SMARCA4 mutation induces tumor cell-intrinsic defects in enhancer landscape and resistance to immunotherapy

Avatar
Poster
Voices Powered byElevenlabs logo
Connected to paperThis paper is a preprint and has not been certified by peer review

SMARCA4 mutation induces tumor cell-intrinsic defects in enhancer landscape and resistance to immunotherapy

Authors

WANG, Y.; Meraz, I.; Qudratullah, M.; Kotagiri, S.; Han, Y.; Xi, Y.; Wang, J.; Lissanu, Y.

Abstract

Cancer genomic studies have identified frequent alterations in components of the SWI/SNF (SWItch/Sucrose Non- Fermenting) chromatin remodeling complex including SMARCA4 and ARID1A. Importantly, clinical reports indicate that SMARCA4-mutant lung cancers respond poorly to immunotherapy and have dismal prognosis. However, the mechanistic basis of immunotherapy resistance is unknown. Here, we corroborated the clinical findings by using immune-humanized, syngeneic, and genetically engineered mouse models of lung cancer harboring SMARCA4 deficiency. Specifically, we show that SMARCA4 loss caused decreased response to anti-PD1 immunotherapy associated with significantly reduced infiltration of dendritic cells (DCs) and CD4+ T cells into the tumor microenvironment (TME) . Mechanistically, we show that SMARCA4 loss in tumor cells led to profound downregulation of STING, IL1{beta} and other components of the innate immune system as well as inflammatory cytokines that are required for efficient recruitment and activity of immune cells. We establish that this deregulation of gene expression is caused by cancer cell-intrinsic reprogramming of the enhancer landscape with marked loss of chromatin accessibility at enhancers of genes involved in innate immune response such as STING, IL1{beta}, type I IFN and inflammatory cytokines. Interestingly, we observed that transcription factor NF-{kappa}B binding motif was highly enriched in enhancers that lose accessibility upon SMARCA4 deficiency. Finally, we confirmed that SMARCA4 and NF-{kappa}B co-occupy the same genomic loci on enhancers associated with STING and IL1{beta}, indicating a functional interplay between SMARCA4 and NF-{kappa}B. Taken together, our findings provide the mechanistic basis for the poor response of SMARCA4-mutant tumors to anti-PD1 immunotherapy and establish a functional link between SMARCA4 and NF-{kappa}B on innate immune and inflammatory gene expression regulation.

Follow Us on

0 comments

Add comment