Skip to main content
Top
Published in: Targeted Oncology 2/2012

01-06-2012 | Review

Sunitinib in pancreatic neuroendocrine tumors

Authors: Eric Raymond, Pascal Hammel, Chantal Dreyer, Christian Maatescu, Olivia Hentic, Philippe Ruszniewski, Sandrine Faivre

Published in: Targeted Oncology | Issue 2/2012

Login to get access

Asbtract

Sunitinib is an oral multitarget tyrosine kinase inhibitor with potent antiangiogenic properties. Preclinical data have demonstrated that pancreatic neuroendocrine tumors depend on vascular endothelial growth factor receptors and platelet growth factor receptors-signaling pathways for tumor angiogenesis. Sunitinib has recently been approved for the treatment of patients with advanced, progressive pancreatic neuroendocrine tumors. Sunitinib has demonstrated clinically meaningful improvements in progression-free survival in a double-blinded randomized trial against placebo, setting progression-free survival as a valid endpoint for the evaluation of novel agents in patients with pancreatic neuroendocrine tumors. Although patients who progressed in this phase III trial were allowed to cross-over, a trend toward improvement in overall survival was also observed. In this trial, side effects reported with sunitinib were those previously reported in other tumor types, including hand–foot syndrome, diarrhea, and hypertension. This trial also investigated patient-reported outcome and showed that treatment with sunitinib did not affect quality of life of patient. Interestingly, this trial showed that sunitinib could be combined with somatostatin analogues without affecting the safety profile of either sunitinib or somatostatin analogues. Since the overall survival of patients with well-differentiated neuroendocrine tumors remains sufficiently long, it is worth considering using alternate sequences of targeted therapy (such as everolimus) and chemotherapy to optimize the care of patients with advanced diseases. The optimal sequence for using chemotherapy, everolimus, and sunitinib will remain to be established in clinical trials.
Literature
1.
go back to reference Halfdanarson TR, Rabe KG, Rubin J, Petersen GM (2008) Pancreatic neuroendocrine tumors (PNETs): incidence, prognosis and recent trend toward improved survival. Ann Oncol 19:1727–1733PubMedCrossRef Halfdanarson TR, Rabe KG, Rubin J, Petersen GM (2008) Pancreatic neuroendocrine tumors (PNETs): incidence, prognosis and recent trend toward improved survival. Ann Oncol 19:1727–1733PubMedCrossRef
2.
go back to reference Yao JC, Hassan M, Phan A, Dagohoy C, Leary C, Mares JE 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):3063–3072PubMedCrossRef Yao JC, Hassan M, Phan A, Dagohoy C, Leary C, Mares JE 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):3063–3072PubMedCrossRef
3.
go back to reference Ballian N, Loeffler AG, Rajamanickam V, Norstedt PA, Weber SM, Cho CS (2009) A simplified prognostic system for resected pancreatic neuroendocrine neoplasms. HPB (Oxford) 11:422–428CrossRef Ballian N, Loeffler AG, Rajamanickam V, Norstedt PA, Weber SM, Cho CS (2009) A simplified prognostic system for resected pancreatic neuroendocrine neoplasms. HPB (Oxford) 11:422–428CrossRef
4.
go back to reference Knigge U, Hansen CP, Stadil F (2008) Interventional treatment of neuroendocrine liver metastases. Surgeon 6:232–239PubMedCrossRef Knigge U, Hansen CP, Stadil F (2008) Interventional treatment of neuroendocrine liver metastases. Surgeon 6:232–239PubMedCrossRef
5.
go back to reference Modlin IM, Pavel M, Kidd M, Gustafsson BI (2010) Somatostatin analogues in the treatment of gastroenteropancreatic neuroendocrine (carcinoid) tumours. Aliment Pharmacol Ther 31:169–188PubMed Modlin IM, Pavel M, Kidd M, Gustafsson BI (2010) Somatostatin analogues in the treatment of gastroenteropancreatic neuroendocrine (carcinoid) tumours. Aliment Pharmacol Ther 31:169–188PubMed
6.
go back to reference Rinke A, Müller HH, Schade-Brittinger C, Klose KJ, Barth P, Wied M 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 27:4656–4663PubMedCrossRef Rinke A, Müller HH, Schade-Brittinger C, Klose KJ, Barth P, Wied M 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 27:4656–4663PubMedCrossRef
7.
go back to reference 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 303:1189–1194PubMedCrossRef 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 303:1189–1194PubMedCrossRef
8.
go back to reference 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 326:519–523PubMedCrossRef 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 326:519–523PubMedCrossRef
9.
go back to reference 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 86:944–948CrossRef 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 86:944–948CrossRef
10.
go back to reference McCollum AD, Kulke MH, Ryan DP, Clark JW, Shulman LN, Mayer RJ et al (2004) Lack of efficacy of streptozocin and doxorubicin in patients with advanced pancreatic endocrine tumors. Am J Clin Oncol 27:485–488PubMedCrossRef McCollum AD, Kulke MH, Ryan DP, Clark JW, Shulman LN, Mayer RJ et al (2004) Lack of efficacy of streptozocin and doxorubicin in patients with advanced pancreatic endocrine tumors. Am J Clin Oncol 27:485–488PubMedCrossRef
11.
go back to reference Klöppel G, Anlauf M (2005) Epidemiology, tumour biology and histopathological classification of neuroendocrine tumours of the gastrointestinal tract. Best Pract Res Clin Gastroenterol 19:507–517PubMedCrossRef Klöppel G, Anlauf M (2005) Epidemiology, tumour biology and histopathological classification of neuroendocrine tumours of the gastrointestinal tract. Best Pract Res Clin Gastroenterol 19:507–517PubMedCrossRef
12.
go back to reference Inoue M, Hager JH, Ferrara N, Gerber HP, Hanahan D (2002) VEGF-A has a critical, nonredundant role in angiogenic switching and pancreatic beta cell carcinogenesis. Cancer Cell 1:193–202PubMedCrossRef Inoue M, Hager JH, Ferrara N, Gerber HP, Hanahan D (2002) VEGF-A has a critical, nonredundant role in angiogenic switching and pancreatic beta cell carcinogenesis. Cancer Cell 1:193–202PubMedCrossRef
13.
go back to reference Fjällskog ML, Lejonklou MH, Oberg KE, Eriksson BK, Janson ET (2003) Expression of molecular targets for tyrosine kinase receptor antagonists in malignant endocrine pancreatic tumors. Clin Cancer Res 9:1469–1473PubMed Fjällskog ML, Lejonklou MH, Oberg KE, Eriksson BK, Janson ET (2003) Expression of molecular targets for tyrosine kinase receptor antagonists in malignant endocrine pancreatic tumors. Clin Cancer Res 9:1469–1473PubMed
14.
go back to reference Yao VJ, Sennino B, Davis RB, Christensen J, Hu-Lowe D, Roberts G et al (2006) Combined anti-VEGFR and anti-PDGFR actions of sunitinib on blood vessels in preclinical tumor models. Eur J Cancer 4(12):27–28 Yao VJ, Sennino B, Davis RB, Christensen J, Hu-Lowe D, Roberts G et al (2006) Combined anti-VEGFR and anti-PDGFR actions of sunitinib on blood vessels in preclinical tumor models. Eur J Cancer 4(12):27–28
15.
go back to reference Missiaglia E, Dalai I, Barbi S, Beghelli S, Falconi M, del la Peruta M et al (2010) Pancreatic endocrine tumors: expression profiling evidences a role for AKT-mTOR pathway. J Clin Oncol 28:245–255PubMedCrossRef Missiaglia E, Dalai I, Barbi S, Beghelli S, Falconi M, del la Peruta M et al (2010) Pancreatic endocrine tumors: expression profiling evidences a role for AKT-mTOR pathway. J Clin Oncol 28:245–255PubMedCrossRef
16.
go back to reference Moreno A, Akcakanat A, Munsell MF, Soni A, Yao JC, Meric-Bernstam F (2008) Antitumor activity of rapamycin and octreotide as single agents or in combination in neuroendocrine tumors. Endocr Relat Cancer 15:257–266PubMedCrossRef Moreno A, Akcakanat A, Munsell MF, Soni A, Yao JC, Meric-Bernstam F (2008) Antitumor activity of rapamycin and octreotide as single agents or in combination in neuroendocrine tumors. Endocr Relat Cancer 15:257–266PubMedCrossRef
17.
go back to reference Grothey A, Galanis E (2009) Targeting angiogenesis: progress with anti-VEGF treatment with large molecules. Nat Rev Clin Oncol 6(9):507–18PubMedCrossRef Grothey A, Galanis E (2009) Targeting angiogenesis: progress with anti-VEGF treatment with large molecules. Nat Rev Clin Oncol 6(9):507–18PubMedCrossRef
18.
go back to reference Rodallec M, Vilgrain V, Couvelard A, Rufat P, O'Toole D, Barrau V et al (2006) Endocrine pancreatic tumours and helical CT: contrast enhancement is correlated with microvascular density, histoprognostic factors and survival. Pancreatology 6:77–85PubMedCrossRef Rodallec M, Vilgrain V, Couvelard A, Rufat P, O'Toole D, Barrau V et al (2006) Endocrine pancreatic tumours and helical CT: contrast enhancement is correlated with microvascular density, histoprognostic factors and survival. Pancreatology 6:77–85PubMedCrossRef
19.
go back to reference Couvelard A, O'Toole D, Turley H, Leek R, Sauvanet A, Degott C et al (2005) Microvascular density and hypoxia-inducible factor pathway in pancreatic endocrine tumours: negative correlation of microvascular density and VEGF expression with tumour progression. Br J Cancer 92:94–101PubMedCrossRef Couvelard A, O'Toole D, Turley H, Leek R, Sauvanet A, Degott C et al (2005) Microvascular density and hypoxia-inducible factor pathway in pancreatic endocrine tumours: negative correlation of microvascular density and VEGF expression with tumour progression. Br J Cancer 92:94–101PubMedCrossRef
20.
go back to reference Chung DC, Smith AP, Louis DN, Graeme-Cook F, Warshaw AL, Arnold A (1997) A novel pancreatic endocrine tumor suppressor gene locus on chromosome 3p with clinical prognostic implications. J Clin Invest 100:404–410PubMedCrossRef Chung DC, Smith AP, Louis DN, Graeme-Cook F, Warshaw AL, Arnold A (1997) A novel pancreatic endocrine tumor suppressor gene locus on chromosome 3p with clinical prognostic implications. J Clin Invest 100:404–410PubMedCrossRef
21.
go back to reference Moore PS, Missiaglia E, Antonello D, Zamò A, Zamboni G, Corleto V et al (2001) Role of disease-causing genes in sporadic pancreatic endocrine tumors: MEN1 and VHL. Genes Chromosomes Cancer 32:177–181PubMedCrossRef Moore PS, Missiaglia E, Antonello D, Zamò A, Zamboni G, Corleto V et al (2001) Role of disease-causing genes in sporadic pancreatic endocrine tumors: MEN1 and VHL. Genes Chromosomes Cancer 32:177–181PubMedCrossRef
22.
go back to reference Schmitt AM, Schmid S, Rudolph T, Anlauf M, Prinz C, Klöppel G et al (2009) VHL inactivation is an important pathway for the development of malignant sporadic pancreatic endocrine tumors. Endocr Relat Cancer 16:1219–1227PubMedCrossRef Schmitt AM, Schmid S, Rudolph T, Anlauf M, Prinz C, Klöppel G et al (2009) VHL inactivation is an important pathway for the development of malignant sporadic pancreatic endocrine tumors. Endocr Relat Cancer 16:1219–1227PubMedCrossRef
23.
go back to reference Couvelard A, Deschamps L, Rebours V, Sauvanet A, Gatter K, Pezzella F et al (2008) Overexpression of the oxygen sensors PHD-1, PHD-2, PHD-3, and FIHIs associated with tumor aggressiveness in pancreatic endocrine tumors. Clin Cancer Res 14:6634–6639PubMedCrossRef Couvelard A, Deschamps L, Rebours V, Sauvanet A, Gatter K, Pezzella F et al (2008) Overexpression of the oxygen sensors PHD-1, PHD-2, PHD-3, and FIHIs associated with tumor aggressiveness in pancreatic endocrine tumors. Clin Cancer Res 14:6634–6639PubMedCrossRef
24.
go back to reference Jiao Y, Shi C, Edil BH, de Wilde RF, Klimstra DS, Maitra A et al (2011) DAXX/ATRX, MEN1, and mTOR pathway genes are frequently altered in pancreatic neuroendocrine tumors. Science 331:1199–1203PubMedCrossRef Jiao Y, Shi C, Edil BH, de Wilde RF, Klimstra DS, Maitra A et al (2011) DAXX/ATRX, MEN1, and mTOR pathway genes are frequently altered in pancreatic neuroendocrine tumors. Science 331:1199–1203PubMedCrossRef
25.
go back to reference Hanahan D (1985) Heritable formation of pancreatic β-cells: tumors in transgenic mice harboring recombinant insulin/simian virus 40 oncogenes. Nature 215:115–122CrossRef Hanahan D (1985) Heritable formation of pancreatic β-cells: tumors in transgenic mice harboring recombinant insulin/simian virus 40 oncogenes. Nature 215:115–122CrossRef
26.
go back to reference Lopez T, Hanahan D (2002) Elevated levels of IGF-1 receptor convey invasive and metastatic capability in a mouse model of pancreatic islet tumorigenesis. Cancer Cell 1:339–353PubMedCrossRef Lopez T, Hanahan D (2002) Elevated levels of IGF-1 receptor convey invasive and metastatic capability in a mouse model of pancreatic islet tumorigenesis. Cancer Cell 1:339–353PubMedCrossRef
27.
go back to reference Bergers G, Brekken R, McMahon G, Vu TH, Itoh T, Tamaki K et al (2000) Matrix metalloproteinase-9 triggers the angiogenic switch during carcinogenesis. Nat Cell Biol 2:737–744PubMedCrossRef Bergers G, Brekken R, McMahon G, Vu TH, Itoh T, Tamaki K et al (2000) Matrix metalloproteinase-9 triggers the angiogenic switch during carcinogenesis. Nat Cell Biol 2:737–744PubMedCrossRef
28.
go back to reference Joyce JA, Laakkonen P, Bernasconi M, Bergers G, Ruoslahti E, Hanahan D (2003) Stage-specific vascular markers revealed by phage display in a mouse model of pancreatic islet tumorigenesis. Cancer Cell 4:393–403PubMedCrossRef Joyce JA, Laakkonen P, Bernasconi M, Bergers G, Ruoslahti E, Hanahan D (2003) Stage-specific vascular markers revealed by phage display in a mouse model of pancreatic islet tumorigenesis. Cancer Cell 4:393–403PubMedCrossRef
29.
go back to reference Bergers G, Javaherian K, Lo KM, Folkman J, Hanahan D (1999) Effects of angiogenesis inhibitors on multistage carcinogenesis in mice. Science 284:808–812PubMedCrossRef Bergers G, Javaherian K, Lo KM, Folkman J, Hanahan D (1999) Effects of angiogenesis inhibitors on multistage carcinogenesis in mice. Science 284:808–812PubMedCrossRef
30.
go back to reference Parangi S, O'Reilly M, Christofori G, Holmgren L, Grosfeld J, Folkman J et al (1996) Antiangiogenic therapy of transgenic mice impairs de novo tumor growth. Proc Natl Acad Sci USA 93:2002–2007PubMedCrossRef Parangi S, O'Reilly M, Christofori G, Holmgren L, Grosfeld J, Folkman J et al (1996) Antiangiogenic therapy of transgenic mice impairs de novo tumor growth. Proc Natl Acad Sci USA 93:2002–2007PubMedCrossRef
31.
go back to reference Chiu CW, Nozawa H, Hanahan D (2010) Survival benefit with proapoptotic molecular and pathologic responses from dual targeting of mammalian target of rapamycin and epidermal growth factor receptor in a preclinical model of pancreatic neuroendocrine carcinogenesis. J Clin Oncol 28:4425–4433PubMedCrossRef Chiu CW, Nozawa H, Hanahan D (2010) Survival benefit with proapoptotic molecular and pathologic responses from dual targeting of mammalian target of rapamycin and epidermal growth factor receptor in a preclinical model of pancreatic neuroendocrine carcinogenesis. J Clin Oncol 28:4425–4433PubMedCrossRef
32.
go back to reference Raymond E, Dahan L, Raoul JL, Bang YJ, Borbath I, Lombard-Bohas C et al (2011) Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. N Engl J Med 364:501–513PubMedCrossRef Raymond E, Dahan L, Raoul JL, Bang YJ, Borbath I, Lombard-Bohas C et al (2011) Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. N Engl J Med 364:501–513PubMedCrossRef
33.
go back to reference Yao JC, Shah MH, Ito T, Bohas CL, Wolin EM, Van Cutsem E et al (2011) Everolimus for advanced pancreatic neuroendocrine tumors. N Engl J Med 364:514–523PubMedCrossRef Yao JC, Shah MH, Ito T, Bohas CL, Wolin EM, Van Cutsem E et al (2011) Everolimus for advanced pancreatic neuroendocrine tumors. N Engl J Med 364:514–523PubMedCrossRef
34.
go back to reference Casanovas O, Hicklin DJ, Bergers G, Hanahan D (2005) Drug resistance by evasion of antiangiogenic targeting of VEGF signaling in late-stage pancreatic islet tumors. Cancer Cell 8:299–309PubMedCrossRef Casanovas O, Hicklin DJ, Bergers G, Hanahan D (2005) Drug resistance by evasion of antiangiogenic targeting of VEGF signaling in late-stage pancreatic islet tumors. Cancer Cell 8:299–309PubMedCrossRef
35.
go back to reference Marijon H, Dokmak S, Paradis V, Zappa M, Bieche I, Bouattour M et al (2011) Epithelial-to-mesenchymal transition and acquired resistance to sunitinib in a patient with hepatocellular carcinoma. J Hepatol 54:1073–1078PubMedCrossRef Marijon H, Dokmak S, Paradis V, Zappa M, Bieche I, Bouattour M et al (2011) Epithelial-to-mesenchymal transition and acquired resistance to sunitinib in a patient with hepatocellular carcinoma. J Hepatol 54:1073–1078PubMedCrossRef
36.
go back to reference Faivre S, Demetri G, Sargent W, Raymond E (2007) Molecular basis for sunitinib efficacy and future clinical development. Nat Rev Drug Discov 6:734–745PubMedCrossRef Faivre S, Demetri G, Sargent W, Raymond E (2007) Molecular basis for sunitinib efficacy and future clinical development. Nat Rev Drug Discov 6:734–745PubMedCrossRef
37.
go back to reference Bergers G, Song S, Meyer-Morse N, Bergsland E, Hanahan D (2003) Benefits of targeting both pericytes and endothelial cells in the tumor vasculature with kinase inhibitors. J Clin Invest 111:1287–1295PubMed Bergers G, Song S, Meyer-Morse N, Bergsland E, Hanahan D (2003) Benefits of targeting both pericytes and endothelial cells in the tumor vasculature with kinase inhibitors. J Clin Invest 111:1287–1295PubMed
38.
go back to reference Tuveson D, Hanahan D (2011) Translational medicine: cancer lessons from mice to humans. Nature 471:316–317PubMedCrossRef Tuveson D, Hanahan D (2011) Translational medicine: cancer lessons from mice to humans. Nature 471:316–317PubMedCrossRef
39.
go back to reference Faivre S, Delbaldo C, Vera K, Robert C, Lozahic S, Lassau N et al (2006) Safety, pharmacokinetic, and antitumor activity of SU11248, a novel oral multitarget tyrosine kinase inhibitor, in patients with cancer. J Clin Oncol 24:25–35PubMedCrossRef Faivre S, Delbaldo C, Vera K, Robert C, Lozahic S, Lassau N et al (2006) Safety, pharmacokinetic, and antitumor activity of SU11248, a novel oral multitarget tyrosine kinase inhibitor, in patients with cancer. J Clin Oncol 24:25–35PubMedCrossRef
40.
go back to reference Kulke MH, Lenz HJ, Meropol NJ, Posey J, Ryan DP, Picus J et al (2008) Activity of sunitinib in patients with advanced neuroendocrine tumors. J Clin Oncol 26:3403–3410PubMedCrossRef Kulke MH, Lenz HJ, Meropol NJ, Posey J, Ryan DP, Picus J et al (2008) Activity of sunitinib in patients with advanced neuroendocrine tumors. J Clin Oncol 26:3403–3410PubMedCrossRef
41.
go back to reference Turner NC, Strauss SJ, Sarker D, Gillmore R, Kirkwood A, Hackshaw A et al (2010) Chemotherapy with 5-fluorouracil, cisplatin and streptozocin for neuroendocrine tumours. Br J Cancer 102:1106–1112PubMedCrossRef Turner NC, Strauss SJ, Sarker D, Gillmore R, Kirkwood A, Hackshaw A et al (2010) Chemotherapy with 5-fluorouracil, cisplatin and streptozocin for neuroendocrine tumours. Br J Cancer 102:1106–1112PubMedCrossRef
Metadata
Title
Sunitinib in pancreatic neuroendocrine tumors
Authors
Eric Raymond
Pascal Hammel
Chantal Dreyer
Christian Maatescu
Olivia Hentic
Philippe Ruszniewski
Sandrine Faivre
Publication date
01-06-2012
Publisher
Springer-Verlag
Published in
Targeted Oncology / Issue 2/2012
Print ISSN: 1776-2596
Electronic ISSN: 1776-260X
DOI
https://doi.org/10.1007/s11523-012-0220-2

Other articles of this Issue 2/2012

Targeted Oncology 2/2012 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine