Skip to main content

Inhibition of mTOR in Neuroendocrine Neoplasms of the Digestive Tract

  • Chapter
  • First Online:

Abstract

The clinical behavior of neuroendocrine neoplasms (NEN) is highly variable. NEN may present as indolent tumors, usually well differentiated, that have limited impact on performance status but are detected at late stages. NEN may also display more aggressive behaviors when cancer cells are poorly differentiated, leading to very rapid tumor growth severely impairing patient general conditions. The term carcinoid was formerly used to identify well-differentiated slowly growing NEN and is actually replaced by the term neuroendocrine tumors G1/G2. Somatostatin analogs have improved the clinical management of patients with NEN by controlling carcinoid symptoms (flushing, diarrhea) and delaying tumor progression. The mammalian target of rapamycin (mTOR), a main protein kinase downstream to the phosphoinositide 3-kinase/Akt signaling pathway, appears as an important intracellular mediator involved in multiple cellular functions including proliferation, differentiation, apoptosis, tumorigenesis, and angiogenesis. Alterations in the normal activity of mTOR and of mTOR-related kinases in this pathway have been found in a diversity of human tumors, including NEN; therefore, mTOR pathway represents an attractive target for new anticancer therapies. While mTOR inhibitors, such as everolimus, are established therapy in pancreatic NET, results from recent clinical trials indicate that mTOR inhibitors may be also of value in the management of non-pancreatic NET. However, ongoing clinical trials will have to confirm efficacy and elucidate, in which subtypes and in which setting these drugs might be most usefully applied.

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

Buying options

Chapter
USD   29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD   39.99
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD   54.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD   54.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Learn about institutional subscriptions

References

  1. Halfdanarson TR, Rabe KG, Rubin J, Petersen GM (2008) Pancreatic neuroendocrine tumors (PNETs): incidence, prognosis and recent trend toward improved survival. Ann Oncol 19:1727–1733

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  2. Yao JC et al (2008) One hundred years after “carcinoid”: epidemiology of and prognostic factors for neuroendocrine tumors in 35,825 cases in the United States. J Clin Oncol 26(18):3063–3072

    Article  PubMed  Google Scholar 

  3. Ballian N, Loeffler AG, Rajamanickam V, Norstedt PA, Weber SM, Cho CS (2009) A simplified prognostic system for resected pancreatic neuroendocrine neoplasms. HPB 11:422–428 (Oxford)

    Article  PubMed Central  PubMed  Google Scholar 

  4. Knigge U, Hansen CP, Stadil F (2008) Interventional treatment of neuroendocrine liver metastases. Surgeon 6:232–239

    Article  CAS  PubMed  Google Scholar 

  5. Modlin IM, Pavel M, Kidd M, Gustafsson BI (2010) Somatostatin analogues in the treatment of gastroenteropancreatic neuroendocrine (carcinoid) tumours. Aliment Pharmacol Ther 31:169–188

    CAS  PubMed  Google Scholar 

  6. Rinke A et al (2009) Placebo-controlled, double-blind, prospective, randomized study on the effect of octreotide LAR in the control of tumor growth in patients with metastatic neuroendocrine midgut tumors: a report from the PROMID Study Group. J Clin Oncol 27:4656–4663

    Article  CAS  PubMed  Google Scholar 

  7. Moertel CG, Hanley JA, Johnson LA (1980) Streptozocin alone compared with streptozocin plus fluorouracil in the treatment of advanced islet-cell carcinoma. N Engl J Med 303:1189–1194

    Article  CAS  PubMed  Google Scholar 

  8. Moertel CG, Lefkopoulo M, Lipsitz S, Hahn RG, Klaassen D (1992) Streptozocin– doxorubicin, streptozocin–fluorouracil, or chlorozotocin in the treatment of advanced islet-cell carcinoma. N Engl J Med 326:519–523

    Article  CAS  PubMed  Google Scholar 

  9. Heng PN, Saltz LB (1999) Failure to confirm major objective antitumor activity for streptozocin and doxorubicin in the treatment of patients with advanced islet cell carcinoma. Cancer 86:944–948

    Article  Google Scholar 

  10. McCollum AD et al (2004) Lack of efficacy of streptozocin and doxorubicin in patients with advanced pancreatic endocrine tumors. Am J Clin Oncol 27:485–488

    Article  CAS  PubMed  Google Scholar 

  11. Missiaglia E et al (2010) Pancreatic endocrine tumors: expression profiling evidences a role for Akt-mTOR pathway. J Clin Oncol 28:245–255

    Article  CAS  PubMed  Google Scholar 

  12. Moreno A et al (2008) Antitumor activity of rapamycin and octreotide as single agents or in combination in neuroendocrine tumors. Endocr Relat Cancer 15:257–266

    Article  CAS  PubMed  Google Scholar 

  13. Chung DC et al (1997) A novel pancreatic endocrine tumor suppressor gene locus on chromosome 3p with clinical prognostic implications. J Clin Invest 100:404–410

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  14. Moore PS et al (2001) Role of disease-causing genes in sporadic pancreatic endocrine tumors: MEN1 and VHL. Genes Chromosomes Cancer 32:177–181

    Article  CAS  PubMed  Google Scholar 

  15. Schmitt AM et al (2009) VHL inactivation is an important pathway for the development of malignant sporadic pancreatic endocrine tumors. Endocr Relat Cancer 16:1219–1227

    Article  CAS  PubMed  Google Scholar 

  16. Bjornsti MA, Houghton PJ (2004) The TOR pathway: a target for cancer therapy. Nat Rev Cancer 4(5):335–348

    Article  CAS  PubMed  Google Scholar 

  17. Couvelard A et al (2008) Overexpression of the oxygen sensors PHD-1, PHD-2, PHD-3, and FIH is associated with tumor aggressiveness in pancreatic endocrine tumors. Clin Cancer Res 14:6634–6639

    Article  CAS  PubMed  Google Scholar 

  18. Yuchen J et al (2011) DAXX/ATRX, MEN1, and mTOR pathway genes are frequently altered in pancreatic neuroendocrine tumors. Science 331:1199–1203

    Article  Google Scholar 

  19. Woodgett JR (2005) Recent advances in the protein kinase B signaling pathway. Curr Opin Cell Biol 17(2):150–157

    Article  CAS  PubMed  Google Scholar 

  20. Luo J, Manning BD, Cantley LC (2003) Targeting the PI3 K-Akt pathway in human cancer: rationale and promise. Cancer Cell 4(4):257–262

    Article  CAS  PubMed  Google Scholar 

  21. Guertin DA, Sabatini DM (2007) Defining the role of mTOR in cancer. Cancer Cell 12(1):9–22

    Article  CAS  PubMed  Google Scholar 

  22. Huang S, Houghton PJ (2003) Targeting mTOR signaling for cancer therapy. Curr Opin Pharmacol 3(4):371–377

    Article  CAS  PubMed  Google Scholar 

  23. Faivre S, Kroemer G, Raymond E (2006) Current development of mTOR inhibitors as anticancer agents. Nat Rev Drug Discov 5(8):671–688

    Article  CAS  PubMed  Google Scholar 

  24. Houghton PJ, Morton CL, Kolb EA et al (2008) Initial testing (stage 1) of the mTOR inhibitor rapamycin by the pediatric preclinical testing program. Pediatr Blood Cancer 50(4):799–805

    Article  PubMed  Google Scholar 

  25. Motzer RJ, Escudier B, Oudard S et al (2008) Efficacy of everolimus in advanced renal cell carcinoma: a double-blind, randomised, placebo-controlled phase III trial. Lancet 372(9637):449–456

    Article  CAS  PubMed  Google Scholar 

  26. Okamoto I, Doi T, Ohtsu A et al (2010) Phase I clinical and pharmacokinetic study of RAD001 (everolimus) administered daily to Japanese patients with advanced solid tumors. Jpn J Clin Oncol 40(1):17–23

    Article  PubMed Central  PubMed  Google Scholar 

  27. Abraham RT, Gibbons JJ (2007) The mammalian target of rapamycin signaling pathway: twists and turns in the road to cancer therapy. Clin Cancer Res 13(11):3109–3114

    Article  CAS  PubMed  Google Scholar 

  28. Faivre S, Raymond E (2008) Mechanism of action of rapalogues: the antiangiogenic hypothesis. Expert Opin Investig Drugs 17:1619–1621

    Article  CAS  PubMed  Google Scholar 

  29. O’Donnell A et al (2008) Phase I pharmacokinetic and pharmacodynamic study of the oral mammalian target of rapamycin inhibitor everolimus in patients with advanced solid tumors. J Clin Oncol 26:1588–1595

    Article  PubMed  Google Scholar 

  30. Yao JC et al (2010) Daily oral everolimus activity in patients with metastatic pancreatic neuroendocrine tumors after failure of cytotoxic chemotherapy: a phase II trial. J Clin Oncol 28:69–76

    Article  CAS  PubMed  Google Scholar 

  31. Yao JC, Shah MH, Ito T et al (2011) Everolimus for advanced pancreatic neuroendocrine tumors. N Engl J Med 364(6):514–523

    Article  CAS  PubMed  Google Scholar 

  32. Yao JC, Phan AT, Chang DZ, et al (2008) Efficacy of RAD001 (everolimus) and octreotide LAR in advanced low- to intermediate-grade neuroendocrine tumors: results of a phase II study. J Clin Oncol 10(26(26)):4311–4318. doi:10.1200/JCO.2008.16.7858

  33. Pavel ME, Hainsworth JD, Baudin E et al (2011) Everolimus plus octreotide long-acting repeatable for the treatment of advanced neuroendocrine tumours associated with carcinoid syndrome (RADIANT-2): a randomised, placebo-controlled, phase 3 study. Lancet 378(9808):2005–2012

    Article  CAS  PubMed  Google Scholar 

  34. Yao J, Ricci S, Winkler R, Jehl V, Pavel M (2011) Everolimus plus octreotide LAR versus placebo plus octreotide LAR in patients with advanced neuroendocrine tumors (NET): updated safety and efficacy results from RADIANT-2. J Clin Oncol 29(4011)

    Google Scholar 

  35. Yao J, Öberg K, Hainsworth J, Lam D, Rouyrre N, Peeters M, Baudin E, Gross D, Pavel M (2013) Everolimus plus octreotide long-acting release for the treatment of advanced neuroendocrine tumors associated with carcinoid syndrome (RADIANT-2): updated overall survival results; presented at North American neuroendocrine tumor society annual conference. Charleston, South Carolina, Oct 4–5, 2013

    Google Scholar 

  36. Pavel M, Baudin E, Couvelard A et al (2012) ENETS consensus guidelines for the management of patients with liver and other distant metastases from neuroendocrine neoplasms of foregut, midgut, hindgut, and unknown primary. Neuroendocrinology 95(2):157–176

    Article  CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Eric Raymond .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2014 Springer-Verlag France

About this chapter

Cite this chapter

Raymond, E., Pavel, M. (2014). Inhibition of mTOR in Neuroendocrine Neoplasms of the Digestive Tract. In: Raymond, E., Faivre, S., Ruszniewski, P. (eds) Management of Neuroendocrine Tumors of the Pancreas and Digestive Tract. Springer, Paris. https://doi.org/10.1007/978-2-8178-0430-9_8

Download citation

  • DOI: https://doi.org/10.1007/978-2-8178-0430-9_8

  • Published:

  • Publisher Name: Springer, Paris

  • Print ISBN: 978-2-8178-0429-3

  • Online ISBN: 978-2-8178-0430-9

  • eBook Packages: MedicineMedicine (R0)

Publish with us

Policies and ethics