Inactivation of tumor suppressor genes constitutes a driving mechanism in oncogenesis. In normal cells, tumor suppressors act as central hubs coordinating cell cycle, cell death, and cell fate via regulation of gene expression. The loss of tumor suppressor activities disrupts these processes, permitting aberrant cell growth. Recent work has shown that ~20% of human cancers carry mutations in at least one subunit of the SWI/SNF chromatin-remodeling complex. While genetic, animal and cell culture studies have established that several SWI/SNF subunits function as tumor suppressors, few studies have comprehensively established the mechanisms by which they act. We recently identified inactivating mutations in the SWI/SNF ATPase subunit, SMARCA4, in the majority of small cell carcinoma of the ovary of hypercalcemic type (SCCOHT), a poorly- differentiated and aggressive tumor, with concomitant protein loss in nearly 90% of primary tumors. We also found loss of expression of the alternative SWI/SNF ATPase subunit, SMARCA2, in all tumors lacking SMARCA4. In contrast to most adult cancers, SCCOHT genomes remain predominantly diploid with rare secondary mutations in other cancer genes. Therefore, we hypothesize that 1) SMARCA4 mutations drive tumor development through disruption of normal differentiation; and 2) that these mutations will define a global therapeutic vulnerability in cancer. To test this hypothesis, we propose to 1) Investigate the epigenetic consequences of SMARCA4/A2 re-expression; 2) Model the histogenesis of SCCOHT; and 3) Identify and validate targets for tumor treatment. To carry out the proposed studies, we take advantage of the cutting-edge technologies available at the University of British Columbia (UBC), the Translational Genomics Research Institute (TGen) and the University of North Carolina (UNC) and the complementary expertise of the 3 multiple principal investigators including ovarian cancer pathology and biology (UBC), systems biology and translational applications (TGen) and chromatin biology and cancer epigenetics (UNC). We will also generate novel cell culture and animal models to parse the mechanisms by which inactivation of SMARCA4 and/or SMARCA2 drive SCCOHT development and to identify and validate new reagents for treatment of this deadly disease. In addition, the predominance of SMARCA4 mutations along with the scarcity of secondary mutations in other cancer genes yields a unique model for understanding the specific contributions of SWI/SNF complex mutations to human cancer development. Thus, the results of these studies will significantly impact the diagnosis and treatment of SCCOHT by providing seminal insights into the mechanisms that drive its development, including the disruption of SWI/SNF complex function. Furthermore, the ubiquitous nature of SWI/SNF complex mutations in human malignancies broadens the scope of our findings to other deadly human cancers including lung cancer, renal cell carcinoma and brain tumors.

Public Health Relevance

This project examines how mutations in genes that regulate the structure of DNA and proteins in the nuclei of cells lead to the development of a rare but deadly cancer of the ovaries. The findings from these studies will identify novel targets for treatment of this disease as well as assess the potency of existing drugs. Furthermore, our results will prove applicable to other deadly human cancers, such as lung and brain tumors, that also contain the same mutations.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
5R01CA195670-02
Application #
9070725
Study Section
Cancer Genetics Study Section (CG)
Program Officer
Mietz, Judy
Project Start
2015-06-01
Project End
2020-05-31
Budget Start
2016-06-01
Budget End
2017-05-31
Support Year
2
Fiscal Year
2016
Total Cost
Indirect Cost
Name
University of North Carolina Chapel Hill
Department
Pathology
Type
Schools of Medicine
DUNS #
608195277
City
Chapel Hill
State
NC
Country
United States
Zip Code
27599
Wang, Yemin; Chen, Shary Yuting; Colborne, Shane et al. (2018) Histone Deacetylase Inhibitors Synergize with Catalytic Inhibitors of EZH2 to Exhibit Antitumor Activity in Small Cell Carcinoma of the Ovary, Hypercalcemic Type. Mol Cancer Ther 17:2767-2779
Lang, Jessica D; Hendricks, William P D (2018) Identification of Driver Mutations in Rare Cancers: The Role of SMARCA4 in Small Cell Carcinoma of the Ovary, Hypercalcemic Type (SCCOHT). Methods Mol Biol 1706:367-379
Kuwahara, Yasumichi; Kennedy, Leslie M; Karnezis, Anthony N et al. (2018) High Frequency of Ovarian Cyst Development in Vhl2B/+;Snf5+/- Mice. Am J Pathol 188:1510-1516
Gao, Yanzhe; Kardos, Jordan; Yang, Yang et al. (2018) The Cancer/Testes (CT) Antigen HORMAD1 promotes Homologous Recombinational DNA Repair and Radioresistance in Lung adenocarcinoma cells. Sci Rep 8:15304
Witkowski, Leora; Goudie, Catherine; Ramos, Pilar et al. (2016) The influence of clinical and genetic factors on patient outcome in small cell carcinoma of the ovary, hypercalcemic type. Gynecol Oncol 141:454-460
Lloyd, K C Kent; Khanna, Chand; Hendricks, William et al. (2016) Precision medicine: an opportunity for a paradigm shift in veterinary medicine. J Am Vet Med Assoc 248:45-8
Karnezis, Anthony N; Wang, Yemin; Ramos, Pilar et al. (2016) Dual loss of the SWI/SNF complex ATPases SMARCA4/BRG1 and SMARCA2/BRM is highly sensitive and specific for small cell carcinoma of the ovary, hypercalcaemic type. J Pathol 238:389-400