PRC2-mediated repression of SMARCA2 predicts EZH2 inhibitor activity in SWI/SNF mutant tumors

Significance Targeting epigenetic dependencies caused by mutations in chromatin-modifying enzymes represents a novel therapeutic approach in cancer. Notably, cancers harboring mutations in the SNF5 subunit of the SWI/SNF chromatin remodeling complex have been shown to be susceptible to small-molecule inhibitors of the EZH2 histone methyltransferase that are currently in clinical development. We demonstrate that EZH2 inhibition can be effective in SMARCA4 mutant cancers that concurrently transcriptionally silence the paralog helicase SMARCA2. SMARCA2 is directly suppressed by EZH2, and SMARCA2 expression levels predict EZH2 inhibitor activity in other SWI/SNF mutant contexts, including ARID1A mutant tumors. These data provide insight into the utility of EZH2 inhibitors in SWI/SNF mutant tumors and have important implications regarding predictive diagnostics. Subunits of the SWI/SNF chromatin remodeling complex are frequently mutated in human cancers leading to epigenetic dependencies that are therapeutically targetable. The dependency on the polycomb repressive complex (PRC2) and EZH2 represents one such vulnerability in tumors with mutations in the SWI/SNF complex subunit, SNF5; however, whether this vulnerability extends to other SWI/SNF subunit mutations is not well understood. Here we show that a subset of cancers harboring mutations in the SWI/SNF ATPase, SMARCA4, is sensitive to EZH2 inhibition. EZH2 inhibition results in a heterogenous phenotypic response characterized by senescence and/or apoptosis in different models, and also leads to tumor growth inhibition in vivo. Lower expression of the SMARCA2 paralog was associated with cellular sensitivity to EZH2 inhibition in SMARCA4 mutant cancer models, independent of tissue derivation. SMARCA2 is suppressed by PRC2 in sensitive models, and induced SMARCA2 expression can compensate for SMARCA4 and antagonize PRC2 targets. The induction of SMARCA2 in response to EZH2 inhibition is required for apoptosis, but not for growth arrest, through a mechanism involving the derepression of the lysomal protease cathepsin B. Expression of SMARCA2 also delineates EZH2 inhibitor sensitivity for other SWI/SNF complex subunit mutant tumors, including SNF5 and ARID1A mutant cancers. Our data support monitoring SMARCA2 expression as a predictive biomarker for EZH2-targeted therapies in the context of SWI/SNF mutant cancers.

[1]  A. Warth,et al.  SMARCA4 and SMARCA2 deficiency in non-small cell lung cancer: immunohistochemical survey of 316 consecutive specimens. , 2017, Annals of diagnostic pathology.

[2]  P. Park,et al.  ARID1A loss impairs enhancer-mediated gene regulation and drives colon cancer in mice , 2016, Nature Genetics.

[3]  K. Helin,et al.  Maintaining cell identity: PRC2-mediated regulation of transcription and cancer , 2016, Nature Reviews Cancer.

[4]  G. Bunt,et al.  Cathepsin B launches an apoptotic exit effort upon cell death-associated disruption of lysosomes , 2016, Cell Death Discovery.

[5]  R. Copeland,et al.  PRC2 and SWI/SNF Chromatin Remodeling Complexes in Health and Disease. , 2016, Biochemistry.

[6]  J. Trent,et al.  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 , 2015, The Journal of pathology.

[7]  Thomas P. Howard,et al.  SWI/SNF-mutant cancers depend on catalytic and non-catalytic activity of EZH2 , 2015, Nature Medicine.

[8]  N. Girard,et al.  SMARCA4 inactivation defines a group of undifferentiated thoracic malignancies transcriptionally related to BAF-deficient sarcomas , 2015, Nature Genetics.

[9]  Biao Liu,et al.  Frequent co‐inactivation of the SWI/SNF subunits SMARCB1, SMARCA2 and PBRM1 in malignant rhabdoid tumours , 2015, Histopathology.

[10]  G. Crabtree,et al.  Mammalian SWI/SNF chromatin remodeling complexes and cancer: Mechanistic insights gained from human genomics , 2015, Science Advances.

[11]  Benjamin G. Bitler,et al.  Synthetic lethality by targeting EZH2 methyltransferase activity in ARID1A-mutated cancers , 2015, Nature Medicine.

[12]  O. Delattre,et al.  SWI/SNF chromatin remodeling and human malignancies. , 2015, Annual review of pathology.

[13]  P. Trojer,et al.  EZH2 inhibitor efficacy in non-Hodgkin's lymphoma does not require suppression of H3K27 monomethylation. , 2014, Chemistry & biology.

[14]  J. Biegel,et al.  SWI/SNF chromatin remodeling complexes and cancer , 2014, American journal of medical genetics. Part C, Seminars in medical genetics.

[15]  W. Foulkes,et al.  No small surprise – small cell carcinoma of the ovary, hypercalcaemic type, is a malignant rhabdoid tumour , 2014, The Journal of pathology.

[16]  D. Reisman,et al.  The silencing of the SWI/SNF subunit and anticancer gene BRM in Rhabdoid tumors , 2014, Oncotarget.

[17]  N. Schultz,et al.  Recurrent SMARCA4 mutations in small cell carcinoma of the ovary , 2014, Nature Genetics.

[18]  R. Siebert,et al.  Germline and somatic SMARCA4 mutations characterize small cell carcinoma of the ovary, hypercalcemic type , 2014, Nature Genetics.

[19]  C. Roberts,et al.  Functional epigenetics approach identifies BRM/SMARCA2 as a critical synthetic lethal target in BRG1-deficient cancers , 2014, Proceedings of the National Academy of Sciences.

[20]  W. Hahn,et al.  Residual Complexes Containing SMARCA2 (BRM) Underlie the Oncogenic Drive of SMARCA4 (BRG1) Mutation , 2014, Molecular and Cellular Biology.

[21]  R. Copeland,et al.  Durable tumor regression in genetically altered malignant rhabdoid tumors by inhibition of methyltransferase EZH2 , 2013, Proceedings of the National Academy of Sciences.

[22]  Yan Liu,et al.  EZH2 inhibition as a therapeutic strategy for lymphoma with EZH2-activating mutations , 2012, Nature.

[23]  D. Reisman,et al.  Pharmacologic reversal of epigenetic silencing of the anticancer protein BRM: a novel targeted treatment strategy , 2011, Oncogene.

[24]  S. Orkin,et al.  Epigenetic antagonism between polycomb and SWI/SNF complexes during oncogenic transformation. , 2010, Cancer cell.

[25]  C. Roberts,et al.  Oncogenesis caused by loss of the SNF5 tumor suppressor is dependent on activity of BRG1, the ATPase of the SWI/SNF chromatin remodeling complex. , 2009, Cancer research.

[26]  E. Moran,et al.  Antagonistic Roles for BRM and BRG1 SWI/SNF Complexes in Differentiation , 2009, Journal of Biological Chemistry.

[27]  D. Metzger,et al.  Targeted knockout of BRG1 potentiates lung cancer development. , 2008, Cancer research.

[28]  M. Bollen,et al.  The transcriptional repressor NIPP1 is an essential player in EZH2-mediated gene silencing , 2008, Oncogene.

[29]  D. Reisman,et al.  The reversible epigenetic silencing of BRM: implications for clinical targeted therapy , 2007, Oncogene.

[30]  Weidong Wang,et al.  Loss of BRG1/BRM in human lung cancer cell lines and primary lung cancers: correlation with poor prognosis. , 2003, Cancer research.

[31]  B. Emerson,et al.  Transcriptional specificity of human SWI/SNF BRG1 and BRM chromatin remodeling complexes. , 2003, Molecular cell.

[32]  Marcel Leist,et al.  Cathepsin B Acts as a Dominant Execution Protease in Tumor Cell Apoptosis Induced by Tumor Necrosis Factor , 2001, The Journal of cell biology.

[33]  A. Sands,et al.  Disruption of Ini1 Leads to Peri-Implantation Lethality and Tumorigenesis in Mice , 2001, Molecular and Cellular Biology.

[34]  C. Roberts,et al.  Haploinsufficiency of Snf5 (integrase interactor 1) predisposes to malignant rhabdoid tumors in mice. , 2000, Proceedings of the National Academy of Sciences of the United States of America.

[35]  Olivier Delattre,et al.  Truncating mutations of hSNF5/INI1 in aggressive paediatric cancer , 1998, Nature.

[36]  Guha,et al.  Functional interactions between the hBRM/hBRG1 transcriptional activators and the pRB family of proteins , 1996, Molecular and cellular biology.

[37]  C. Roberts,et al.  ARID1A mutations in cancer: another epigenetic tumor suppressor? , 2013, Cancer discovery.