PT - JOURNAL ARTICLE AU - Prasath Pararajalingam AU - Krysta M. Coyle AU - Sarah E. Arthur AU - Nicole Thomas AU - Miguel Alcaide AU - Barbara Meissner AU - Merrill Boyle AU - Bruno M. Grande AU - Graham Slack AU - Andrew J. Mungall AU - Randy D. Gascoyne AU - Christian Steidl AU - Joseph Connors AU - Diego Villa AU - Marco A. Marra AU - Nathalie Johnson AU - David W. Scott AU - Ryan D. Morin TI - Coding and non-coding drivers of mantle cell lymphoma identified through exome and genome sequencing AID - 10.1101/686956 DP - 2019 Jan 01 TA - bioRxiv PG - 686956 4099 - http://biorxiv.org/content/early/2019/07/08/686956.short 4100 - http://biorxiv.org/content/early/2019/07/08/686956.full AB - Mantle cell lymphoma (MCL) is an uncommon B-cell non-Hodgkin lymphoma (NHL) that is incurable with standard therapies. The genetic drivers of this cancer have not been firmly established and the features known to contribute to differences in clinical course remain limited. To extend our understanding of the biological pathways involved in this malignancy, we performed a large-scale genomic analysis of MCL using data from 51 exomes alongside previously published exome cohorts. To confirm our findings, we re-sequenced the genes identified in the exome cohort in 212 MCL tumors, each having clinical follow-up data. We confirmed the prognostic association of TP53 and NOTCH1 mutations and further nominate two additional genes, EWSR1 and MEF2B, whose mutation respectively associated with poor and good outcome. Our sequencing revealed novel recurrent mutations including a unique missense hot spot in MEF2B and a pattern of non-coding mutations surrounding a single exon of the HNRNPH1 gene. We sequenced the whole genomes of 34 MCLs to confirm the focal nature of HNRNPH1 mutations. Using RNA-seq data from 110 of these cases, we identified a functional role for recurrent non-coding HNRNPH1 mutations in disrupting an auto-regulatory feedback mechanism. Overall, we identified three novel MCL-related genes with roles in RNA trafficking or splicing, namely DAZAP1, EWSR1, and HNRNPH1. Taken together, these data strongly implicate a role for aberrant regulation of splicing in MCL pathobiology.Key pointsRNA-binding proteins with roles in regulating alternative splicing, DAZAP1, EWSR1, HNRNPH1, are frequently mutated in MCLThe majority of recurrent somatic HNRNPH1 mutations are intronic and HNRNPH1 exhibits self-regulation through alternative splicing