RT Journal Article SR Electronic T1 CDK19 is a Regulator of Triple-Negative Breast Cancer Growth JF bioRxiv FD Cold Spring Harbor Laboratory SP 317776 DO 10.1101/317776 A1 Robert W. Hsieh A1 Angera H. Kuo A1 Ferenc A. Scheeren A1 Mark A. Zarnegar A1 Shaheen S. Sikandar A1 Jane Antony A1 Luuk S. Heitink A1 Divya Periyakoil A1 Tomer Kalisky A1 Sopheak Sim A1 Dalong Qian A1 Sanjay V. Malhotra A1 George Somlo A1 Frederick M. Dirbas A1 Ajit Jadhav A1 Aaron M. Newman A1 Michael F. Clarke YR 2018 UL http://biorxiv.org/content/early/2018/05/10/317776.abstract AB Triple-negative breast cancer (TNBC) is a poor prognosis disease with no clinically approved targeted therapies. Here, using in vitro and in vivo RNA interference (RNAi) screens in TNBC patient-derived xenografts (PDX), we identify cyclin dependent kinase 19 (CDK19) as a potential therapeutic target. Using in vitro and in vivo TNBC PDX models, we validated the inhibitory effect of CDK19 knockdown on tumor initiation, proliferation and metastases. Despite this, CDK19 knockdown did not affect the growth of non-transformed mammary epithelial cells. Using CD10 and EpCAM as novel tumor initiating cell (TIC) markers, we found the EpCAMmed/high/CD10−/low TIC sub-population to be enriched in CDK19 and a putative cellular target of CDK19 inhibition. Comparative gene expression analysis of CDK19 and CDK8 knockdowns revealed that CDK19 regulates a number of cancer-relevant pathways, uniquely through its own action and others in common with CDK8. Furthermore, although it is known that CDK19 can act at enhancers, our CHIP-Seq studies showed that CDK19 can also epigenetically modulate specific H3K27Ac enhancer signals which correlate with gene expression changes. Finally, to assess the potential therapeutic utility of CDK19, we showed that both CDK19 knockdown and chemical inhibition of CDK19 kinase activity impaired the growth of pre-established PDX tumors in vivo. Current strategies inhibiting transcriptional co-factors and targeting TICs have been limited by toxicity to normal cells. Because of CDK19’s limited tissue distribution and the viability of CDK19 knockout mice, CDK19 represents a promising therapeutic target for TNBC.