Zur Kurzanzeige

Stromal and epithelial mechanisms of chemotherapeutic resistance in pancreatic cancer

dc.contributor.advisorNeeße, Albrecht PD Dr. Dr.
dc.contributor.authorPatzak, Melanie Susanne
dc.date.accessioned2019-05-24T10:37:27Z
dc.date.available2020-01-28T23:50:02Z
dc.date.issued2019-05-24
dc.identifier.urihttp://hdl.handle.net/11858/00-1735-0000-002E-E650-E
dc.identifier.urihttp://dx.doi.org/10.53846/goediss-7441
dc.language.isoengde
dc.rights.urihttp://creativecommons.org/licenses/by-nc-nd/4.0/
dc.subject.ddc610
dc.titleStromal and epithelial mechanisms of chemotherapeutic resistance in pancreatic cancerde
dc.typedoctoralThesisde
dc.contributor.refereeNeeße, Albrecht PD Dr. Dr.
dc.date.examination2019-01-29
dc.description.abstractengPancreatic ductal adenocarcinoma (PDAC) is a devastating disease with a 5-year survival rate of less than 8 %. Hallmarks of pancreatic cancer are extensive desmoplasia and strong resistance to standard chemotherapeutic agents, e.g. gemcitabine. In this context, impaired drug delivery and drug metabolism pathways might play a crucial role in mediating this pronounced chemoresistance. In this study, I investigated tumor cell intrinsic and extrinsic mechanisms of chemotherapeutic resistance in PDAC. Pharmacokinetic characteristics of gemcitabine (dFdC) were analyzed in the widely used LSL-Kras<sup>G12D/+</sup>;LSL-Trp53<sup>R172H/+</sup>;Pdx-1-Cre (KPC) mouse model by liquid chromatography tandem mass-spectrometry (LC-MS/MS). Surprisingly, the levels of gemcitabine were elevated in the primary, stroma-rich and hypovascular tumor samples compared with matched normal liver samples and samples from liver metastases. A more detailed analysis by our group revealed an increased ratio of cancer-associated fibroblasts (CAFs) in primary tumors compared to liver metastases. Notably, gemcitabine metabolizing enzymes were highly expressed in epithelial but not stromal cells. The inactivating enzymes cytidine deaminase (CDA), deoxycytidylate deaminase (DCTD), and cytosolic 5’-nucleotidase 1A (NT5C1A) were hardly expressed in the stromal compartment of murine and human PDAC samples in vivo. In contrast, these enzymes were robustly expressed in the epithelial compartment. Consequently, the cytotoxic gemcitabine metabolite dFdCTP accumulated in murine CAFs, as the phosphorylated metabolites are unable to cross the cell membrane. Drug scavenging of CAFs was confirmed in conditioned medium (CM) assays. Incubation of tumor cells with CM of gemcitabine treated CAFs reduced the available amount of gemcitabine for tumor cells in vitro. Due to the low proliferation rate of CAFs in vivo, these cells might have exhibited intrinsic resistance to the increased amounts of dFdCTP. Further experiments were aimed to characterize NT5C1A, a previously unrecognized gemcitabine inactivating enzyme in pancreatic cancer that reverses the initial phosphorylation step of gemcitabine. Immunohistochemical staining of tissue microarrays (TMAs) with more than 400 tumor samples, from two independent cohorts of resected PDAC patients, were used to study the expression pattern of NT5C1A in PDAC. We found robust protein expression in the epithelial compartment of 64-70 % of PDAC patients, whereas robust stromal expression of NT5C1A was detectable in less than 20 % of these patients. A prognostic role of NT5C1A was not observed in both patient cohorts. Recombinant expression of this enzyme was used to elucidate its impact on chemotherapeutic resistance. Re-expression of NT5C1A in pancreatic stellate cells (PSCs) reduced the intracellular levels of the active gemcitabine metabolite dFdCTP, suggesting NT5C1A as novel target for stromal reprogramming. Gemcitabine response in tumor cells overexpressing NT5C1A was investigated using standard biochemical assays and orthotopic transplantation of the modified tumor cells into mice. Indeed, cells overexpressing NT5C1A showed higher resistance towards gemcitabine and had decreased levels of intracellular dFdCTP and of cleaved caspase 3 (CC3) levels following treatment with gemcitabine. Tumor weights were increased in mice that were transplanted with NT5C1A expressing cells compared to control cells upon gemcitabine treatment, showing the relevance of this enzyme in therapeutic effectiveness. Given its role in dephosphorylation of nucleoside monophosphates, NT5C1A overexpression in pancreatic cancer cells did not reduce chemosensitivity towards paclitaxel, a standard chemotherapeutic agent that acts independently of intracellular phosphorylation. In conclusion, our study gave new insight into the impact of drug metabolizing enzymes on chemotherapeutic resistance in PDAC. We demonstrated that alterations in drug metabolism and not impaired drug delivery mainly determine the response to gemcitabine in PDAC. Our results further demonstrated NT5C1A as target for stromal reprogramming. Most importantly, our findings pave the way for a more detailed stratification of patients for treatments and suggest NT5C1A to be considered as a possible predictor of treatment response to gemcitabine in PDAC patients.de
dc.contributor.coRefereeJohnsen, Steven Prof. Dr.
dc.subject.engpancreatic cancerde
dc.subject.engchemotherapeutic resistancede
dc.subject.enggemcitabinede
dc.subject.engNT5C1Ade
dc.identifier.urnurn:nbn:de:gbv:7-11858/00-1735-0000-002E-E650-E-6
dc.affiliation.instituteMedizinische Fakultät
dc.subject.gokfullInnere Medizin - Allgemein- und Gesamtdarstellungen (PPN619875747)de
dc.description.embargoed2020-01-28
dc.identifier.ppn1666651680


Dateien

Thumbnail

Das Dokument erscheint in:

Zur Kurzanzeige