-Author name in bold denotes the presenting author
-Asterisk * with author name denotes a Non-ASH member
Clinically Relevant Abstract denotes an abstract that is clinically relevant.

PhD Trainee denotes that this is a recommended PHD Trainee Session.

Ticketed Session denotes that this is a ticketed session.

940 NSD2 Regulates Pro-Metastatic Gene PTP4A3 through Chromatin Remodeling in Multiple Myeloma

Program: Oral and Poster Abstracts
Session: 602. Disordered Gene Expression in Hematologic Malignancy, including Disordered Epigenetic Regulation: Poster I
Hematology Disease Topics & Pathways:
autoimmune disorders, Biological, multiple myeloma, Diseases, Biological Processes, epigenetics, Plasma Cell Disorders, Lymphoid Malignancies
Saturday, December 5, 2020, 7:00 AM-3:30 PM

Phyllis SY Chong, PhD1*, Jing Yuan Chooi, BSc2*, Julia Lim, Bsc3* and Wee-Joo Chng, MBBS, PhD4

1National University of Singapore, Singapore, Singapore
2Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore, Singapore
3Cancer Science Institute of Singapore, Singapore, Singapore
4National University Cancer Institute of Singapore, Singapore, Singapore

Nuclear receptor-binding SET domain 2 (NSD2) is the primary oncogenic driver and represent a good druggable target due to its universal overexpression in t(4;14) MM. However, given the challenges in the development of small molecule inhibitors against NSD2, we took on a discovery approach to characterize NSD2 interactome as an alternative strategy to target NSD2. We employed SILAC-based mass-spectrometry coupled with gene ontology analysis to uncover the spectrum of NSD2-interacting proteins which are relevant in t(4;14) MM. Validation using in silico protein-protein interaction analysis and immunoprecipitation confirmed that SMARCA2, a bromodomain-containing chromatin remodeler, interacts with NSD2 independent of the SWI/SNF complex. SMARCA2 is primarily expressed in t(4;14) cell lines, and functional assays indicated that dual inhibition of SMARCA2 and NSD2 impairs growth of t(4;14)+ cells. Next, we performed RNA sequencing which led to the identification of PTP4A3 as a downstream target of NSD2 that is co-regulated by SMARCA2. Mechanistically, SMARCA2 is required for the recruitment of NSD2 to PTP4A3 promoter, leading to a transcriptionally permissive state for expression. Importantly, upregulation of PTP4A3 is sufficient for the activation of MYC, which is crucial for t(4;14) myelomagenesis. PFI-3, which belongs to the class of bromodomain and extra-terminal motif protein inhibitors (BETi), displaced SMARCA2 and NSD2 occupancy from PTP4A3 promoter, and selectively inhibited t(4;14) myeloma cell viability. Interestingly, PFI-3 treatment did not perturb the levels of NSD2 nor SMARCA2, rather, the targeting effect is achieved through their occupancy on oncogenes. Finally, high expression of NSD2 and SMARCA2 were predictive of poor survival outcome in a large cohort of myeloma patients. Collectively, we demonstrated a proof-of-concept of how histone modifying enzyme and chromatin remodeling complex cooperatively caused an altered epigenetic state in myelomagenesis through the regulation of an important myeloma gene. Our study proposed that the pharmacological inhibition of SMARCA2 may represent a novel strategy to target t(4;14) myeloma cells using BETi and also explore as combinational therapy with anti-myeloma agents that are currently in the clinics.

Disclosures: No relevant conflicts of interest to declare.

Previous Abstract | Next Abstract >>
*signifies non-member of ASH