OCCC. In addition to EZH2, our unexplored data suggest that ARID1A-mutated OCCC cells are also selectively sensitive to inhibition of HDAC2. ARID1A mutation is mutually exclusive from TP53 mutation, and expression of p53 target genes is impaired by ARID1A mutation. However, the mechanism by which ARID1A regulates p53 activity has never been explored. Our preliminary data indicate that ARID1A mutation correlates with the association of myosin IIa (MyoIIa) with the SWI/SNF complex in an ARID1A status-dependent manner. MyoIIa is a known tumor suppressor that is necessary for p53 activity. Thus, these studies suggest that ARID1A mutation impairs p53 activity through MyoIIa sequestration. Our central hypothesis is that ARID1A-mutated OCCC can be treated and ultimately eradicated by targeting the four proteins (namely EZH2, HDAC2, PI3K and p53) we have implicated. Accordingly, three specific aims are proposed:
Aim 1 : To investigate the role of HDAC2 in the observed synthetic lethality and gene expression antagonism between ARID1A and EZH2;
Aim 2. To investigate the mechanisms by which ARID1A regulates p53 activity through MyoIIa;
and Aim 3. To develop novel therapeutic strategies for ARID1A mutation. The proposed studies are highly innovative because they challenge current research/clinical paradigms, contribute to new concepts for epigenetic therapeutics, and utilize innovative methods to explore new intervention strategies for ARID1A-mutated OCCC. The research proposed is of high impact because it has the potential to establish the first effective targeted therapeutic strategy for OCCC. Since genetic alterations in components of the SWI/SNF chromatin-remodeling complex occur in more than 20% of all human cancers, the mechanistic insights gained from the current studies will have broad implications for many different types of cancers as well.

Public Health Relevance

The proposed research is relevant to public health because it will not only reveal critical molecular insights into how ARID1A mutation contributes to ovarian clear cell carcinoma, but also provide scientific rationale for developing urgently needed new therapeutic strategies for these devastating diseases. These studies will also serve as a prototype for more epigenetic therapeutic strategies for cancers with genetic alterations in regulators of chromatin structure, a well-recognized feature of many cancer types. Therefore, the proposed research is relevant to the part of the NIH's mission that pertains to developing fundamental knowledge that will reduce the burden of human illness.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
5R01CA202919-05
Application #
9829039
Study Section
Tumor Cell Biology Study Section (TCB)
Program Officer
Okano, Paul
Project Start
2015-12-01
Project End
2020-11-30
Budget Start
2019-12-01
Budget End
2020-11-30
Support Year
5
Fiscal Year
2020
Total Cost
Indirect Cost
Name
Wistar Institute
Department
Type
DUNS #
075524595
City
Philadelphia
State
PA
Country
United States
Zip Code
19104
Trizzino, Marco; Barbieri, Elisa; Petracovici, Ana et al. (2018) The Tumor Suppressor ARID1A Controls Global Transcription via Pausing of RNA Polymerase II. Cell Rep 23:3933-3945
Fukumoto, Takeshi; Zhang, Rugang; Bitler, Benjamin G (2018) Epigenetic inhibitors for the precision treatment of ARID1A-mutant ovarian cancers: what are the next steps? Expert Rev Precis Med Drug Dev 3:233-236
Wu, Shuai; Fatkhutdinov, Nail; Fukumoto, Takeshi et al. (2018) SWI/SNF catalytic subunits' switch drives resistance to EZH2 inhibitors in ARID1A-mutated cells. Nat Commun 9:4116
Fukumoto, Takeshi; Magno, Elizabeth; Zhang, Rugang (2018) SWI/SNF Complexes in Ovarian Cancer: Mechanistic Insights and Therapeutic Implications. Mol Cancer Res 16:1819-1825
Karakashev, Sergey; Zhu, Hengrui; Wu, Shuai et al. (2018) CARM1-expressing ovarian cancer depends on the histone methyltransferase EZH2 activity. Nat Commun 9:631
Fukumoto, Takeshi; Park, Pyoung Hwa; Wu, Shuai et al. (2018) Repurposing Pan-HDAC Inhibitors for ARID1A-Mutated Ovarian Cancer. Cell Rep 22:3393-3400
Wu, Shuai; Fatkhutdinov, Nail; Zhang, Rugang (2017) Harnessing mutual exclusivity between TP53 and ARID1 A mutations. Cell Cycle 16:2313-2314
Kim, Hyoung; George, Erin; Ragland, Ryan et al. (2017) Targeting the ATR/CHK1 Axis with PARP Inhibition Results in Tumor Regression in BRCA-Mutant Ovarian Cancer Models. Clin Cancer Res 23:3097-3108
Svoronos, Nikolaos; Perales-Puchalt, Alfredo; Allegrezza, Michael J et al. (2017) Tumor Cell-Independent Estrogen Signaling Drives Disease Progression through Mobilization of Myeloid-Derived Suppressor Cells. Cancer Discov 7:72-85
Stephen, Tom L; Payne, Kyle K; Chaurio, Ricardo A et al. (2017) SATB1 Expression Governs Epigenetic Repression of PD-1 in Tumor-Reactive T Cells. Immunity 46:51-64

Showing the most recent 10 out of 21 publications