4.8 Article

Synergistic targeting and resistance to PARP inhibition in DNA damage repair-deficient pancreatic cancer

Journal

GUT
Volume 70, Issue 4, Pages 743-760

Publisher

BMJ PUBLISHING GROUP
DOI: 10.1136/gutjnl-2019-319970

Keywords

-

Funding

  1. German Cancer Aid grant [111879]
  2. Deutsche Forschungsgemeinschaft (DFG) 'Sachbeihilfe' [KL 2544/1-1, KL 2544/1-2, KL 2544/5-1, KL 2544/7-1]
  3. Deutsche Forschungsgemeinschaft (DFG) Heisenberg-Programm [KL 2544/6-1]
  4. Baden-Wurttemberg-Foundation ExPoChip
  5. INDIMED-Verbund PancChip
  6. DFG [GRK 2254/1]
  7. Ulm University
  8. Max Eder Fellowship of the German Cancer Aid [111746]
  9. German Cancer Aid Priority Program 'Translational Oncology' [70112505]
  10. Collaborative Research Centre grant of the German Research Foundation [316249678 - SFB 1279]
  11. German Cancer Aid (PiPAC) [70112505]
  12. Volkswagenstiftung/Ministry for Science and Culture in Lower Saxony [ZN3222]
  13. Deutsche Forschungsgemeinschaft [AZ.96/1-3]
  14. Deutsche Krebshilfe [111264]
  15. German Cancer Aid Priority Program Translational Oncology [70112504]

Ask authors/readers for more resources

The study identified synergistic effects in inhibiting PARP, ATR, and DNA-PKcs that lead to synthetic lethality in ATM-deficient pancreatic ductal adenocarcinoma. Chemical inhibition of ATM sensitized human PDAC cells towards this combinatorial therapy, which was also found to overcome PARP inhibitor resistance. These findings suggest potential mutation-specific therapeutic strategies that may be applied through ATM inhibition regardless of genotype.
Objective ATM serine/threonine kinase (ATM) is the most frequently mutated DNA damage response gene, involved in homologous recombination (HR), in pancreatic ductal adenocarcinoma (PDAC). Design Combinational synergy screening was performed to endeavour a genotype-tailored targeted therapy. Results Synergy was found on inhibition of PARP, ATR and DNA-PKcs (PAD) leading to synthetic lethality in ATM-deficient murine and human PDAC. Mechanistically, PAD-induced PARP trapping, replication fork stalling and mitosis defects leading to P53-mediated apoptosis. Most importantly, chemical inhibition of ATM sensitises human PDAC cells toward PAD with long-term tumour control in vivo. Finally, we anticipated and elucidated PARP inhibitor resistance within the ATM-null background via whole exome sequencing. Arising cells were aneuploid, underwent epithelial-mesenchymal-transition and acquired multidrug resistance (MDR) due to upregulation of drug transporters and a bypass within the DNA repair machinery. These functional observations were mirrored in copy number variations affecting a region on chromosome 5 comprising several of the upregulated MDR genes. Using these findings, we ultimately propose alternative strategies to overcome the resistance. Conclusion Analysis of the molecular susceptibilities triggered by ATM deficiency in PDAC allow elaboration of an efficient mutation-specific combinational therapeutic approach that can be also implemented in a genotype-independent manner by ATM inhibition.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.8
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available