Abstract C39: A novel rewired pathway of nucleotide metabolism drives chemoresistance in pancreatic cancer

2019 
Pancreatic ductal adenocarcinoma (PDAC) is characterized by two molecular subtypes, of which the basal-like subtype is associated with the worst survival and is highly resistant to the currently available first-line chemotherapy. Our laboratory has identified that keratin 17 (K17) is a novel negative prognostic biomarker, as accurate as molecular subtyping in predicting patient survival. Patient-derived data analysis suggests that K17 expression correlates with increased resistance to chemotherapeutic agents. The goal of this study is to determine the role of K17 in chemoresistance, and to identify novel therapeutic approaches for around 50% of PDAC patients with tumors that express high levels of K17. In multiple in vivo and in vitro models of PDAC, spanning human and murine PDAC cells, patient-derived organoids, and orthotopic xenograft models, we determined that K17 expression causes more than two-fold increase in resistance to gemcitabine (Gem) and 5-fluorouracil (5-FU), key components of the current standard-of-care chemotherapeutic regimens. To uncover the mechanism associated to K17-induced chemoresistance, we performed unbiased metabolomic studies in isogenic PDAC cell lines and found that K17 reprograms several key metabolic pathways. In particular, K17 increases pyrimidine biosynthesis, a pathway has been linked to chemoresistance. Rescue experiments showed that deoxycytidine (dC) was sufficient to promote Gem (dC analogue) resistance in K17-nonexpressing PDAC cells, suggesting that upregulation of pyrimidine synthesis by K17 underlies resistance to chemotherapeutic agents. Through unbiased RNA-sequencing studies, we identified that gene expression of enzymes involved in pyrimidine biosynthesis was increased specifically in high K17-expressing cells. Previous reports from our group and others suggest that nuclear K17 regulates cell-cycle progression and gene expression. Through domain-prediction analyses, we discovered a novel domain on K17 involved in transcriptional regulation that is required for metabolic reprogramming. Currently, we are testing the role of this domain in metabolic reprograming. In addition, are pursuing two approaches to determine the “druggability” of these findings. First, we are testing if interrupting K17-mediated nucleotide metabolism, by means of small-molecule inhibitors, resensitizes tumor cells to pyrimidine analogues. Second, we are validating the results of a large-scale small-molecule inhibitor screen of FDA-approved, pharma-developed tools to identify compounds that target DNA metabolism and transcription in K17-expressing PDAC cells. In summary, we identified a novel and potentially druggable pathway of chemoresistance that could ultimately result in developing novel therapeutic strategies to enhance patient survival. Citation Format: Chun-Hao Pan, Cindy V. Leiton, Lucia Roa-Pena, Ryan R. Kawalerski, Richard A. Moffitt, Jiang Zhao, Timothy Spicer, Peter Bailey, David K. Chang, Andrew Biankin, Tim Duong, Pankaj K. Singh, Kenneth R. Shroyer, Luisa F. Escobar-Hoyos. A novel rewired pathway of nucleotide metabolism drives chemoresistance in pancreatic cancer [abstract]. In: Proceedings of the AACR Special Conference on Pancreatic Cancer: Advances in Science and Clinical Care; 2019 Sept 6-9; Boston, MA. Philadelphia (PA): AACR; Cancer Res 2019;79(24 Suppl):Abstract nr C39.
    • Correction
    • Source
    • Cite
    • Save
    • Machine Reading By IdeaReader
    0
    References
    0
    Citations
    NaN
    KQI
    []