Skip to main content

Emerging Therapeutic Targets in Pancreatic Adenocarcinoma

  • Living reference work entry
  • First Online:
Pancreatic Cancer

Abstract

Pancreatic adenocarcinoma is one of the most lethal cancers but has limited therapeutic options necessitating continued investigation of new therapeutic agents. Recently, improved overall survival has been achieved with cytotoxic drug combinations including 5-fluorouracil, leucovorin, irinotecan, and oxaliplatin (FOLFIRINOX) and gemcitabine/nab-paclitaxel, but the success has been modest at best. More targeted approaches focusing on EGFR and MAPK signaling have also enjoyed marginal success. Accumulating evidence suggests that pancreatic tumors have increased dependence on metabolic pathways through both KRAS and KRAS-independent mechanisms and are broadly resistant to drug therapy due to stromal remodeling. Genetic and epigenetic vulnerabilities, such as inactivating aberrations in DNA damage repair, chromatin remodeling, and microRNA dysregulation, may reveal exploitable weaknesses. Modern approaches to drug development tailored to molecularly defined subsets of patients likely to respond to targeted therapies are needed to achieve more substantial progress in this disease in an era of precision medicine.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Institutional subscriptions

References

  1. Ying H, Kimmelman AC, Lyssiotis CA, Hua S, Chu GC, Fletcher-Sananikone E, et al. Oncogenic Kras maintains pancreatic tumors through regulation of anabolic glucose metabolism. Cell. 2012;149(3):656–70. doi:10.1016/j.cell.2012.01.058.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Samatar AA, Poulikakos PI. Targeting RAS-ERK signalling in cancer: promises and challenges. Nat Rev Drug Discov. 2014;13(12):928–42. doi:10.1038/nrd4281.

    Article  CAS  PubMed  Google Scholar 

  3. Lito P, Saborowski A, Yue J, Solomon M, Joseph E, Gadal S, et al. Disruption of CRAF-mediated MEK activation is required for effective MEK inhibition in KRAS mutant tumors. Cancer Cell. 2014;25(5):697–710. doi:10.1016/j.ccr.2014.03.011.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Vander Heiden MG, Cantley LC, Thompson CB. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science. 2009;324(5930):1029–33. doi:10.1126/science.1160809.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Yun J, Rago C, Cheong I, Pagliarini R, Angenendt P, Rajagopalan H, et al. Glucose deprivation contributes to the development of KRAS pathway mutations in tumor cells. Science. 2009;325(5947):1555–9. doi:10.1126/science.1174229.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Daemen A, Peterson D, Sahu N, McCord R, Du X, Liu B, et al. Metabolite profiling stratifies pancreatic ductal adenocarcinomas into subtypes with distinct sensitivities to metabolic inhibitors. Proc Natl Acad Sci U S A. 2015;112(32):E4410–7. doi:10.1073/pnas.1501605112.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Capello M, Ferri-Borgogno S, Riganti C, Chattaragada MS, Principe M, Roux C, et al. Targeting the Warburg effect in cancer cells through ENO1 knockdown rescues oxidative phosphorylation and induces growth arrest. Oncotarget. 2016;7(5):5598–612. doi:10.18632/oncotarget.6798.

    PubMed  Google Scholar 

  8. Iacobuzio-Donahue CA, Ashfaq R, Maitra A, Adsay NV, Shen-Ong GL, Berg K, et al. Highly expressed genes in pancreatic ductal adenocarcinomas: a comprehensive characterization and comparison of the transcription profiles obtained from three major technologies. Cancer Res. 2003;63(24):8614–22.

    CAS  PubMed  Google Scholar 

  9. Amedei A, Niccolai E, Benagiano M, Della Bella C, Cianchi F, Bechi P, et al. Ex vivo analysis of pancreatic cancer-infiltrating T lymphocytes reveals that ENO-specific Tregs accumulate in tumor tissue and inhibit Th1/Th17 effector cell functions. Cancer Immunol Immunother. 2013;62(7):1249–60. doi:10.1007/s00262-013-1429-3.

    Article  CAS  PubMed  Google Scholar 

  10. Principe M, Ceruti P, Shih NY, Chattaragada MS, Rolla S, Conti L, et al. Targeting of surface alpha-enolase inhibits the invasiveness of pancreatic cancer cells. Oncotarget. 2015;6(13):11098–113. doi:10.18632/oncotarget.3572.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Goldman RD, Kaplan NO, Hall TC. Lactic dehydrogenase in human neoplastic tissues. Cancer Res. 1964;24:389–99.

    CAS  PubMed  Google Scholar 

  12. Le A, Cooper CR, Gouw AM, Dinavahi R, Maitra A, Deck LM, et al. Inhibition of lactate dehydrogenase A induces oxidative stress and inhibits tumor progression. Proc Natl Acad Sci U S A. 2010;107(5):2037–42. doi:10.1073/pnas.0914433107.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Mohammad GH, Olde Damink SW, Malago M, Dhar DK, Pereira SP. Pyruvate kinase M2 and lactate dehydrogenase A are overexpressed in pancreatic cancer and correlate with poor outcome. PLoS One. 2016;11(3):e0151635. doi:10.1371/journal.pone.0151635.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Cohen R, Neuzillet C, Tijeras-Raballand A, Faivre S, de Gramont A, Raymond E. Targeting cancer cell metabolism in pancreatic adenocarcinoma. Oncotarget. 2015;6(19):16832–47. doi:10.18632/oncotarget.4160.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Xie H, Hanai J, Ren JG, Kats L, Burgess K, Bhargava P, et al. Targeting lactate dehydrogenase – a inhibits tumorigenesis and tumor progression in mouse models of lung cancer and impacts tumor-initiating cells. Cell Metab. 2014;19(5):795–809. doi:10.1016/j.cmet.2014.03.003.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Weinberg F, Hamanaka R, Wheaton WW, Weinberg S, Joseph J, Lopez M, et al. Mitochondrial metabolism and ROS generation are essential for Kras-mediated tumorigenicity. Proc Natl Acad Sci U S A. 2010;107(19):8788–93. doi:10.1073/pnas.1003428107.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Son J, Lyssiotis CA, Ying H, Wang X, Hua S, Ligorio M, et al. Glutamine supports pancreatic cancer growth through a KRAS-regulated metabolic pathway. Nature. 2013;496(7443):101–5. doi:10.1038/nature12040.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Wise DR, DeBerardinis RJ, Mancuso A, Sayed N, Zhang XY, Pfeiffer HK, et al. Myc regulates a transcriptional program that stimulates mitochondrial glutaminolysis and leads to glutamine addiction. Proc Natl Acad Sci U S A. 2008;105(48):18782–7. doi:10.1073/pnas.0810199105.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Commisso C, Davidson SM, Soydaner-Azeloglu RG, Parker SJ, Kamphorst JJ, Hackett S, et al. Macropinocytosis of protein is an amino acid supply route in Ras-transformed cells. Nature. 2013;497(7451):633–7. doi:10.1038/nature12138.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Kamphorst JJ, Nofal M, Commisso C, Hackett SR, Lu W, Grabocka E, et al. Human pancreatic cancer tumors are nutrient poor and tumor cells actively scavenge extracellular protein. Cancer Res. 2015;75(3):544–53. doi:10.1158/0008-5472.CAN-14-2211.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Perera RM, Stoykova S, Nicolay BN, Ross KN, Fitamant J, Boukhali M, et al. Transcriptional control of autophagy-lysosome function drives pancreatic cancer metabolism. Nature. 2015;524(7565):361–5. doi:10.1038/nature14587.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Schutte M, Hruban RH, Geradts J, Maynard R, Hilgers W, Rabindran SK, et al. Abrogation of the Rb/p16 tumor-suppressive pathway in virtually all pancreatic carcinomas. Cancer Res. 1997;57(15):3126–30.

    CAS  PubMed  Google Scholar 

  23. Mavrakis KJ, McDonald 3rd ER, Schlabach MR, Billy E, Hoffman GR, de Weck A, et al. Disordered methionine metabolism in MTAP/CDKN2A-deleted cancers leads to dependence on PRMT5. Science. 2016;351(6278):1208–13. doi:10.1126/science.aad5944.

    Article  CAS  PubMed  Google Scholar 

  24. Hustinx SR, Hruban RH, Leoni LM, Iacobuzio-Donahue C, Cameron JL, Yeo CJ, et al. Homozygous deletion of the MTAP gene in invasive adenocarcinoma of the pancreas and in periampullary cancer: a potential new target for therapy. Cancer Biol Ther. 2005;4(1):83–6.

    Article  CAS  PubMed  Google Scholar 

  25. Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA, et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2012;2(5):401–4. doi:10.1158/2159-8290.CD-12-0095.

    Article  PubMed  Google Scholar 

  26. Chen ZH, Olopade OI, Savarese TM. Expression of methylthioadenosine phosphorylase cDNA in p16-, MTAP- malignant cells: restoration of methylthioadenosine phosphorylase-dependent salvage pathways and alterations of sensitivity to inhibitors of purine de novo synthesis. Mol Pharmacol. 1997;52(5):903–11.

    Article  CAS  PubMed  Google Scholar 

  27. Kindler HL, Burris 3rd HA, Sandler AB, Oliff IA. A phase II multicenter study of l-alanosine, a potent inhibitor of adenine biosynthesis, in patients with MTAP-deficient cancer. Investig New Drugs. 2009;27(1):75–81. doi:10.1007/s10637-008-9160-1.

    Article  CAS  Google Scholar 

  28. Kryukov GV, Wilson FH, Ruth JR, Paulk J, Tsherniak A, Marlow SE, et al. MTAP deletion confers enhanced dependency on the PRMT5 arginine methyltransferase in cancer cells. Science. 2016;351(6278):1214–8. doi:10.1126/science.aad5214.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Jansson M, Durant ST, Cho EC, Sheahan S, Edelmann M, Kessler B, et al. Arginine methylation regulates the p53 response. Nat Cell Biol. 2008;10(12):1431–9. doi:10.1038/ncb1802.

    Article  CAS  PubMed  Google Scholar 

  30. Hou Z, Peng H, Ayyanathan K, Yan KP, Langer EM, Longmore GD, et al. The LIM protein AJUBA recruits protein arginine methyltransferase 5 to mediate SNAIL-dependent transcriptional repression. Mol Cell Biol. 2008;28(10):3198–207. doi:10.1128/MCB.01435-07.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Marjon K, Cameron MJ, Quang P, Clasquin MF, Mandley E, Kunii K, et al. MTAP deletions in cancer create vulnerability to targeting of the MAT2A/PRMT5/RIOK1 axis. Cell Rep. 2016;15(3):574–87. doi:10.1016/j.celrep.2016.03.043.

    Article  CAS  PubMed  Google Scholar 

  32. Myers K, Gagou ME, Zuazua-Villar P, Rodriguez R, Meuth M. ATR and Chk1 suppress a caspase-3-dependent apoptotic response following DNA replication stress. PLoS Genet. 2009;5(1):e1000324. doi:10.1371/journal.pgen.1000324.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  33. Murphy KM, Brune KA, Griffin C, Sollenberger JE, Petersen GM, Bansal R, et al. Evaluation of candidate genes MAP2K4, MADH4, ACVR1B, and BRCA2 in familial pancreatic cancer: deleterious BRCA2 mutations in 17%. Cancer Res. 2002;62(13):3789–93.

    CAS  PubMed  Google Scholar 

  34. Risch HA, McLaughlin JR, Cole DE, Rosen B, Bradley L, Fan I, et al. Population BRCA1 and BRCA2 mutation frequencies and cancer penetrances: a kin-cohort study in Ontario. Canada J Natl Cancer Inst. 2006;98(23):1694–706. doi:10.1093/jnci/djj465.

    Article  CAS  PubMed  Google Scholar 

  35. Waddell N, Pajic M, Patch AM, Chang DK, Kassahn KS, Bailey P, et al. Whole genomes redefine the mutational landscape of pancreatic cancer. Nature. 2015;518(7540):495–501. doi:10.1038/nature14169.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Bailey P, Chang DK, Nones K, Johns AL, Patch AM, Gingras MC, et al. Genomic analyses identify molecular subtypes of pancreatic cancer. Nature. 2016;531(7592):47–52. doi:10.1038/nature16965.

    Article  CAS  PubMed  Google Scholar 

  37. Lowery MA, Kelsen DP, Stadler ZK, Yu KH, Janjigian YY, Ludwig E, et al. An emerging entity: pancreatic adenocarcinoma associated with a known BRCA mutation: clinical descriptors, treatment implications, and future directions. Oncologist. 2011;16(10):1397–402. doi:10.1634/theoncologist.2011-0185.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Jones S, Hruban RH, Kamiyama M, Borges M, Zhang X, Parsons DW, et al. Exomic sequencing identifies PALB2 as a pancreatic cancer susceptibility gene. Science. 2009;324(5924):217. doi:10.1126/science.1171202.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Tischkowitz MD, Sabbaghian N, Hamel N, Borgida A, Rosner C, Taherian N, et al. Analysis of the gene coding for the BRCA2-interacting protein PALB2 in familial and sporadic pancreatic cancer. Gastroenterology. 2009;137(3):1183–6. doi:10.1053/j.gastro.2009.06.055.

    Article  PubMed  PubMed Central  Google Scholar 

  40. Roberts NJ, Norris AL, Petersen GM, Bondy ML, Brand R, Gallinger S, et al. Whole genome sequencing defines the genetic heterogeneity of familial pancreatic cancer. Cancer Discov. 2016;6(2):166–75. doi:10.1158/2159-8290.CD-15-0402.

    Article  CAS  PubMed  Google Scholar 

  41. Biankin AV, Waddell N, Kassahn KS, Gingras MC, Muthuswamy LB, Johns AL, et al. Pancreatic cancer genomes reveal aberrations in axon guidance pathway genes. Nature. 2012;491(7424):399–405. doi:10.1038/nature11547.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Russell R, Perkhofer L, Liebau S, Lin Q, Lechel A, Feld FM, et al. Loss of ATM accelerates pancreatic cancer formation and epithelial-mesenchymal transition. Nat Commun. 2015;6:7677. doi:10.1038/ncomms8677.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Ray PD, Huang BW, Tsuji Y. Reactive oxygen species (ROS) homeostasis and redox regulation in cellular signaling. Cell Signal. 2012;24(5):981–90. doi:10.1016/j.cellsig.2012.01.008.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Mateo J, Carreira S, Sandhu S, Miranda S, Mossop H, Perez-Lopez R, et al. DNA-repair defects and olaparib in metastatic prostate cancer. N Engl J Med. 2015;373(18):1697–708. doi:10.1056/NEJMoa1506859.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Prevo R, Fokas E, Reaper PM, Charlton PA, Pollard JR, McKenna WG, et al. The novel ATR inhibitor VE-821 increases sensitivity of pancreatic cancer cells to radiation and chemotherapy. Cancer Biol Ther. 2012;13(11):1072–81. doi:10.4161/cbt.21093.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Weber AM, Ryan AJ. ATM and ATR as therapeutic targets in cancer. Pharmacol Ther. 2015;149:124–38. doi:10.1016/j.pharmthera.2014.12.001.

    Article  CAS  PubMed  Google Scholar 

  47. Scintu M, Vitale R, Prencipe M, Gallo AP, Bonghi L, Valori VM, et al. Genomic instability and increased expression of BUB1B and MAD2L1 genes in ductal breast carcinoma. Cancer Lett. 2007;254(2):298–307. doi:10.1016/j.canlet.2007.03.021.

    Article  CAS  PubMed  Google Scholar 

  48. Long J, Zhang Z, Liu Z, Xu Y, Ge C. Identification of genes and pathways associated with pancreatic ductal adenocarcinoma by bioinformatics analyses. Oncol Lett. 2016;11(2):1391–7. doi:10.3892/ol.2015.4042.

    PubMed  Google Scholar 

  49. Kapoor A, Yao W, Ying H, Hua S, Liewen A, Wang Q, et al. Yap1 activation enables bypass of oncogenic Kras addiction in pancreatic cancer. Cell. 2014;158(1):185–97. doi:10.1016/j.cell.2014.06.003.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Shain AH, Giacomini CP, Matsukuma K, Karikari CA, Bashyam MD, Hidalgo M, et al. Convergent structural alterations define SWItch/Sucrose NonFermentable (SWI/SNF) chromatin remodeler as a central tumor suppressive complex in pancreatic cancer. Proc Natl Acad Sci U S A. 2012;109(5):E252–9. doi:10.1073/pnas.1114817109.

    Article  CAS  PubMed  Google Scholar 

  51. Guan B, Rahmanto YS, Wu RC, Wang Y, Wang Z, Wang TL et al. Roles of deletion of Arid1a, a tumor suppressor, in mouse ovarian tumorigenesis. J Natl Cancer Inst. 2014;106(7). doi:10.1093/jnci/dju146.

    Google Scholar 

  52. Witkiewicz AK, McMillan EA, Balaji U, Baek G, Lin WC, Mansour J, et al. Whole-exome sequencing of pancreatic cancer defines genetic diversity and therapeutic targets. Nat Commun. 2015;6:6744. doi:10.1038/ncomms7744.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Helming KC, Wang X, Wilson BG, Vazquez F, Haswell JR, Manchester HE, et al. ARID1B is a specific vulnerability in ARID1A-mutant cancers. Nat Med. 2014;20(3):251–4. doi:10.1038/nm.3480.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. van den Beucken T, Koch E, Chu K, Rupaimoole R, Prickaerts P, Adriaens M, et al. Hypoxia promotes stem cell phenotypes and poor prognosis through epigenetic regulation of DICER. Nat Commun. 2014;5:5203. doi:10.1038/ncomms6203.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  55. Ougolkov AV, Bilim VN, Billadeau DD. Regulation of pancreatic tumor cell proliferation and chemoresistance by the histone methyltransferase enhancer of zeste homologue 2. Clin Cancer Res. 2008;14(21):6790–6. doi:10.1158/1078-0432.CCR-08-1013.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Qi W, Chan H, Teng L, Li L, Chuai S, Zhang R, et al. Selective inhibition of Ezh2 by a small molecule inhibitor blocks tumor cells proliferation. Proc Natl Acad Sci U S A. 2012;109(52):21360–5. doi:10.1073/pnas.1210371110.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Kim KH, Kim W, Howard TP, Vazquez F, Tsherniak A, Wu JN, et al. SWI/SNF-mutant cancers depend on catalytic and non-catalytic activity of EZH2. Nat Med. 2015;21(12):1491–6. doi:10.1038/nm.3968.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Pal S, Vishwanath SN, Erdjument-Bromage H, Tempst P, Sif S. Human SWI/SNF-associated PRMT5 methylates histone H3 arginine 8 and negatively regulates expression of ST7 and NM23 tumor suppressor genes. Mol Cell Biol. 2004;24(21):9630–45. doi:10.1128/MCB.24.21.9630-9645.2004.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Tae S, Karkhanis V, Velasco K, Yaneva M, Erdjument-Bromage H, Tempst P, et al. Bromodomain protein 7 interacts with PRMT5 and PRC2, and is involved in transcriptional repression of their target genes. Nucleic Acids Res. 2011;39(13):5424–38. doi:10.1093/nar/gkr170.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. van Hattem WA, Carvalho R, Li A, Offerhaus GJ, Goggins M. Amplification of EMSY gene in a subset of sporadic pancreatic adenocarcinomas. Int J Clin Exp Pathol. 2008;1(4):343–51.

    PubMed  PubMed Central  Google Scholar 

  61. Varier RA, Carrillo de Santa Pau E, van der Groep P, Lindeboom RG, Matarese F, Mensinga A, et al. Recruitment of the mammalian histone-modifying EMSY complex to target genes is regulated by ZNF131. J Biol Chem. 2016;291(14):7313–24. doi:10.1074/jbc.M115.701227.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Zhao Y, Sun J, Zhang H, Guo S, Gu J, Wang W, et al. High-frequency aberrantly methylated targets in pancreatic adenocarcinoma identified via global DNA methylation analysis using methylCap-seq. Clin Epigenetics. 2014;6(1):18. doi:10.1186/1868-7083-6-18.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  63. Giaginis C, Damaskos C, Koutsounas I, Zizi-Serbetzoglou A, Tsoukalas N, Patsouris E et al. Histone deacetylase (HDAC)-1, -2, -4 and -6 expression in human pancreatic adenocarcinoma: associations with clinicopathological parameters, tumor proliferative capacity and patients’ survival. BMC Gastroenterol. 2015;15:148. doi:10.1186/s12876-015-0379-y.

    Google Scholar 

  64. Engelmann D, Putzer BM. Emerging from the shade of p53 mutants: N-terminally truncated variants of the p53 family in EMT signaling and cancer progression. Sci Signal. 2014;7(345):re9. doi:10.1126/scisignal.2005699.

    Article  PubMed  CAS  Google Scholar 

  65. Li N, Singh S, Cherukuri P, Li H, Yuan Z, Ellisen LW, et al. Reciprocal intraepithelial interactions between TP63 and hedgehog signaling regulate quiescence and activation of progenitor elaboration by mammary stem cells. Stem Cells. 2008;26(5):1253–64. doi:10.1634/stemcells.2007-0691.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Ma J, Meng Y, Kwiatkowski DJ, Chen X, Peng H, Sun Q, et al. Mammalian target of rapamycin regulates murine and human cell differentiation through STAT3/p63/Jagged/Notch cascade. J Clin Invest. 2010;120(1):103–14. doi:10.1172/JCI37964.

    Article  CAS  PubMed  Google Scholar 

  67. Childs EJ, Mocci E, Campa D, Bracci PM, Gallinger S, Goggins M, et al. Common variation at 2p13.3, 3q29, 7p13 and 17q25.1 associated with susceptibility to pancreatic cancer. Nat Genet. 2015;47(8):911–6. doi:10.1038/ng.3341.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Su X, Chakravarti D, Cho MS, Liu L, Gi YJ, Lin YL, et al. TAp63 suppresses metastasis through coordinate regulation of Dicer and miRNAs. Nature. 2010;467(7318):986–90. doi:10.1038/nature09459.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Su X, Gi YJ, Chakravarti D, Chan IL, Zhang A, Xia X, et al. TAp63 is a master transcriptional regulator of lipid and glucose metabolism. Cell Metab. 2012;16(4):511–25. doi:10.1016/j.cmet.2012.09.006.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Danilov AV, Neupane D, Nagaraja AS, Feofanova EV, Humphries LA, DiRenzo J, et al. DeltaNp63alpha-mediated induction of epidermal growth factor receptor promotes pancreatic cancer cell growth and chemoresistance. PLoS One. 2011;6(10):e26815. doi:10.1371/journal.pone.0026815.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Moreno-Bueno G, Portillo F, Cano A. Transcriptional regulation of cell polarity in EMT and cancer. Oncogene. 2008;27(55):6958–69. doi:10.1038/onc.2008.346.

    Article  CAS  PubMed  Google Scholar 

  72. Gregory PA, Bert AG, Paterson EL, Barry SC, Tsykin A, Farshid G, et al. The miR-200 family and miR-205 regulate epithelial to mesenchymal transition by targeting ZEB1 and SIP1. Nat Cell Biol. 2008;10(5):593–601. doi:10.1038/ncb1722.

    Article  CAS  PubMed  Google Scholar 

  73. Burk U, Schubert J, Wellner U, Schmalhofer O, Vincan E, Spaderna S, et al. A reciprocal repression between ZEB1 and members of the miR-200 family promotes EMT and invasion in cancer cells. EMBO Rep. 2008;9(6):582–9. doi:10.1038/embor.2008.74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Ellenrieder V, Hendler SF, Boeck W, Seufferlein T, Menke A, Ruhland C, et al. Transforming growth factor beta1 treatment leads to an epithelial-mesenchymal transdifferentiation of pancreatic cancer cells requiring extracellular signal-regulated kinase 2 activation. Cancer Res. 2001;61(10):4222–8.

    CAS  PubMed  Google Scholar 

  75. Singh A, Greninger P, Rhodes D, Koopman L, Violette S, Bardeesy N, et al. A gene expression signature associated with “K-Ras addiction” reveals regulators of EMT and tumor cell survival. Cancer Cell. 2009;15(6):489–500. doi:10.1016/j.ccr.2009.03.022.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Rosenbluh J, Nijhawan D, Cox AG, Li X, Neal JT, Schafer EJ, et al. beta-Catenin-driven cancers require a YAP1 transcriptional complex for survival and tumorigenesis. Cell. 2012;151(7):1457–73. doi:10.1016/j.cell.2012.11.026.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Cebola I, Rodriguez-Segui SA, Cho CH, Bessa J, Rovira M, Luengo M, et al. TEAD and YAP regulate the enhancer network of human embryonic pancreatic progenitors. Nat Cell Biol. 2015;17(5):615–26. doi:10.1038/ncb3160.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Shao DD, Xue W, Krall EB, Bhutkar A, Piccioni F, Wang X, et al. KRAS and YAP1 converge to regulate EMT and tumor survival. Cell. 2014;158(1):171–84. doi:10.1016/j.cell.2014.06.004.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Mei L, Du W, Ma WW. Targeting stromal microenvironment in pancreatic ductal adenocarcinoma: controversies and promises. J Gastrointest Oncol. 2016;7(3):487–94. doi:10.21037/jgo.2016.03.03.

    Article  PubMed  PubMed Central  Google Scholar 

  80. Olive KP, Jacobetz MA, Davidson CJ, Gopinathan A, McIntyre D, Honess D, et al. Inhibition of Hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science. 2009;324(5933):1457–61. doi:10.1126/science.1171362.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Remmers N, Anderson JM, Linde EM, DiMaio DJ, Lazenby AJ, Wandall HH, et al. Aberrant expression of mucin core proteins and o-linked glycans associated with progression of pancreatic cancer. Clin Cancer Res. 2013;19(8):1981–93. doi:10.1158/1078-0432.CCR-12-2662.

    Article  CAS  PubMed  Google Scholar 

  82. Radhakrishnan P, Dabelsteen S, Madsen FB, Francavilla C, Kopp KL, Steentoft C, et al. Immature truncated O-glycophenotype of cancer directly induces oncogenic features. Proc Natl Acad Sci U S A. 2014;111(39):E4066–75. doi:10.1073/pnas.1406619111.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Hockl PF, Wolosiuk A, Perez-Saez JM, Bordoni AV, Croci DO, Toum-Terrones Y, et al. Glyco-nano-oncology: novel therapeutic opportunities by combining small and sweet. Pharmacol Res. 2016;109:45–54. doi:10.1016/j.phrs.2016.02.005.

    Article  CAS  PubMed  Google Scholar 

  84. Berberat PO, Friess H, Wang L, Zhu Z, Bley T, Frigeri L, et al. Comparative analysis of galectins in primary tumors and tumor metastasis in human pancreatic cancer. J Histochem Cytochem. 2001;49(4):539–49.

    Article  CAS  PubMed  Google Scholar 

  85. Song S, Ji B, Ramachandran V, Wang H, Hafley M, Logsdon C, et al. Overexpressed galectin-3 in pancreatic cancer induces cell proliferation and invasion by binding Ras and activating Ras signaling. PLoS One. 2012;7(8):e42699. doi:10.1371/journal.pone.0042699.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Martinez-Bosch N, Fernandez-Barrena MG, Moreno M, Ortiz-Zapater E, Munne-Collado J, Iglesias M, et al. Galectin-1 drives pancreatic carcinogenesis through stroma remodeling and Hedgehog signaling activation. Cancer Res. 2014;74(13):3512–24. doi:10.1158/0008-5472.CAN-13-3013.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Martinez-Bosch N, Navarro P. Targeting Galectin-1 in pancreatic cancer: immune surveillance on guard. Oncoimmunology. 2014;3(8):e952201. doi:10.4161/21624011.2014.952201.

    Article  PubMed  PubMed Central  Google Scholar 

  88. Pour PM, Bell RH, Batra SK. Neural invasion in the staging of pancreatic cancer. Pancreas. 2003;26(4):322–5.

    Article  PubMed  Google Scholar 

  89. Li J, Ma Q, Liu H, Guo K, Li F, Li W, et al. Relationship between neural alteration and perineural invasion in pancreatic cancer patients with hyperglycemia. PLoS One. 2011;6(2):e17385. doi:10.1371/journal.pone.0017385.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Kayahara M, Nakagawara H, Kitagawa H, Ohta T. The nature of neural invasion by pancreatic cancer. Pancreas. 2007;35(3):218–23. doi:10.1097/mpa.0b013e3180619677.

    Article  PubMed  Google Scholar 

  91. Salvioli B, Bovara M, Barbara G, De Ponti F, Stanghellini V, Tonini M, et al. Neurology and neuropathology of the pancreatic innervation. JOP. 2002;3(2):26–33.

    PubMed  Google Scholar 

  92. Eichmann A, Makinen T, Alitalo K. Neural guidance molecules regulate vascular remodeling and vessel navigation. Genes Dev. 2005;19(9):1013–21. doi:10.1101/gad.1305405.

    Article  CAS  PubMed  Google Scholar 

  93. Brose K, Bland KS, Wang KH, Arnott D, Henzel W, Goodman CS, et al. Slit proteins bind Robo receptors and have an evolutionarily conserved role in repulsive axon guidance. Cell. 1999;96(6):795–806.

    Article  CAS  PubMed  Google Scholar 

  94. Ypsilanti AR, Zagar Y, Chedotal A. Moving away from the midline: new developments for Slit and Robo. Development. 2010;137(12):1939–52. doi:10.1242/dev.044511.

    Article  CAS  PubMed  Google Scholar 

  95. Gohrig A, Detjen KM, Hilfenhaus G, Korner JL, Welzel M, Arsenic R, et al. Axon guidance factor SLIT2 inhibits neural invasion and metastasis in pancreatic cancer. Cancer Res. 2014;74(5):1529–40. doi:10.1158/0008-5472.CAN-13-1012.

    Article  PubMed  CAS  Google Scholar 

  96. Nones K, Waddell N, Song S, Patch AM, Miller D, Johns A, et al. Genome-wide DNA methylation patterns in pancreatic ductal adenocarcinoma reveal epigenetic deregulation of SLIT-ROBO, ITGA2 and MET signaling. Int J Cancer. 2014;135(5):1110–8. doi:10.1002/ijc.28765.

    Article  CAS  PubMed  Google Scholar 

  97. Taucher V, Mangge H, Haybaeck J. Non-coding RNAs in pancreatic cancer: challenges and opportunities for clinical application. Cell Oncol (Dordr). 2016;39(4):295–318. doi:10.1007/s13402-016-0275-7.

    Article  CAS  Google Scholar 

  98. Bloomston M, Frankel WL, Petrocca F, Volinia S, Alder H, Hagan JP, et al. MicroRNA expression patterns to differentiate pancreatic adenocarcinoma from normal pancreas and chronic pancreatitis. JAMA. 2007;297(17):1901–8. doi:10.1001/jama.297.17.1901.

    Article  CAS  PubMed  Google Scholar 

  99. Han S, Gonzalo DH, Feely M, Delitto D, Behrns KE, Beveridge M, et al. The pancreatic tumor microenvironment drives changes in miRNA expression that promote cytokine production and inhibit migration by the tumor associated stroma. Oncotarget. 2016; doi:10.18632/oncotarget.10722.

    Google Scholar 

  100. Zhu ZM, Xu YF, Su QJ, Du JD, Tan XL, Tu YL, et al. Prognostic significance of microRNA-141 expression and its tumor suppressor function in human pancreatic ductal adenocarcinoma. Mol Cell Biochem. 2014;388(1–2):39–49. doi:10.1007/s11010-013-1897-y.

    Article  CAS  PubMed  Google Scholar 

  101. Imanaka Y, Tsuchiya S, Sato F, Shimada Y, Shimizu K, Tsujimoto G. MicroRNA-141 confers resistance to cisplatin-induced apoptosis by targeting YAP1 in human esophageal squamous cell carcinoma. J Hum Genet. 2011;56(4):270–6. doi:10.1038/jhg.2011.1.

    Article  CAS  PubMed  Google Scholar 

  102. Gironella M, Seux M, Xie MJ, Cano C, Tomasini R, Gommeaux J, et al. Tumor protein 53-induced nuclear protein 1 expression is repressed by miR-155, and its restoration inhibits pancreatic tumor development. Proc Natl Acad Sci U S A. 2007;104(41):16170–5. doi:10.1073/pnas.0703942104.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Kuninty PR, Schnittert J, Storm G, Prakash J. MicroRNA targeting to modulate tumor microenvironment. Front Oncol. 2016;6:3. doi:10.3389/fonc.2016.00003.

    Article  PubMed  PubMed Central  Google Scholar 

  104. Zhou K, Nguyen LH, Miller JB, Yan Y, Kos P, Xiong H, et al. Modular degradable dendrimers enable small RNAs to extend survival in an aggressive liver cancer model. Proc Natl Acad Sci U S A. 2016;113(3):520–5. doi:10.1073/pnas.1520756113.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Jennifer H. Choe .

Editor information

Editors and Affiliations

Section Editor information

Rights and permissions

Reprints and permissions

Copyright information

© 2016 Springer Science+Business Media LLC

About this entry

Cite this entry

Choe, J.H., Abbruzzese, J.L. (2016). Emerging Therapeutic Targets in Pancreatic Adenocarcinoma. In: Neoptolemos, J., Urrutia, R., Abbruzzese, J., Büchler, M. (eds) Pancreatic Cancer. Springer, New York, NY. https://doi.org/10.1007/978-1-4939-6631-8_92-1

Download citation

  • DOI: https://doi.org/10.1007/978-1-4939-6631-8_92-1

  • Received:

  • Accepted:

  • Published:

  • Publisher Name: Springer, New York, NY

  • Print ISBN: 978-1-4939-6631-8

  • Online ISBN: 978-1-4939-6631-8

  • eBook Packages: Springer Reference Biomedicine and Life SciencesReference Module Biomedical and Life Sciences

Publish with us

Policies and ethics