Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Inhibition of geranylgeranyl diphosphate synthase is a novel therapeutic strategy for pancreatic ductal adenocarcinoma

Abstract

Rab proteins play an essential role in regulating intracellular membrane trafficking processes. Rab activity is dependent upon geranylgeranylation, a post-translational modification that involves the addition of 20-carbon isoprenoid chains via the enzyme geranylgeranyl transferase (GGTase) II. We have focused on the development of inhibitors against geranylgeranyl diphosphate synthase (GGDPS), which generates the isoprenoid donor (GGPP), as anti-Rab agents. Pancreatic ductal adenocarcinoma (PDAC) is characterized by abnormal mucin production and these mucins play important roles in tumor development, metastasis and chemo-resistance. We hypothesized that GGDPS inhibitor (GGDPSi) treatment would induce PDAC cell death by disrupting mucin trafficking, thereby inducing the unfolded protein response pathway (UPR) and apoptosis. To this end, we evaluated the effects of RAM2061, a potent GGDPSi, against PDAC. Our studies revealed that GGDPSi treatment activates the UPR and triggers apoptosis in a variety of human and mouse PDAC cell lines. Furthermore, GGDPSi treatment was found to disrupt the intracellular trafficking of key mucins such as MUC1. These effects could be recapitulated by incubation with a specific GGTase II inhibitor, but not a GGTase I inhibitor, consistent with the effect being dependent on disruption of Rab-mediated activities. In addition, siRNA-mediated knockdown of GGDPS induces upregulation of UPR markers and disrupts MUC1 trafficking in PDAC cells. Experiments in two mouse models of PDAC demonstrated that GGDPSi treatment significantly slows tumor growth. Collectively, these data support further development of GGDPSi therapy as a novel strategy for the treatment of PDAC.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7
Fig. 8

Similar content being viewed by others

References

  1. Luberice K, Downs D, Sadowitz B, Ross S, Rosemurgy A. Has survival improved following resection for pancreatic adenocarcinoma? Am J Surg. 2017;214:341–6.

    Article  Google Scholar 

  2. Suh H, Pillai K, Morris DL. Mucins in pancreatic cancer: biological role, implications in carcinogenesis and applications in diagnosis and therapy. Am J Cancer Res. 2017;7:1372–83.

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Besmer DM, Curry JM, Roy LD, Tinder TL, Sahraei M, Schettini J, et al. Pancreatic ductal adenocarcinoma mice lacking mucin 1 have a profound defect in tumor growth and metastasis. Cancer Res. 2011;71:4432–42.

    Article  CAS  Google Scholar 

  4. Jonckheere N, Skrypek N, Van Seuningen I. Mucins and tumor resistance to chemotherapeutic drugs. Biochim Biophys Acta. 2014;1846:142–51.

    CAS  PubMed  Google Scholar 

  5. Kaur S, Kumar S, Momi N, Sasson AR, Batra SK. Mucins in pancreatic cancer and its microenvironment. Nat Rev Gastroenterol Hepatol. 2013;10:607–20.

    Article  CAS  Google Scholar 

  6. Taylor-Papadimitriou J, Burchell JM, Graham R, Beatson R. Latest developments in MUC1 immunotherapy. Biochem Soc Trans. 2018;46:659–68.

    Article  CAS  Google Scholar 

  7. Haney SL, Wills VS, Wiemer DF, Holstein SA. Recent advances in the development of mammalian geranylgeranyl diphosphate synthase inhibitors. Molecules. 2017;22:886.

    Article  Google Scholar 

  8. Wills VS, Allen C, Holstein SA, Wiemer DF. Potent triazole bisphosphonate inhibitor of geranylgeranyl diphosphate synthase. ACS Med Chem Lett. 2015;6:1195–8.

    Article  CAS  Google Scholar 

  9. Haney SL, Chhonker YS, Varney ML, Talmon G, Murry DJ, Holstein SA. Preclinical investigation of a potent geranylgeranyl diphosphate synthase inhibitor. Invest New Drugs. 2018;36:810–8.

    Article  CAS  Google Scholar 

  10. Matthiesen RA, Varney ML, Xu PC, Rier AS, Wiemer DF, Holstein SA. alpha-Methylation enhances the potency of isoprenoid triazole bisphosphonates as geranylgeranyl diphosphate synthase inhibitors. Bioorg Med Chem. 2018;26:376–85.

    Article  CAS  Google Scholar 

  11. Holstein SA, Hohl RJ. Isoprenoid biosynthetic pathway inhibition disrupts monoclonal protein secretion and induces the unfolded protein response pathway in multiple myeloma cells. Leuk Res. 2011;35:551–9.

    Article  CAS  Google Scholar 

  12. Born EJ, Hartman SV, Holstein SA. Targeting HSP90 and monoclonal protein trafficking modulates the unfolded protein response, chaperone regulation and apoptosis in myeloma cells.Blood Cancer J. 2013;3:e167.

    Article  CAS  Google Scholar 

  13. Sorensen AL, Reis CA, Tarp MA, Mandel U, Ramachandran K, Sankaranarayanan V, et al. Chemoenzymatically synthesized multimeric Tn/STn MUC1 glycopeptides elicit cancer-specific anti-MUC1 antibody responses and override tolerance. Glycobiology. 2006;16:96–107.

    Article  CAS  Google Scholar 

  14. Taylor-Papadimitriou J, Peterson JA, Arklie J, Burchell J, Ceriani RL, Bodmer WF. Monoclonal antibodies to epithelium-specific components of the human milk fat globule membrane: production and reaction with cells in culture. Int J Cancer. 1981;28:17–21.

    Article  CAS  Google Scholar 

  15. Hanson RL, Hollingsworth MA. Functional Consequences of Differential O-glycosylation of MUC1, MUC4, and MUC16 (Downstream Effects on Signaling). Biomolecules. 2016;6:E34.

    Article  Google Scholar 

  16. Misumi Y, Miki K, Takatsuki A, Tamura G, Ikehara Y. Novel blockade by brefeldin A of intracellular transport of secretory proteins in cultured rat hepatocytes. J Biol Chem. 1986;261:11398–403.

    CAS  PubMed  Google Scholar 

  17. Deer EL, Gonzalez-Hernandez J, Coursen JD, Shea JE, Ngatia J, Scaife CL, et al. Phenotype and genotype of pancreatic cancer cell lines. Pancreas. 2010;39:425–35.

    Article  CAS  Google Scholar 

  18. Yu DC, Liu J, Chen J, Shao JJ, Shen X, Xia HG, et al. GGPPS1 predicts the biological character of hepatocellular carcinoma in patients with cirrhosis. BMC Cancer. 2014;14:248.

    Article  Google Scholar 

  19. Igarashi T, Araki K, Yokobori T, Altan B, Yamanaka T, Ishii N, et al. Association of RAB5 overexpression in pancreatic cancer with cancer progression and poor prognosis via E-cadherin suppression. Oncotarget. 2017;8:12290–300.

    PubMed  PubMed Central  Google Scholar 

  20. Li Y, Sun X, Ji D, Kong X, Liu D, Zhao Z, et al. Expression of Rab5a correlates with tumor progression in pancreatic carcinoma. Virchows Arch. 2017;470:527–36.

    Article  CAS  Google Scholar 

  21. Nath S, Mukherjee P. MUC1: a multifaceted oncoprotein with a key role in cancer progression. Trends Mol Med. 2014;20:332–42.

    Article  CAS  Google Scholar 

  22. Liu X, Yuan Z, Chung M. MUC1 intra-cellular trafficking is clathrin, dynamin, and rab5 dependent. Biochem Biophys Res Commun. 2008;376:688–93.

    Article  CAS  Google Scholar 

  23. Jin C, Rajabi H, Pitroda S, Li A, Kharbanda A, Weichselbaum R, et al. Cooperative interaction between the MUC1-C oncoprotein and the Rab31 GTPase in estrogen receptor-positive breast cancer cells. PLoS ONE. 2012;7:e39432.

    Article  CAS  Google Scholar 

  24. Kinlough CL, McMahan RJ, Poland PA, Bruns JB, Harkleroad KL, Stremple RJ, et al. Recycling of MUC1 is dependent on its palmitoylation. J Biol Chem. 2006;281:12112–22.

    Article  CAS  Google Scholar 

  25. Wissniowski TT, Meister S, Hahn EG, Kalden JR, Voll R, Ocker M. Mucin production determines sensitivity to bortezomib and gemcitabine in pancreatic cancer cells. Int J Oncol. 2012;40:1581–9.

    CAS  PubMed  Google Scholar 

  26. Marten A, Zeiss N, Serba S, Mehrle S, von Lilienfeld-Toal M, Schmidt J. Bortezomib is ineffective in an orthotopic mouse model of pancreatic adenocarcinoma. Mol Cancer Ther. 2008;7:3624–31.

    Article  Google Scholar 

  27. Wang H, Cao Q, Dudek AZ. Phase II study of panobinostat and bortezomib in patients with pancreatic cancer progressing on gemcitabine-based therapy. Anticancer Res. 2012;32:1027–31.

    CAS  PubMed  Google Scholar 

  28. Iwamura T, Taniguchi S, Kitamura N, Yamanari H, Kojima A, Hidaka K, et al. Correlation between CA19-9 production in vitro and histological grades of differentiation in vivo in clones isolated from a human pancreatic cancer cell line (SUIT-2). J Gastroenterol Hepatol. 1992;7:512–9.

    Article  CAS  Google Scholar 

  29. Hingorani SR, Wang L, Multani AS, Combs C, Deramaudt TB, Hruban RH, et al. Trp53R172H and KrasG12D cooperate to promote chromosomal instability and widely metastatic pancreatic ductal adenocarcinoma in mice. Cancer Cell. 2005;7:469–83.

    Article  CAS  Google Scholar 

  30. Coxon FP, Joachimiak L, Najumudeen AK, Breen G, Gmach J, Oetken-Lindholm C, et al. Synthesis and characterization of novel phosphonocarboxylate inhibitors of RGGT. Eur J Med Chem. 2014;84:77–89.

    Article  CAS  Google Scholar 

  31. Chhonker YS, Haney SL, Bala V, Holstein SA, Murry DJ. Simultaneous Quantitation of Isoprenoid Pyrophosphates in Plasma and Cancer Cells Using LC-MS/MS. Molecules. 2018;23:E3275.

    Article  Google Scholar 

  32. Holstein SA, Hohl RJ. Interaction of cytosine arabinoside and lovastatin in human leukemia cells. Leuk Res. 2001;25:651–60.

    Article  CAS  Google Scholar 

  33. Qi W, Schultes BC, Liu D, Kuzma M, Decker W, Madiyalakan R. Characterization of an anti-MUC1 monoclonal antibody with potential as a cancer vaccine. Hybrid Hybridomics. 2001;20:313–24.

    Article  CAS  Google Scholar 

  34. Yan Y, Gao YY, Liu BQ, Niu XF, Zhuang Y, Wang HQ. Resveratrol-induced cytotoxicity in human Burkitt’s lymphoma cells is coupled to the unfolded protein response. BMC Cancer. 2010;10:445.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by the Fred & Pamela Buffett Cancer Center Support Grant from the National Cancer Institute under award P30 CA036727 and the National Institutes of Health P50CA127297. The authors wish to acknowledge the support of the Fred & Pamela Buffett Cancer Center Advanced Microscopy Core Facility and the Flow Cytometry Research Facility, supported by the National Cancer Institute under award P30 CA036727. We thank Janice A. Taylor and James R. Talaska of the Advanced Microscopy Core Facility for providing assistance with confocal microscopy.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Sarah A. Holstein.

Ethics declarations

Conflict of interest

The authors have no relevant conflicts of interest to disclose.

Additional information

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Haney, S.L., Varney, M.L., Chhonker, Y.S. et al. Inhibition of geranylgeranyl diphosphate synthase is a novel therapeutic strategy for pancreatic ductal adenocarcinoma. Oncogene 38, 5308–5320 (2019). https://doi.org/10.1038/s41388-019-0794-6

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41388-019-0794-6

Search

Quick links