Abstract

Abstract Background: The 5-year survival rate of pancreatic cancer is extremely low compared with other cancers, although it has increased to 12% in 2023 in the United States. 80% of pancreatic ductal adenocarcinoma (PDAC) patients are not eligible for surgical procedure, and unfortunately, PDAC is refractory to standard-of-care chemotherapy. Thus, developing novel and more effective treatment strategies for PDAC is urgently needed. We previously reported keratin 17 (K17) as a novel negative prognostic and predictive biomarker, whose overexpression confers the resistance to chemotherapies. Here, we investigated the mechanisms of chemoresistance and tumor-specific vulnerabilities that can be exploited for K17-mediated targeted therapies. Methods: Survival analyses, immunohistochemistry (IHC), and matrix assisted laser desorption/ionization mass spectrometry (MALDI MSI) metabolite imaging were applied in patient derived samples. The K17 stable expression cell line models (KPC and PANC-1) and K17 loss-of-function cell line model (L3.6) were generated, where the analyses were applied including western blots, qRT-PCR, mass spectrometric metabolomics analysis, immunofluorescence imaging, cell viability and proliferation assays. KPC cells with or without K17 expression were orthotopically implanted into the head of the pancreas of c57B6J mice for tumor growth and treatment study. Results: Unbiased metabolomic studies in isogenic PDAC models identified several key metabolic pathways that are upregulated in the presence of K17. We demonstrate that K17 increases pyrimidine biosynthesis, a pathway that drives chemoresistance. Patient dataset analysis revealed that K17 expression and enzymes involved in pyrimidine, but not purine, de novo biosynthesis is associated with shorter patient survival. Furthermore, K17-expressing cells were more sensitive to Brequinar, a specific inhibitor of dihydroorotate dehydrogenase (DHODH), the rate-limiting enzyme in de novo pyrimidine biosynthetic pathway. Targeting DHODH by small interfering RNA or by Brequinar with Gemcitabine synergistically inhibited the viability of K17- positive cells. Importantly, the combination of Gemcitabine and Brequinar significantly inhibited the growth of K17-expressing tumors and extended survival of mice bearing K17-positive tumors. Mechanistically, K17 increased mitochondrial DHODH translocation which made these cells more sensitive to Brequinar. We also found that the coil 2 domain of K17 was crucial for triggering metabolic reprogramming and promoting tumor aggression. Conclusions: Overall, we define a novel connection between K17, nucleotide metabolism, and chemoresistance in PDAC. We report for the first time that K17, a negative biomarker of the most aggressive PDAC subtype, drives chemoresistance by up-regulating de novo pyrimidine biosynthesis, which provides a tumor-specific vulnerability: DHODH. Further study could lead to the development of a DHODH targeted therapy for K17-expressing PDAC. Citation Format: Yinghuan Lyu, Kenneth Shroyer, Chun-Hao Pan, Robert Tseng, Md Afjalus Siraj, Girish Halemirle Rajacharya, Bo Chen, Katie L Donnelly, Michael Horowitz, Carlos Mauricio Mejia Arbelaez, Sumedha Chowdhury, Lyanne Oblein, Natalia Marchenko, Luisa Escobar-Hoyos, Pankaj K. Singh. K17-induced pyrimidine biosynthesis drives chemoresistance in PDAC [abstract]. In: Proceedings of the AACR Special Conference in Cancer Research: Pancreatic Cancer; 2023 Sep 27-30; Boston, Massachusetts. Philadelphia (PA): AACR; Cancer Res 2024;84(2 Suppl):Abstract nr B085.

Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call