Pancreatic Cancer Treatments: The Promise of Signal Transduction Pathways

Jill R. Glausier, Ph.D.

Successfully treating pancreatic cancer represents one of the biggest challenges in oncology to date. Pancreatic cancer has the lowest five-year survival rate of all cancers1; equivalent incidence and mortality rates2; and an underwhelming response to all widely available treatments, including surgical resection3,4. The poor prognosis of pancreatic cancer highlights the need to identify additional therapeutic targets in order to produce more efficacious therapies. To this end, signal transduction cascades associated with pancreatic cancer cells are an area of intense research that has much promise5,6.

Signal transduction pathways include both the membrane-bound receptor that receives the signal, and the complex molecular pathway which then transduces the signal into intracellular activity. Alterations to epidermal growth factor receptors (EGFR) and several molecules within two EGFR-initiated signal transduction pathways are heavily implicated in pancreatic cancer5,7,8. EGFR is overexpressed in the majority of pancreatic cancers9,10, and this excessive expression is associated with an even worse prognosis for the individual11,12. Interestingly, EGFR inhibitors have shown only modest clinical benefits7,13,14, and this may be due to the concomitant mutations and alterations to EGFR signal transduction molecules. For example, one key EGFR-initiated pathway includes K-Ras/RAF/MEK/ERK/MAP2K. Single point mutations in the K-Ras gene have been identified in pancreatic cancers, causing constitutive activation and initiation of various cellular processes that contribute to tumorogenesis5,15-17. The PI3K/AKT/mTOR pathway is another EGFR signal transduction cascade implicated in pancreatic cancer5,6,18. This pathway is involved with the enhancement of cell growth and survival, and also appears to be excessively activated in pancreatic cancers19,20. Numerous other signal transduction molecules and pathways are implicated in pancreatic cancer cells and the stroma, including: 1) JAK/STAT, 2) cyclooxygenase-2 (COX-2), 3) sonic hedgehog (SHH), 4) Notch, and 5) WNT. For example, many pancreatic cancers are associated with persistently active STAT321,22, and excessive COX-2 and WNT expression23,24.

Detection of MafA in FFPE mouse pancreatic islet.

Detection of MafA in FFPE mouse pancreatic islet. Antibody: Rabbit anti-MafA (IHC-00352). Secondary: HRP-conjugated goat anti-rabbit IgG (A120-501P). Substrate: DAB.

The abundance of signaling pathways implicated in pancreatic cancer etiology and progression provides a wealth of possible therapeutic targets that may apply to all, or to subpopulations, of patients. This characteristic of pancreatic cancer may position it as an ideal model for the refinement and application of personalized medicine25,26.


References

1. American Cancer Society. 2013. Cancer facts & figures. American Cancer Society Atlanta, GA.

2. World Health Organization. 2012. WHO mortality database. World Health Organization Statistical Information System.

3. Cooperman, AM. 2001. Pancreatic cancer: the bigger picture. Surg Clin North Am. Jun;81(3):557-574.

4. Siegel R, Ma J, Zou Z, Jemal A. 2014. Cancer statistics, 2014. CA Cancer J Clin, Jan-Feb;64(1)9-29.

5. Wormann SM, Algul H. 2013. Risk Factors and Therapeutic Targets in Pancreatic Cancer. Front Oncol. Nov 18;3:282.

6. Iovanna J, Mallmann MC, Goncalves A, Turrini O, Dagorn JC. 2012. Current knowledge on pancreatic cancer. Front Oncol. Jan 31;2:6.

7. Chong CR, Janne PA. 2013. The quest to overcome resistance to EGFR-targeted therapies in cancer. Nat Med. No;19(11):1389-1400.

8. Mahipal A, Kothari N, Gupta S. 2014 Epidermal growth factor receptor inhibitors: coming of age. Cancer Control. Jan;21(1):74-79.

9. Lemoine NR, Hughes CM, Barton CM, Poulsom R, Jeffery RE, Klöppel G, Hall PA, Gullick WJ. The epidermal growth factor receptor in human pancreatic cancer. J Pathol. Jan;166(1):7-12.

10. Troiani T, Martinelli E, Capasso A, Morgillo F, Orditura M, De Vita F, Ciardiello F. 2012. Targeting EGFR in pancreatic cancer treatment. Curr Drug Targets. Jun;13(6):802-810.

11. Luo G, Long J, Qiu L, Liu C, Xu J, Yu X. 2011. Role of epidermal growth factor receptor expression on patient survival in pancreatic cancer: a meta-analysis. Pancreatology. 11(6):595-600.

12. Mahipal A, Mcdonald MJ, Witkiewicz A, Carr BI. 2012. Cell membrane and cytoplasmic epidermal growth factor receptor expression in pancreatic ductal adenocarcinoma. Med Oncol. Mar;29(1):134-139.

13. Moore MJ, Goldstein D, Hamm J, Figer A, Hecht JR, Gallinger S, Au HJ, Murawa P, Walde D, Wolff RA, et al. 2007. Erlotinib plus gemcitabine compared with gemcitabine alone in patients with advanced pancreatic cancer: a phase III trial of the National Cancer Institute of Canada Clinical Trials Group. J Clin Oncol. May 20; 25(15):1960-1966.

14. Welch SA, Moore MJ. 2007. Erlotinib: success of a molecularly targeted agent for the treatment of advanced pancreatic cancer. Future Oncol. 3(3):247-254.

15. Almoguera C, Shibata D, Forrester K, Martin J, Arnheim N, Perucho M. 1988. Most human carcinomas of the exocrine pancreas contain mutant c-K-ras genes. Cell. May 20;53(4):549-554.

16. Neuzillet C, Hammel P, Tijeras-Raballand A, Couvelard A, Raymond E. 2013. Targeting the Ras-ERK pathway in pancreatic adenocarcinoma. Cancer Metastasis Rev. Jun;32(1-2):147-162.

17. Deramaudt T, Rustgi AK. 2005. Mutant KRAS in the initiation of pancreatic cancer. Biochim Biophys Acta. Nov 25;1756(2):97-101.

18. Michl P, Downward J. 2005. Mechanisms of disease: PI3K/AKT signaling in gastrointestinal cancers. Z. Gastroenterol. Oct;43(10):1133-1139.

19. Schlieman MG, Fahy BN, Ramsamooj R, Beckett L, Bold RJ. 2003. Incidence, mechanism and prognostic value of activated AKT in pancreas cancer. Br J Cancer. Dec 1;89(11):2110-2115.

20. Asano T, Yao Y, Zhu J, Li D, Abbruzzese JL, Reddy SA. 2014. The PI 3-kinase/Akt signaling pathway is activated due to aberrant Pten expression and targets transcription factors NF-kappaB and c-Myc in pancreatic cancer cells. Oncogene. Nov 11;23(53):8571-8580.

21. Toyonaga T, Nakano K, Nagano M, Zhao G, Yamaguchi K, Kuroki S, Eguchi T, Chijiiwa K, Tsuneyoshi M, Tanaka M. 2003. Blockade of constitutively activated Janus kinase/signal transducer and activator of transcription-3 pathway inhibits growth of human pancreatic cancer. Cancer Lett. Nov 10;201(1):107-116.

22. Scholz A, Heinze S, Detjen KM, Peters M, Welzel M, Hauff P, Schirner M, Wiedenmann B, Rosewicz S. 2003. Activated signal transducer and activator of transcription 3 (STAT3) supports the malignant phenotype of human pancreatic cancer Gastroenterology. Sept;125(3):891-905.

23. Weekes CD, Winn RA. 2011. The many faces of wnt and pancreatic ductal adenocarcinoma oncogenesis. Cancers (Basel). Sept 21;3(3):3676-3686.

24. Tucker ON, Dannenberg AJ, Yang EK, Zhang F, Teng L, Daly JM, Soslow RA, Masferrer JL, Woerner BM, Koki AT, Fahey TJ 3rd.1999. Cyclooxygenase-2 expression is up-regulated in human pancreatic cancer. Cancer Res. Mar 1;59(5):987-990.

25. Utomo WK, Narayanan V, Biermann K, van Eijck CH, Bruno MJ, Peppelenbosch MP, Braat H.. 2014. mTOR is a promising therapeutical target in a subpopulation of pancreatic adenocarcinoma. Cancer Lett. May 1;346(2):309-317.

26. Braat H, Bruno M, Kuipers EJ, Peppelenbosch MP. 2012. Pancreatic cancer: promise for personalised medicine? Cancer Lett. May 1;318(1):1-8.