Skip to main content
Log in

Potential Synergies for Combined Targeted Therapy in the Treatment of Neuroendocrine Cancer

  • Leading Article
  • Published:
Drugs Aims and scope Submit manuscript

Abstract

Well differentiated neuroendocrine tumours (WDNET) are a diverse group of cancers that are often advanced at the time of diagnosis and generally do not respond significantly to traditional chemotherapy. A number of intriguing therapeutic targets have emerged, including somatostatin receptors, insulin-like growth factor-1 (IGF-1) and its receptor (IGF-1R), the phosphatidylinositol 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) pathway, and vascular endothelial growth factor receptor. Functional somatostatin receptors and IGF-1R as well as dysregulated mTOR-a key pathway component for both growth factor signalling and protein synthesis — have been identified in human neuroendocrine tumour (NET) cell lines. Somatostatin analogues (SSA) and mTOR inhibitors have exhibited in vitro and in vivo antitumour activity against NET and have shown effects on the IGF-1 pathway in preclinical studies. SSA inhibit PI3K/Akt signalling upstream of mTOR, suggesting that the combination of an SSA and an mTOR inhibitor may have greater efficacy than either as single agents. Recent clinical trial experience has provided some encouraging findings and prompted the design of additional studies of this dual-targeted approach to treating advanced WDNET. Results of ongoing trials of dual-targeted therapy combinations will define future therapies for advanced WDNET.

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

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Institutional subscriptions

Similar content being viewed by others

References

  1. Yao JC, Hassan M, Phan A, et al. One hundred years after “carcinoid”: epidemiology of and prognostic factors for neuroendocrine tumors in 35,825 cases in the United States. J Clin Oncol 2008; 26: 3063–72

    Article  PubMed  Google Scholar 

  2. Modlin IM, Moss SF, Chung DC, et al. Priorities for improving the management of gastroenteropancreatic neuro-endocrine tumors. J Natl Cancer Inst 2008; 100: 1282–9

    Article  PubMed  Google Scholar 

  3. Grozinsky-Glasberg S, Shimon I, Korbonits M, et al. Somatostatin analogues in the control of neuroendocrine tumors: efficacy and mechanisms. Endocr Relat Cancer 2008; 15: 701–20

    Article  PubMed  CAS  Google Scholar 

  4. Öberg K, Jelic S, on behalf of the ESMO Guidelines Working Group. Neuroendocrine gastroenteropancreatic tumors: ESMO Clinical Recommendation for diagnosis, treatment and follow-up. Ann Oncol 2009; 20 Suppl. 4: iv150–3

    Article  Google Scholar 

  5. Florio T. Molecular mechanisms of the antiproliferative activity of somatostatin receptors (sstrs) in neuroendocrine tumors. Front Biosci 2008; 13: 822–40

    Article  PubMed  CAS  Google Scholar 

  6. von Wichert G, Jehle PM, Hoeflich A, et al. Insulin-like growth factor-I is an autocrine regulator of chromagranin A secretion and growth in human neuroendocrine tumor cells. Cancer Res 2000; 60: 4573–81

    Google Scholar 

  7. Albanell J, Dalmases A, Rovira A, et al. mTOR signalling in human cancer. Clin Transl Oncol 2007; 9: 484–93

    Article  PubMed  CAS  Google Scholar 

  8. Nilsson O, Wängberg B, Theodorsson E, et al. Presence of IGF-I in human midgut carcinoid tumours: an autocrine regulator of carcinoid tumour growth? Int J Cancer 1992; 51: 195–203

    Article  PubMed  CAS  Google Scholar 

  9. Hörsch D, Tielke S, Schrader J. Expression and activation of mTOR in neuroendocrine tumors: effects of mTOR inhibition by RAD001 upon growth, cell cycle regulation, and signaling in neuroendocrine cell lines [abstract no. 10570]. J Clin Oncol 2007; 25(18S): 582s

    Google Scholar 

  10. Grozinsky-Glasberg S, Franchi G, Teng M, et al. Octreotide and the mTOR inhibitor RAD001 (everolimus) block proliferation and interact with the Akt-mTOR-p70S6K pathway in a neuro-endocrine tumour cell line. Neuro-endocrinology 2008; 87: 168–81

    CAS  Google Scholar 

  11. Hofland LJ, Lamberts SWJ. The pathophysiological consequences of somatostatin receptor internalization and resistance. Endocr Rev 2003; 24: 28–47

    Article  PubMed  CAS  Google Scholar 

  12. Lamberts SW, van der Lely AJ, de Herder WW, et al. Octreotide. N Engl J Med 1996; 334: 246–54

    Article  PubMed  CAS  Google Scholar 

  13. Öberg K, Kvols L, Caplin M, et al. Consensus report on the use of somatostatin analogs for the management of neuroendocrine tumors of the gastroenteropancreatic system. Ann Oncol 2004; 15: 966–73

    Article  PubMed  Google Scholar 

  14. Modlin IM, Pavel M, Kidd M, et al. Review articles: somatostatin analogues in the treatment of gastroenteropancreatic neuroendocrine (carcinoid) tumours. Aliment Pharmacol Ther 2010; 31: 169–88

    PubMed  CAS  Google Scholar 

  15. Kurosaki M, Saeger W, Abe T, et al. Expression of vascular endothelial growth factor in growth hormone-secreting pituitary adenomas: special reference to the octreotide treatment. Neurol Res 2008; 30: 518–22

    Article  PubMed  CAS  Google Scholar 

  16. Anthony LB, Martin W, Delbeke D, et al. Somatostatin receptor imaging: predictive and prognostic considerations. Digestion 1996; 57: 50–3

    Article  PubMed  CAS  Google Scholar 

  17. Townsend A, Price T, Yeend S, et al. Metastatic carcinoid tumor: changing patterns of care over two decades. J Clin Gastroenterol 2010; 44: 195–9

    Article  PubMed  Google Scholar 

  18. Aparicio T, Ducreux M, Baudin E, et al. Antitumor activity of somatostatin analogues in progressive metastatic neuro-endocrine tumors. Eur J Cancer 2001; 37: 1014–9

    Article  PubMed  CAS  Google Scholar 

  19. Panzuto F, Di Fonzo M, Iannicelli E, et al. Long-term clinical outcome of somatostatin analogues for treatment of progressive, metastatic, well-differentiated entero-pancreatic endocrine carcinoma. Ann Oncol 2006; 17: 461–6

    Article  PubMed  CAS  Google Scholar 

  20. Rinke A, Müller HH, Schade-Brittinger C, et al. 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 2009; 27: 4656–63

    Article  PubMed  CAS  Google Scholar 

  21. Kvols L, Wiedenmann B, Öberg K, et al. Efficacy and safety results from a phase II study of pasireotide (SOM230) in the treatment of patients with metastatic NET refractory or resistant to octreotide LAR [poster presentation C57]. Conference of the European Neuroendocrine Tumour Society; 2010 Mar 11–12; Berlin

  22. Novartis Pharmaceuticals. An open label, multicenter, single arm study of pasireotide LAR in patients with rare tumors of neuroendocrine origin [ClinicalTrials.gov identifier NCT00958841]. US National Institutes of Health, Clinical Trials.gov [online]. Available from URL: http://www.clinicaltrials.gov [Accessed 2009 Jul 21]

  23. Novartis Pharmaceuticals. A multicenter, randomized, blinded efficacy and safety study of pasireotide LAR vs octreotide LAR in patients with metastatic carcinoid tumors whose disease-related symptoms are inadequately controlled by somatostatin analogues [ClinicalTrials.gov identifier NCT00690430]. US National Institutes of Health, ClinicalTrials.gov [online]. Available from URL: http://www.clinicaltrials.gov [Accessed 2009 Jul 21]

  24. LeRoith D, Yakar S. Mechanisms of disease: metabolic effects of growth hormone and insulin-like growth factor 1. Nat Clin Pract Endocrinol Metab 2007; 3: 302–10

    Article  PubMed  CAS  Google Scholar 

  25. Pollak MN, Polychronakos C, Guyda H. Somatostatin analogue SMS 201-995 reduces serum IGF-I levels in patients with neoplasms potentially dependent on IGF-I. Anticancer Res 1989; 9: 889–92

    PubMed  CAS  Google Scholar 

  26. Schmid HA, Stumm M, McSheehy P, et al. Inhibitory effect of pasireotide in a rat pancreatic tumor model (CA20948) alone and in combination with everolimus (RAD001) [poster]. Annual Meeting of the American Association for Cancer Research; 2009 Apr 18–22; Denver (CO)

  27. Friedlander T, Weinberg V, Sharib J, et al. Effect of the somatostatin analog octreotide acetate on circulating IGF-1 and on PSA in patients with castration-resistant prostate cancer (CRPC): results of a phase II study [abstract no. 99 plus poster]. Genitourinary Cancers Symposium; 2010 Mar 5–7; San Francisco (CA)

  28. Pokrajac A, Frystyk J, Flyvbjerg A, et al. Pituitary-independent effect of octreotide on IGF1 generation. Eur J Endocrinol 2009; 160: 543–8

    Article  PubMed  CAS  Google Scholar 

  29. Reidy DL, Hollywood E, Segal M, et al. A phase II clinical trial of MK-0646, an insulin-like growth factor-1 receptor inhibitor (IGF-1R), in patients with metastatic well-differentiated neuroendocrine tumors (NETs) [abstract no. 4163]. J Clin Oncol 2010; 28 Suppl.: 15s

    Article  Google Scholar 

  30. Faivre S, Kroemer G, Raymond E. Current development of mTOR inhibitors as anticancer agents. Nat Rev Drug Discov 2006; 5(8): 671–88

    Article  PubMed  CAS  Google Scholar 

  31. Meric-Bernstam F, Gonzalez-Angulo AM. Targeting the mTOR signaling network for cancer therapy. J Clin Oncol 2009; 27(13): 2278–87

    Article  PubMed  CAS  Google Scholar 

  32. Shida T, Kishimoto T, Furuya M, et al. Expression of an activated mammalian target of rapamycin (mTOR) in gastroenteropancreatic neuroendocrine tumors. Cancer Chemother Pharmacol 2010; 65: 889–93

    Article  PubMed  CAS  Google Scholar 

  33. Samlowski WE, Vogelzang NJ. Emerging drugs for the treatment of metastatic renal cancer. Expert Opin Emerg Drugs 2007; 12: 605–18

    Article  PubMed  CAS  Google Scholar 

  34. Missiaglia E, Dalai I, Barbi S, et al. Pancreatic endocrine tumors: expression profiling evidences a role for Akt-mTOR pathway. J Clin Oncol 2010; 28: 245–55

    Article  PubMed  CAS  Google Scholar 

  35. Ter-Minassian M, Wang Z, Asomaning K, et al. Association of a TSC2 SNP with sporadic neuroendocrine tumor risk [abstract no. 3050]. Proceedings of the Annual Meeting of the American Association for Cancer Research; 2009 Apr 18–22; Denver (CO)

  36. Moreno A, Akcakanat A, Munsell MF, et al. Antitumor activity of rapamycin and octreotide as single agents or in combination in neuroendocrine tumors. Endocr Relat Cancer 2008; 15: 257–66

    Article  PubMed  CAS  Google Scholar 

  37. Neshat MS, Mellinghoff IK, Tran C, et al. Enhanced sensitivity of PTEN-deficient tumors to inhibition of FRAP/mTOR. Proc Natl Acad Sci USA 2001; 98: 10314–9

    Article  PubMed  CAS  Google Scholar 

  38. Artime MC, Blackman S, Ebbinghaus S, et al. PI3K suppression by the mTOR inhibitor ridaforolimus and the AKT inhibitor MK-2206 is associated with enhanced anti-tumor activity and hyperglycemia in preclinical models [abstract no. 3478]. Annual Meeting of the American Association for Cancer Research; 2010 Apr 17–21; Washington (DC)

  39. Zitzmann K, De Toni EN, Brand S, et al. The novel mTOR inhibitor RAD001 (everolimus) induces antiproliferative effects in human pancreatic neuroendocrine tumor cells. Neuroendocrinology 2007; 85: 54–60

    Article  PubMed  CAS  Google Scholar 

  40. Duran I, Kortmansky J, Singh D, et al. A phase II clinical and pharmacodynamic study of temsirolimus in advanced neuroendocrine carcinomas. Br J Cancer 2006; 95: 1148–54

    Article  PubMed  CAS  Google Scholar 

  41. Yao JC, Lombard-Bohas C, Baudin E, et al. Daily oral everolimus activity in patients with metastatic pancreatic neuroendocrine tumors after failure of cytotoxic chemotherapy: a phase II trial. J Clin Oncol 2010; 28: 69–76

    Article  PubMed  CAS  Google Scholar 

  42. Yao JC, Shah MH, Ito T, et al. Everolimus for advanced pancreatic neuroendocrine tumors. N Engl J Med 2011; 364: 514–23

    Article  PubMed  CAS  Google Scholar 

  43. Folkman J, Shing Y. Angiogenesis. J Biol Chem 1992; 267: 10931–4

    PubMed  CAS  Google Scholar 

  44. Holash J, Wiegand SJ, Yancopoulos GD. New model of tumor angiogenesis: dynamic balance between vessel regression and growth mediated by angiopoietin and VEGF. Oncogene 1999; 18: 5356–62

    Article  PubMed  CAS  Google Scholar 

  45. Casanovas O, Hicklin DJ, Bergers G, et al. Drug resistance by evasion of antiangiogenic targeting of VEGF signaling in late-stage pancreatic islet tumors. Cancer Cell 2005; 8: 299–309

    Article  PubMed  CAS  Google Scholar 

  46. Inoue M, Hager JH, Ferrara N, et al. VEGF-A has a critical, nonredundant role in angiogenic switching and pancreatic beta cell carcinogenesis. Cancer Cell 2002; 1: 193–202

    Article  PubMed  CAS  Google Scholar 

  47. La Rosa S, Uccella S, Finzi G, et al. Localization of vascular endothelial growth factor and its receptors in digestive endocrine tumors: correlation with microvessel density and clinicopathologic features. Hum Pathol 2003; 34: 18–27

    Article  PubMed  Google Scholar 

  48. Zhang J, Jia Z, Li Q, et al. Elevated expression of vascular endothelial growth factor correlates with increased angiogenesis and decreased progression-free survival among patients with low-grade neuroendocrine tumors. Cancer 2007; 109: 1478–86

    Article  PubMed  CAS  Google Scholar 

  49. Hansel DE, Rahman A, Hermans J, et al. Liver metastases arising from well-differentiated pancreatic endocrine neoplasms demonstrate increased VEGF-C expression. Mod Pathol 2003; 16: 652–9

    Article  PubMed  Google Scholar 

  50. Pietras K, Hanahan D. A multitargeted, metronomic, and maximum-tolerated dose “chemo-switch” regimen is antiangiogenic, producing objective responses and survival benefit in a mouse model of cancer. J Clin Oncol 2005; 23: 939–52

    Article  PubMed  CAS  Google Scholar 

  51. Yao VJ, Sennino B, Davis RB, et al. Combined anti-VEGFR and anti-PDGFR actions of sunitinib on blood vessels in preclinical tumor models [abstract]. Eur J Cancer 2006; 4 Suppl.: 27–8

    Google Scholar 

  52. Faivre S, Delbaldo C, Vera K, et al. Safety, pharmaco-kinetic, and antitumor activity of SU11248, a novel oral multitarget tyrosine kinase inhibitor, in patients with cancer. J Clin Oncol 2006; 24: 25–35

    Article  PubMed  CAS  Google Scholar 

  53. Kulke MH, Lenz HJ, Meropol NJ, et al. Activity of sunitinib in patients with advanced neuroendocrine tumors. J Clin Oncol 2008; 26: 3403–10

    Article  PubMed  CAS  Google Scholar 

  54. Raymond E, Dahan L, Raoul J-L, et al. Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. N Engl J Med 2011; 364: 501–13

    Article  PubMed  CAS  Google Scholar 

  55. Yao JC, Phan A, Hoff PM, et al. Targeting vascular endothelial growth factor in advanced carcinoid tumor: a random assignment phase II study of depot octreotide with bevacizumab and pegylated interferon alfa-2b. J Clin Oncol 2008; 26: 1316–23

    Article  PubMed  CAS  Google Scholar 

  56. Yao JC, Phan AT, Fogelman D, et al. Randomized run-in study of bevacizumab (B) and everolimus (E) in low- to intermediate-grade neuroendocrine tumors (LGNETs) using perfusion CT as functional biomarker [abstract no. 4002]. J Clin Oncol 2010; 28 Suppl.: 15s

    Article  Google Scholar 

  57. Kwak EL, Clark JW, Chabner B. Targeted agents: the rules of combination. Clin Cancer Res 2007; 13: 5232–7

    Article  PubMed  CAS  Google Scholar 

  58. O'Reilly KE, Rojo F, She QB, et al. mTOR inhibition induces upstream receptor tyrosine kinase signaling and activates Akt. Cancer Res 2006; 66: 1500–8

    Article  PubMed  Google Scholar 

  59. Van Gompel JJ, Chen H. Insulin-like growth factor 1 signaling in human gastrointestinal carcinoid tumor cells. Surgery 2004; 136: 1297–302

    Article  PubMed  Google Scholar 

  60. Charland S, Boucher M-J, Houde M, et al. Somatostatin inhibits Akt phosphorylation and cell cycle entry, but not p42/p44 mitogen-activated protein (MAP) kinase activation in normal and tumoral pancreatic acinar cells. Endocrinology 2001; 142: 121–8

    Article  PubMed  CAS  Google Scholar 

  61. Cerovac V, Monteserin-Garcia J, Rubinfeld H, et al. The somatostatin analogue octreotide confers sensitivity to rapamycin treatment on pituitary tumor cells. Cancer Res 2010; 70: 666–74

    Article  PubMed  CAS  Google Scholar 

  62. Yao JC, Phan AT, Chang DZ, et al. Efficacy of RAD001 (everolimus) and octreotide LAR in advanced low- to intermediate-grade neuroendocrine tumors: results of a phase II study. J Clin Oncol 2008; 26: 4311–8

    Article  PubMed  Google Scholar 

  63. Pavel M, Hainsworth JD, Baudin E, et al. A randomized, double blind, placebo-controlled, multicenter phase III trial of everolimus 1 octreotide lar vs placebo 1 octreotide in patients with advanced neuroendocrine tumors (NET) (RADIANT-2) [abstract]. Ann Oncol 2010; 21: 4

    Article  Google Scholar 

  64. Sathyanarayanan S, Jha S, Klinghoffer R, et al. Combination treatment with the anti-IGF1R antibody MK-0646 and the mTOR inhibitor deforolimus leads to more effective PI3K pathway targeting and anti-tumor activity [poster]. Annual Meeting of the American Association for Cancer Research; 2009 Apr 18–22; Denver (CO)

  65. Merck. A phase I study of ridaforolimus (MK8669) and MK0646 in patients with advanced cancer [ClinicalTrials. gov identifier NCT00730379]. US National Institutes of Health, ClinicalTrials.gov [online]. Available from URL: http://www.clinicaltrials.gov [Accessed 2009 Jul 21]

  66. Hoffmann-La Roche. A multiple ascending dose study of the mTOR inhibitor (RAD001) in combination with R1 507 in patients with advanced solid tumors [ClinicalTrials.gov identifier NCT00985374]. US National Institutes of Health, ClinicalTrials.gov [online]. Available from URL: http://www.clinicaltrials.gov [Accessed 2010 Aug 17]

  67. M.D. Anderson Cancer Center. Phase I study of anti-IGF1R monoclonal antibody, IMC-A12, and mTOR inhibitor, everolimus, in advanced low to intermediate grade neuro-endocrine carcinoma [ClinicalTrials.gov identifier NCT01204476]. US National Institutes of Health, ClinicalTrials.gov [online]. Available from URL: http://www.clinicaltrials.gov [Accessed 2011 Feb 20]

  68. Montagut C, Settleman J. Targeting the RAF-MEK-ERK pathway in cancer therapy. Cancer Lett 2009; 283: 125–34

    Article  PubMed  CAS  Google Scholar 

  69. Hobday TJ, Rubin J, Holen K, et al. MCO44h, a phase II trial of sorafenib in patients (pts) with metastatic neuroendocrine tumors (NET): a phase II consortium (P2C) study, [abstract no. 4504] J Clin Oncol 2007; 25 Suppl. 18: 4504

    Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Ramon Salazar.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Salazar, R., Reidy-Lagunes, D. & Yao, J. Potential Synergies for Combined Targeted Therapy in the Treatment of Neuroendocrine Cancer. Drugs 71, 841–852 (2011). https://doi.org/10.2165/11585500-000000000-00000

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.2165/11585500-000000000-00000

Keywords

Navigation