Skip to main content
Top
Published in: Current Treatment Options in Oncology 3/2014

Open Access 01-09-2014 | Upper Gastrointestinal Cancers (L Rajdev, Section Editor)

Targeting the mTOR Signaling Pathway in Neuroendocrine Tumors

Authors: Jennifer Chan, MD, MPH, Matthew Kulke, MD, MMSc

Published in: Current Treatment Options in Oncology | Issue 3/2014

Login to get access

Opinion statement

Neuroendocrine tumors (NETs) are a heterogeneous group of malignancies characterized by variable but most often indolent biologic behavior. Well-differentiated NETs can be broadly classified as either carcinoid or pancreatic NET. Although they have similar characteristics on routine histologic evaluation, the 2 tumor subtypes have different biology and respond differently to treatment, with most therapeutic agents demonstrating higher response rates in pancreatic NETs compared with carcinoid. Until recently, systemic treatment options for patients with advanced NETs were limited. However, improvements in our understanding of signaling pathways involved in the pathogenesis, growth, and spread of NETs have translated into an expansion of treatment options. Aberrant signaling through the mechanistic pathway of rapamycin (mTOR) pathway has been implicated in neuroendocrine tumorigenesis. Additionally, altered expression of mTOR pathway components has been observed in NETs and has been associated with clinical outcomes. Targeting the mTOR pathway has emerged as an effective treatment strategy in the management of advanced NETs. In a randomized, placebo-controlled study of patients with advanced pancreatic NET, treatment with the mTOR inhibitor everolimus was associated with improved progression-free survival (PFS). Largely based upon these data, everolimus has been approved in the United States and Europe for the treatment of patients with advanced pancreatic NET. The activity of everolimus remains under investigation in patients with carcinoid tumors. In a randomized study of patients with advanced carcinoid tumors associated with carcinoid syndrome, the addition of everolimus to octreotide was associated with improved PFS compared with octreotide. However, the results did not meet the prespecified level of statistical significance based on central review of radiographic imaging. Results from a randomized study examining the efficacy of everolimus in patients with nonfunctional gastrointestinal and lung NETs are awaited. In addition, further investigation is needed to determine whether primary tumor site or other clinical and molecular factors can impact response to mTOR inhibition. Although everolimus can slow tumor progression, significant tumor reduction is rarely obtained. Targeting multiple signaling pathways is a treatment strategy that may provide better tumor control and overcome resistance mechanisms involved with targeting a single pathway. Results of ongoing and future studies will provide important information regarding the added benefit of combining mTOR inhibitors with other targeted agents, such as VEGF pathway inhibitors, and cytotoxic chemotherapy in the treatment of advanced NETs.
Literature
1.•
go back to reference Klimstra DS, Modlin IR, Coppola D, Lloyd RV, Suster S. The pathologic classification of neuroendocrine tumors: a review of nomenclature, grading, and staging systems. Pancreas. 2010;39(6):707–12. This paper provides an excellent overview of the pathologic classification of NETs. Klimstra DS, Modlin IR, Coppola D, Lloyd RV, Suster S. The pathologic classification of neuroendocrine tumors: a review of nomenclature, grading, and staging systems. Pancreas. 2010;39(6):707–12. This paper provides an excellent overview of the pathologic classification of NETs.
2.
go back to reference Kulke MH, Siu LL, Tepper JE, Fisher G, Jaffe D, Haller DG, et al. Future directions in the treatment of neuroendocrine tumors: consensus report of the National Cancer Institute Neuroendocrine Tumor clinical trials planning meeting. J Clin Oncol. 2011;29(7):934–43.PubMedCentralPubMedCrossRef Kulke MH, Siu LL, Tepper JE, Fisher G, Jaffe D, Haller DG, et al. Future directions in the treatment of neuroendocrine tumors: consensus report of the National Cancer Institute Neuroendocrine Tumor clinical trials planning meeting. J Clin Oncol. 2011;29(7):934–43.PubMedCentralPubMedCrossRef
3.
go back to reference Rindi G, Arnold R, Bosman FT, et al. Nomenclature and classification of neuroendocrine neoplasms of the digestive system. In: Carneiro F, Bosman TF, Hruban RH, Theise ND, editors. WHO classification of tumours of the digestive system. Lyon: International Agency for Research on Cancer (IARC) Press; 2010. Rindi G, Arnold R, Bosman FT, et al. Nomenclature and classification of neuroendocrine neoplasms of the digestive system. In: Carneiro F, Bosman TF, Hruban RH, Theise ND, editors. WHO classification of tumours of the digestive system. Lyon: International Agency for Research on Cancer (IARC) Press; 2010.
5.
go back to reference Rinke A, Muller HH, Schade-Brittinger C, Klose KJ, Barth P, Wied M, et al. Placebocontrolled, 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(28):4656–63.PubMedCrossRef Rinke A, Muller HH, Schade-Brittinger C, Klose KJ, Barth P, Wied M, et al. Placebocontrolled, 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(28):4656–63.PubMedCrossRef
6.
go back to reference Caplin M, Ruszniewski P, Pavel M, et al. A randomized, double-blind, placebo-Controlled study of Lanreotide Antiproliferative Response in patients with gastroenteropancreatic NeuroEndocrine Tumors (CLARINET). In: 2013 European Cancer Congress. Amsterdam, Netherlands; 2013. Caplin M, Ruszniewski P, Pavel M, et al. A randomized, double-blind, placebo-Controlled study of Lanreotide Antiproliferative Response in patients with gastroenteropancreatic NeuroEndocrine Tumors (CLARINET). In: 2013 European Cancer Congress. Amsterdam, Netherlands; 2013.
7.
go back to reference Raymond E, Dahan L, Raoul JL, Bang YJ, Borbath I, Lombard-Bohas C, et al. Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. N Engl J Med. 2011;364(6):501–13.PubMedCrossRef Raymond E, Dahan L, Raoul JL, Bang YJ, Borbath I, Lombard-Bohas C, et al. Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. N Engl J Med. 2011;364(6):501–13.PubMedCrossRef
8.••
go back to reference Yao JC, Shah MH, Ito T, Bohas CL, Wolin EM, Van Cutsem E, et al. Everolimus for advanced pancreatic neuroendocrine tumors. N Engl J Med. 2011;364(6):514–23. The results of the RADIANT-3 study are presented in this paper. Everolimus monotherapy was compared to best supportive care alone in patients with advanced pancreatic NET this placebo-controlled phase III trial. Everolimus was associated with a significant prolongation in median PFS (11.0 versus 4.6 months, hazard ratio [HR] for progression 0.35, 95 % confidence interval [CI] 0.27 to 0.45). Yao JC, Shah MH, Ito T, Bohas CL, Wolin EM, Van Cutsem E, et al. Everolimus for advanced pancreatic neuroendocrine tumors. N Engl J Med. 2011;364(6):514–23. The results of the RADIANT-3 study are presented in this paper. Everolimus monotherapy was compared to best supportive care alone in patients with advanced pancreatic NET this placebo-controlled phase III trial. Everolimus was associated with a significant prolongation in median PFS (11.0 versus 4.6 months, hazard ratio [HR] for progression 0.35, 95 % confidence interval [CI] 0.27 to 0.45).
9.
go back to reference Sorbye H, Welin S, Langer SW, Vestermark LW, Holt N, Osterlund P, et al. Predictive and prognostic factors for treatment and survival in 305 patients with advanced gastrointestinal neuroendocrine carcinoma (WHO G3): the NORDIC NEC study. Ann Oncol. 2013;24(1):152–60.PubMedCrossRef Sorbye H, Welin S, Langer SW, Vestermark LW, Holt N, Osterlund P, et al. Predictive and prognostic factors for treatment and survival in 305 patients with advanced gastrointestinal neuroendocrine carcinoma (WHO G3): the NORDIC NEC study. Ann Oncol. 2013;24(1):152–60.PubMedCrossRef
10.
13.
go back to reference Wullschleger S, Loewith R, Hall MN. TOR signaling in growth and metabolism. Cell. 2006;124(3):471–84.PubMedCrossRef Wullschleger S, Loewith R, Hall MN. TOR signaling in growth and metabolism. Cell. 2006;124(3):471–84.PubMedCrossRef
14.•
go back to reference Ma XM, Blenis J. Molecular mechanisms of mTOR-mediated translational control. Nat Rev Mol Cell Biol. 2009;10(5):307–18. This paper presents an excellent review of the mTOR pathway and how its dysregulation contributes to diseases, including cancer. Ma XM, Blenis J. Molecular mechanisms of mTOR-mediated translational control. Nat Rev Mol Cell Biol. 2009;10(5):307–18. This paper presents an excellent review of the mTOR pathway and how its dysregulation contributes to diseases, including cancer.
15.
go back to reference Jacinto E, Loewith R, Schmidt A, Lin S, Ruegg MA, Hall A, et al. Mammalian TOR complex 2 controls the actin cytoskeleton and is rapamycin insensitive. Nat Cell Biol. 2004;6(11):1122–8.PubMedCrossRef Jacinto E, Loewith R, Schmidt A, Lin S, Ruegg MA, Hall A, et al. Mammalian TOR complex 2 controls the actin cytoskeleton and is rapamycin insensitive. Nat Cell Biol. 2004;6(11):1122–8.PubMedCrossRef
16.
go back to reference Loewith R, Jacinto E, Wullschleger S, Lorberg A, Crespo JL, Bonenfant D, et al. Two TOR complexes, only one of which is rapamycin sensitive, have distinct roles in cell growth control. Mol Cell. 2002;10(3):457–68.PubMedCrossRef Loewith R, Jacinto E, Wullschleger S, Lorberg A, Crespo JL, Bonenfant D, et al. Two TOR complexes, only one of which is rapamycin sensitive, have distinct roles in cell growth control. Mol Cell. 2002;10(3):457–68.PubMedCrossRef
17.
go back to reference Sarbassov DD, Guertin DA, Ali SM, Sabatini DM. Phosphorylation and regulation of Akt/PKB by the rictor-mTOR complex. Science. 2005;307(5712):1098–101.PubMedCrossRef Sarbassov DD, Guertin DA, Ali SM, Sabatini DM. Phosphorylation and regulation of Akt/PKB by the rictor-mTOR complex. Science. 2005;307(5712):1098–101.PubMedCrossRef
18.
go back to reference Inoki K, Li Y, Zhu T, Wu J, Guan KL. TSC2 is phosphorylated and inhibited by Akt and suppresses mTOR signalling. Nat Cell Biol. 2002;4(9):648–57.PubMedCrossRef Inoki K, Li Y, Zhu T, Wu J, Guan KL. TSC2 is phosphorylated and inhibited by Akt and suppresses mTOR signalling. Nat Cell Biol. 2002;4(9):648–57.PubMedCrossRef
19.
go back to reference Potter CJ, Pedraza LG, Xu T. Akt regulates growth by directly phosphorylating Tsc2. Nat Cell Biol. 2002;4(9):658–65.PubMedCrossRef Potter CJ, Pedraza LG, Xu T. Akt regulates growth by directly phosphorylating Tsc2. Nat Cell Biol. 2002;4(9):658–65.PubMedCrossRef
20.
go back to reference Manning BD, Tee AR, Logsdon MN, Blenis J, Cantley LC. Identification of the tuberous sclerosis complex-2 tumor suppressor gene product tuberin as a target of the phosphoinositide 3-kinase/akt pathway. Mol Cell. 2002;10(1):151–62.PubMedCrossRef Manning BD, Tee AR, Logsdon MN, Blenis J, Cantley LC. Identification of the tuberous sclerosis complex-2 tumor suppressor gene product tuberin as a target of the phosphoinositide 3-kinase/akt pathway. Mol Cell. 2002;10(1):151–62.PubMedCrossRef
21.
go back to reference Starker LF, Carling T. Molecular genetics of gastroenteropancreatic neuroendocrine tumors. Curr Opin Oncol. 2009;21(1):29–33.PubMedCrossRef Starker LF, Carling T. Molecular genetics of gastroenteropancreatic neuroendocrine tumors. Curr Opin Oncol. 2009;21(1):29–33.PubMedCrossRef
22.
go back to reference Johannessen CM, Reczek EE, James MF, Brems H, Legius E, Cichowski K. The NF1 tumor suppressor critically regulates TSC2 and mTOR. Proc Natl Acad Sci U S A. 2005;102(24):8573–8.PubMedCentralPubMedCrossRef Johannessen CM, Reczek EE, James MF, Brems H, Legius E, Cichowski K. The NF1 tumor suppressor critically regulates TSC2 and mTOR. Proc Natl Acad Sci U S A. 2005;102(24):8573–8.PubMedCentralPubMedCrossRef
24.•
go back to reference Jiao Y, Shi C, Edil BH, de Wilde RF, Klimstra DS, Maitra A, et al. DAXX/ATRX, MEN1, and mTOR pathway genes are frequently altered in pancreatic neuroendocrine tumors. Science. 2011;331(6021):1199–203. Whole exome sequencing was performed in this study to characterize genetic alterations in sporadic pancreatic NET. Somatic mutations in genes involved in the mTOR pathway, including PTEN, TS2, and PIK3CA, were identified in 15 % of cases. Jiao Y, Shi C, Edil BH, de Wilde RF, Klimstra DS, Maitra A, et al. DAXX/ATRX, MEN1, and mTOR pathway genes are frequently altered in pancreatic neuroendocrine tumors. Science. 2011;331(6021):1199–203. Whole exome sequencing was performed in this study to characterize genetic alterations in sporadic pancreatic NET. Somatic mutations in genes involved in the mTOR pathway, including PTEN, TS2, and PIK3CA, were identified in 15 % of cases.
25.
go back to reference Chung DC, Brown SB, Graeme-Cook F, Tillotson LG, Warshaw AL, Jensen RT, et al. Localization of putative tumor suppressor loci by genome-wide allelotyping in human pancreatic endocrine tumors. Cancer Res. 1998;58(16):3706–11.PubMed Chung DC, Brown SB, Graeme-Cook F, Tillotson LG, Warshaw AL, Jensen RT, et al. Localization of putative tumor suppressor loci by genome-wide allelotyping in human pancreatic endocrine tumors. Cancer Res. 1998;58(16):3706–11.PubMed
26.
go back to reference Perren A, Komminoth P, Saremaslani P, Matter C, Feurer S, Lees JA, et al. Mutation and expression analyses reveal differential subcellular compartmentalization of PTEN in endocrine pancreatic tumors compared to normal islet cells. Am J Pathol. 2000;157(4):1097–103.PubMedCentralPubMedCrossRef Perren A, Komminoth P, Saremaslani P, Matter C, Feurer S, Lees JA, et al. Mutation and expression analyses reveal differential subcellular compartmentalization of PTEN in endocrine pancreatic tumors compared to normal islet cells. Am J Pathol. 2000;157(4):1097–103.PubMedCentralPubMedCrossRef
27.•
go back to reference Missiaglia E, Dalai I, Barbi S, Beghelli S, Falconi M, della Peruta M, et al. Pancreatic endocrine tumors: expression profiling evidences a role for AKT-mTOR pathway. J Clin Oncol. 2010;28(2):245–55. In this analysis of gene expression profiles of pancreatic NETs, TSC2 and PTEN, both inhibitors of the mTOR pathway, were found to be downregulated in most of the primary tumors. Furthermore, lower expression was associated with adverse clinical outcome. Missiaglia E, Dalai I, Barbi S, Beghelli S, Falconi M, della Peruta M, et al. Pancreatic endocrine tumors: expression profiling evidences a role for AKT-mTOR pathway. J Clin Oncol. 2010;28(2):245–55. In this analysis of gene expression profiles of pancreatic NETs, TSC2 and PTEN, both inhibitors of the mTOR pathway, were found to be downregulated in most of the primary tumors. Furthermore, lower expression was associated with adverse clinical outcome.
28.
go back to reference Kasajima A, Pavel M, Darb-Esfahani S, Noske A, Stenzinger A, Sasano H, et al. mTOR expression and activity patterns in gastroenteropancreatic neuroendocrine tumours. Endocr Relat Cancer. 2011;18(1):181–92.PubMedCrossRef Kasajima A, Pavel M, Darb-Esfahani S, Noske A, Stenzinger A, Sasano H, et al. mTOR expression and activity patterns in gastroenteropancreatic neuroendocrine tumours. Endocr Relat Cancer. 2011;18(1):181–92.PubMedCrossRef
29.•
go back to reference Qian ZR, Ter-Minassian M, Chan JA, Imamura Y, Hooshmand SM, Kuchiba A, et al. Prognostic significance of MTOR pathway component expression in neuroendocrine tumors. J Clin Oncol. 2013;31(27):3418–25. In this analysis of tumor samples from patients with NETs arising in various sites, primarily small intestine, expression of mTOR or its activated downstream targets were found to be associated with higher proliferation index and adverse clinical outcome. Qian ZR, Ter-Minassian M, Chan JA, Imamura Y, Hooshmand SM, Kuchiba A, et al. Prognostic significance of MTOR pathway component expression in neuroendocrine tumors. J Clin Oncol. 2013;31(27):3418–25. In this analysis of tumor samples from patients with NETs arising in various sites, primarily small intestine, expression of mTOR or its activated downstream targets were found to be associated with higher proliferation index and adverse clinical outcome.
31.
go back to reference Yao JC, Lombard-Bohas C, Baudin E, Kvols LK, Rougier P, Ruszniewski P, 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(1):69–76.PubMedCrossRef Yao JC, Lombard-Bohas C, Baudin E, Kvols LK, Rougier P, Ruszniewski P, 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(1):69–76.PubMedCrossRef
32.
go back to reference Moreno A, Akcakanat A, Munsell MF, Soni A, Yao JC, Meric-Bernstam F. Antitumor activity of rapamycin and octreotide as single agents or in combination in neuroendocrine tumors. Endocr Relat Cancer. 2008;15(1):257–66.PubMedCrossRef Moreno A, Akcakanat A, Munsell MF, Soni A, Yao JC, Meric-Bernstam F. Antitumor activity of rapamycin and octreotide as single agents or in combination in neuroendocrine tumors. Endocr Relat Cancer. 2008;15(1):257–66.PubMedCrossRef
33.
go back to reference O'Reilly KE, Rojo F, She QB, Solit D, Mills GB, Smith D, et al. mTOR inhibition induces upstream receptor tyrosine kinase signaling and activates Akt. Cancer Res. 2006;66(3):1500–8.PubMedCentralPubMedCrossRef O'Reilly KE, Rojo F, She QB, Solit D, Mills GB, Smith D, et al. mTOR inhibition induces upstream receptor tyrosine kinase signaling and activates Akt. Cancer Res. 2006;66(3):1500–8.PubMedCentralPubMedCrossRef
34.
go back to reference van der Veldt AA, Kleijn SA. Advances in pancreatic neuroendocrine tumor treatment. N Engl J Med. 2011;364(19):1873. author reply 1873–5. van der Veldt AA, Kleijn SA. Advances in pancreatic neuroendocrine tumor treatment. N Engl J Med. 2011;364(19):1873. author reply 1873–5.
35.
go back to reference Kulke MH, Bergsland EK, Yao JC. Glycemic control in patients with insulinoma treated with everolimus. N Engl J Med. 2009;360(2):195–7.PubMedCrossRef Kulke MH, Bergsland EK, Yao JC. Glycemic control in patients with insulinoma treated with everolimus. N Engl J Med. 2009;360(2):195–7.PubMedCrossRef
36.
go back to reference Fiebrich HB, Siemerink EJ, Brouwers AH, Links TP, Remkes WS, Hospers GA, et al. Everolimus induces rapid plasma glucose normalization in insulinoma patients by effects on tumor as well as normal tissues. Oncologist. 2011;16(6):783–7.PubMedCentralPubMedCrossRef Fiebrich HB, Siemerink EJ, Brouwers AH, Links TP, Remkes WS, Hospers GA, et al. Everolimus induces rapid plasma glucose normalization in insulinoma patients by effects on tumor as well as normal tissues. Oncologist. 2011;16(6):783–7.PubMedCentralPubMedCrossRef
37.
go back to reference Yao JC, Phan AT, Chang DZ, Wolff RA, Hess K, Gupta S, 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(26):4311–8.PubMedCentralPubMedCrossRef Yao JC, Phan AT, Chang DZ, Wolff RA, Hess K, Gupta S, 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(26):4311–8.PubMedCentralPubMedCrossRef
38.••
go back to reference Pavel ME, Hainsworth JD, Baudin E, Peeters M, Horsch D, Winkler RE, et al. 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. 2011;378(9808):2005–12. The results of the RADIANT-2 study are presented in this paper. In this study, patients with advanced NETs associated with carcinoid syndrome and radiographic disease progression in the preceding 12 months were randomly assigned to octreotide LAR with either everolimus or placebo. The median PFS as assessed by central radiology review was 16.4 months for patients receiving everolimus and octreotide LAR compared with 11.3 months for patients receiving placebo and octreotide LAR (HR 0.77, 95 % CI 0.59-1.00; p = 0.026). These results did not meet the pre-specified level of statistical significance. However, based on local investigator radiology assessment, combined therapy was associated with a median PFS duration of 12.0 months as compared to 8.6 months with placebo (HR 0.78, 95 % CI 0.62-0.98; p = 0.018). Pavel ME, Hainsworth JD, Baudin E, Peeters M, Horsch D, Winkler RE, et al. 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. 2011;378(9808):2005–12. The results of the RADIANT-2 study are presented in this paper. In this study, patients with advanced NETs associated with carcinoid syndrome and radiographic disease progression in the preceding 12 months were randomly assigned to octreotide LAR with either everolimus or placebo. The median PFS as assessed by central radiology review was 16.4 months for patients receiving everolimus and octreotide LAR compared with 11.3 months for patients receiving placebo and octreotide LAR (HR 0.77, 95 % CI 0.59-1.00; p = 0.026). These results did not meet the pre-specified level of statistical significance. However, based on local investigator radiology assessment, combined therapy was associated with a median PFS duration of 12.0 months as compared to 8.6 months with placebo (HR 0.78, 95 % CI 0.62-0.98; p = 0.018).
39.
go back to reference Yao JC, Hainsworth JD, Wolin EM, Pavel ME, Baudin E, Gross D, et al. Multivariate analysis including biomarkers in the phase III RADIANT-2 study of octreotide LAR plus everolimus (E+O) or placebo (P+O) among patients with advanced neuroendocrine tumors (NET). J Clin Oncol. 2012;30(suppl 4):abstr 157. Yao JC, Hainsworth JD, Wolin EM, Pavel ME, Baudin E, Gross D, et al. Multivariate analysis including biomarkers in the phase III RADIANT-2 study of octreotide LAR plus everolimus (E+O) or placebo (P+O) among patients with advanced neuroendocrine tumors (NET). J Clin Oncol. 2012;30(suppl 4):abstr 157.
40.
go back to reference Castellano D, Bajetta E, Panneerselvam A, Saletan S, Kocha W, O'Dorisio T, et al. Everolimus plus octreotide long-acting repeatable in patients with colorectal neuroendocrine tumors: a subgroup analysis of the phase III RADIANT-2 study. Oncologist. 2013;18(1):46–53.PubMedCentralPubMedCrossRef Castellano D, Bajetta E, Panneerselvam A, Saletan S, Kocha W, O'Dorisio T, et al. Everolimus plus octreotide long-acting repeatable in patients with colorectal neuroendocrine tumors: a subgroup analysis of the phase III RADIANT-2 study. Oncologist. 2013;18(1):46–53.PubMedCentralPubMedCrossRef
41.
go back to reference Duran I, Kortmansky J, Singh D, Hirte H, Kocha W, Goss G, et al. A phase II clinical and pharmacodynamic study of temsirolimus in advanced neuroendocrine carcinomas. Br J Cancer. 2006;95(9):1148–54.PubMedCentralPubMedCrossRef Duran I, Kortmansky J, Singh D, Hirte H, Kocha W, Goss G, et al. A phase II clinical and pharmacodynamic study of temsirolimus in advanced neuroendocrine carcinomas. Br J Cancer. 2006;95(9):1148–54.PubMedCentralPubMedCrossRef
42.
go back to reference Shi Y, Yan H, Frost P, Gera J, Lichtenstein A. Mammalian target of rapamycin inhibitors activate the AKT kinase in multiple myeloma cells by up-regulating the insulin-like growth factor receptor/insulin receptor substrate-1/phosphatidylinositol 3-kinase cascade. Mol Cancer Ther. 2005;4(10):1533–40.PubMedCrossRef Shi Y, Yan H, Frost P, Gera J, Lichtenstein A. Mammalian target of rapamycin inhibitors activate the AKT kinase in multiple myeloma cells by up-regulating the insulin-like growth factor receptor/insulin receptor substrate-1/phosphatidylinositol 3-kinase cascade. Mol Cancer Ther. 2005;4(10):1533–40.PubMedCrossRef
43.
go back to reference Chandarlapaty S, Sawai A, Scaltriti M, Rodrik-Outmezguine V, Grbovic-Huezo O, Serra V, et al. AKT inhibition relieves feedback suppression of receptor tyrosine kinase expression and activity. Cancer Cell. 2011;19(1):58–71.PubMedCentralPubMedCrossRef Chandarlapaty S, Sawai A, Scaltriti M, Rodrik-Outmezguine V, Grbovic-Huezo O, Serra V, et al. AKT inhibition relieves feedback suppression of receptor tyrosine kinase expression and activity. Cancer Cell. 2011;19(1):58–71.PubMedCentralPubMedCrossRef
44.
go back to reference Bousquet C, Lasfargues C, Chalabi M, Billah SM, Susini C, Vezzosi D, et al. Clinical review: current scientific rationale for the use of somatostatin analogs and mTOR inhibitors in neuroendocrine tumor therapy. J Clin Endocrinol Metab. 2012;97(3):727–37.PubMedCrossRef Bousquet C, Lasfargues C, Chalabi M, Billah SM, Susini C, Vezzosi D, et al. Clinical review: current scientific rationale for the use of somatostatin analogs and mTOR inhibitors in neuroendocrine tumor therapy. J Clin Endocrinol Metab. 2012;97(3):727–37.PubMedCrossRef
45.
go back to reference Schmid HA. Pasireotide (SOM230): development, mechanism of action and potential applications. Mol Cell Endocrinol. 2008;286(1–2):69–74.PubMedCrossRef Schmid HA. Pasireotide (SOM230): development, mechanism of action and potential applications. Mol Cell Endocrinol. 2008;286(1–2):69–74.PubMedCrossRef
46.
go back to reference Chan J, Ryan D, Fuchs C, Zhu A, Abrams T, Wolpin B, et al. Phase I study of pasireotide (SOM230) in combination with everolimus (RAD001) in patients (pts) with advanced neuroendocrine tumors (NET) Gastrointestinal Cancers Symposium Annual Meeting Proceedings; 2010: p. Abstract 238. Chan J, Ryan D, Fuchs C, Zhu A, Abrams T, Wolpin B, et al. Phase I study of pasireotide (SOM230) in combination with everolimus (RAD001) in patients (pts) with advanced neuroendocrine tumors (NET) Gastrointestinal Cancers Symposium Annual Meeting Proceedings; 2010: p. Abstract 238.
47.
go back to reference Terris B, Scoazec JY, Rubbia L, Bregeaud L, Pepper MS, Ruszniewski P, et al. Expression of vascular endothelial growth factor in digestive neuroendocrine tumours. Histopathology. 1998;32(2):133–8.PubMedCrossRef Terris B, Scoazec JY, Rubbia L, Bregeaud L, Pepper MS, Ruszniewski P, et al. Expression of vascular endothelial growth factor in digestive neuroendocrine tumours. Histopathology. 1998;32(2):133–8.PubMedCrossRef
48.
go back to reference Zhang J, Jia Z, Li Q, Wang L, Rashid A, Zhu Z, 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(8):1478–86.PubMedCrossRef Zhang J, Jia Z, Li Q, Wang L, Rashid A, Zhu Z, 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(8):1478–86.PubMedCrossRef
49.
go back to reference Bowen KA, Silva SR, Johnson JN, Doan HQ, Jackson LN, Gulhati P, et al. An analysis of trends and growth factor receptor expression of GI carcinoid tumors. J Gastrointest Surg. 2009;13(10):1773–80.PubMedCentralPubMedCrossRef Bowen KA, Silva SR, Johnson JN, Doan HQ, Jackson LN, Gulhati P, et al. An analysis of trends and growth factor receptor expression of GI carcinoid tumors. J Gastrointest Surg. 2009;13(10):1773–80.PubMedCentralPubMedCrossRef
50.
go back to reference Silva SR, Bowen KA, Rychahou PG, Jackson LN, Weiss HL, Lee EY, et al. VEGFR-2 expression in carcinoid cancer cells and its role in tumor growth and metastasis. Int J Cancer. 2011;128(5):1045–56.PubMedCentralPubMedCrossRef Silva SR, Bowen KA, Rychahou PG, Jackson LN, Weiss HL, Lee EY, et al. VEGFR-2 expression in carcinoid cancer cells and its role in tumor growth and metastasis. Int J Cancer. 2011;128(5):1045–56.PubMedCentralPubMedCrossRef
51.
go back to reference Fjallskog ML, Hessman O, Eriksson B, Janson ET. Upregulated expression of PDGF receptor beta in endocrine pancreatic tumors and metastases compared to normal endocrine pancreas. Acta Oncol. 2007;46(6):741–6.PubMedCrossRef Fjallskog ML, Hessman O, Eriksson B, Janson ET. Upregulated expression of PDGF receptor beta in endocrine pancreatic tumors and metastases compared to normal endocrine pancreas. Acta Oncol. 2007;46(6):741–6.PubMedCrossRef
52.
go back to reference Fjallskog ML, Lejonklou MH, Oberg KE, Eriksson BK, Janson ET. Expression of molecular targets for tyrosine kinase receptor antagonists in malignant endocrine pancreatic tumors. Clin Cancer Res. 2003;9(4):1469–73.PubMed Fjallskog ML, Lejonklou MH, Oberg KE, Eriksson BK, Janson ET. Expression of molecular targets for tyrosine kinase receptor antagonists in malignant endocrine pancreatic tumors. Clin Cancer Res. 2003;9(4):1469–73.PubMed
53.
go back to reference Hansel DE, Rahman A, Hermans J, de Krijger RR, Ashfaq R, Yeo CJ, et al. Liver metastases arising from well-differentiated pancreatic endocrine neoplasms demonstrate increased VEGF-C expression. Mod Pathol. 2003;16(7):652–9.PubMedCrossRef Hansel DE, Rahman A, Hermans J, de Krijger RR, Ashfaq R, Yeo CJ, et al. Liver metastases arising from well-differentiated pancreatic endocrine neoplasms demonstrate increased VEGF-C expression. Mod Pathol. 2003;16(7):652–9.PubMedCrossRef
54.
go back to reference Hobday TJ, Rubin J, Holen K, Picus J, Donehower R, Marschke R, et al. MC044h, a phase II trial of sorafenib in patients (pts) with metastatic neuroendocrine tumors (NET): a Phase II Consortium (P2C) study. J Clin Oncol, 2007 ASCO Annual Meeting Proceedings Part I, 2007;25(18S):p. Abstract 4504. Hobday TJ, Rubin J, Holen K, Picus J, Donehower R, Marschke R, et al. MC044h, a phase II trial of sorafenib in patients (pts) with metastatic neuroendocrine tumors (NET): a Phase II Consortium (P2C) study. J Clin Oncol, 2007 ASCO Annual Meeting Proceedings Part I, 2007;25(18S):p. Abstract 4504.
55.
go back to reference Phan A, Yao J, Fogelman D, Hess K, Ng C, Bullock S, et al. A prospective, multi-institutional phase II study of GW786034 (pazopanib) and depot octreotide (sandostatin LAR) in advanced low-grade neuroendocrine carcinoma (LGNEC). J Clin Oncol, 2010 ASCO Annual Meeting Proceedings 2010;28(15S):p. Abstract 4044. Phan A, Yao J, Fogelman D, Hess K, Ng C, Bullock S, et al. A prospective, multi-institutional phase II study of GW786034 (pazopanib) and depot octreotide (sandostatin LAR) in advanced low-grade neuroendocrine carcinoma (LGNEC). J Clin Oncol, 2010 ASCO Annual Meeting Proceedings 2010;28(15S):p. Abstract 4044.
56.
go back to reference Yao JC, Phan A, Hoff PM, Chen HX, Charnsangavej C, Yeung SC, 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 alpha-2b. J Clin Oncol. 2008;26(8):1316–23.PubMedCrossRef Yao JC, Phan A, Hoff PM, Chen HX, Charnsangavej C, Yeung SC, 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 alpha-2b. J Clin Oncol. 2008;26(8):1316–23.PubMedCrossRef
57.
go back to reference Chan J, Mayer R, Jackson N, Malinowski P, Regan E, Kulke M, Phase I study of sorafenib in combination with everolimus (RAD001) in patients with advanced neuroendocrine tumors (NET). J Clin Oncol. 2010 (suppl; abstr e14597) 2010. Chan J, Mayer R, Jackson N, Malinowski P, Regan E, Kulke M, Phase I study of sorafenib in combination with everolimus (RAD001) in patients with advanced neuroendocrine tumors (NET). J Clin Oncol. 2010 (suppl; abstr e14597) 2010.
58.
go back to reference Yao J, Phan A, Fogleman D, Ng C, Jacobs C, Dagohoy C, 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. J Clin Oncol, 2010 ASCO Annual Meeting Proceedings. 2010. 28:15s:(suppl; abstr 4002). Yao J, Phan A, Fogleman D, Ng C, Jacobs C, Dagohoy C, 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. J Clin Oncol, 2010 ASCO Annual Meeting Proceedings. 2010. 28:15s:(suppl; abstr 4002).
59.
go back to reference Hobday T, Qin R, Reidy D, Moore M, Strosberg J, Kaubisch A, et al. Multicenter phase II trial of temsirolimus (TEM) and bevacizumab (BEV) in pancreatic neuroendocrine tumor (PNET). J Clin Oncol , 2012 ASCO Annual Meeting Proceedings. 2012;30: p. (suppl 4; abstr 260). Hobday T, Qin R, Reidy D, Moore M, Strosberg J, Kaubisch A, et al. Multicenter phase II trial of temsirolimus (TEM) and bevacizumab (BEV) in pancreatic neuroendocrine tumor (PNET). J Clin Oncol , 2012 ASCO Annual Meeting Proceedings. 2012;30: p. (suppl 4; abstr 260).
60.
go back to reference Moertel C, Lefkopoulo M, Lipsitz S, Hahn R, Klaassen D. Streptozocin-doxorubicin, stretpozocin-fluorouracil, or chlorozotocin in the treatment of advanced islet-cell carcinoma. N Engl J Med. 1992;326:519–23.PubMedCrossRef Moertel C, Lefkopoulo M, Lipsitz S, Hahn R, Klaassen D. Streptozocin-doxorubicin, stretpozocin-fluorouracil, or chlorozotocin in the treatment of advanced islet-cell carcinoma. N Engl J Med. 1992;326:519–23.PubMedCrossRef
61.
go back to reference Kouvaraki M, Ajani J, Hoff P, Wolff R, Evans D, Lozano R, et al. Fluorouracil, doxorubicin, and streptozocin in the treatment of patients with locally advanced and metastatic pancreatic endocrine carcinomas. J Clin Oncol. 2004;22:4762–71.PubMedCrossRef Kouvaraki M, Ajani J, Hoff P, Wolff R, Evans D, Lozano R, et al. Fluorouracil, doxorubicin, and streptozocin in the treatment of patients with locally advanced and metastatic pancreatic endocrine carcinomas. J Clin Oncol. 2004;22:4762–71.PubMedCrossRef
62.
go back to reference Ekeblad S, Sundin A, Janson ET, Welin S, Granberg D, Kindmark H, et al. Temozolomide as monotherapy is effective in treatment of advanced malignant neuroendocrine tumors. Clin Cancer Res. 2007;13(10):2986–91.PubMedCrossRef Ekeblad S, Sundin A, Janson ET, Welin S, Granberg D, Kindmark H, et al. Temozolomide as monotherapy is effective in treatment of advanced malignant neuroendocrine tumors. Clin Cancer Res. 2007;13(10):2986–91.PubMedCrossRef
63.
go back to reference Kulke MH, Hornick JL, Frauenhoffer C, Hooshmand S, Ryan DP, Enzinger PC, et al. O6-methylguanine DNA methyltransferase deficiency and response to temozolomide-based therapy in patients with neuroendocrine tumors. Clin Cancer Res. 2009;15(1):338–45.PubMedCentralPubMedCrossRef Kulke MH, Hornick JL, Frauenhoffer C, Hooshmand S, Ryan DP, Enzinger PC, et al. O6-methylguanine DNA methyltransferase deficiency and response to temozolomide-based therapy in patients with neuroendocrine tumors. Clin Cancer Res. 2009;15(1):338–45.PubMedCentralPubMedCrossRef
64.
go back to reference Strosberg JR, Fine RL, Choi J, Nasir A, Coppola D, Chen DT, et al. First-line chemotherapy with capecitabine and temozolomide in patients with metastatic pancreatic endocrine carcinomas. Cancer. 2011;117(2):268–75.PubMedCrossRef Strosberg JR, Fine RL, Choi J, Nasir A, Coppola D, Chen DT, et al. First-line chemotherapy with capecitabine and temozolomide in patients with metastatic pancreatic endocrine carcinomas. Cancer. 2011;117(2):268–75.PubMedCrossRef
65.
go back to reference Chan JA, Blaszkowsky L, Stuart K, Zhu AX, Allen J, Wadlow R, et al. A prospective, phase 1/2 study of everolimus and temozolomide in patients with advanced pancreatic neuroendocrine tumor. Cancer. 2013;119(17):3212–8.PubMedCrossRef Chan JA, Blaszkowsky L, Stuart K, Zhu AX, Allen J, Wadlow R, et al. A prospective, phase 1/2 study of everolimus and temozolomide in patients with advanced pancreatic neuroendocrine tumor. Cancer. 2013;119(17):3212–8.PubMedCrossRef
66.
go back to reference Yao JC, Phan AT, Jehl V, Shah G, Meric-Bernstam F. Everolimus in advanced pancreatic neuroendocrine tumors: the clinical experience. Cancer Res. 2013;73(5):1449–53.PubMedCrossRef Yao JC, Phan AT, Jehl V, Shah G, Meric-Bernstam F. Everolimus in advanced pancreatic neuroendocrine tumors: the clinical experience. Cancer Res. 2013;73(5):1449–53.PubMedCrossRef
Metadata
Title
Targeting the mTOR Signaling Pathway in Neuroendocrine Tumors
Authors
Jennifer Chan, MD, MPH
Matthew Kulke, MD, MMSc
Publication date
01-09-2014
Publisher
Springer US
Published in
Current Treatment Options in Oncology / Issue 3/2014
Print ISSN: 1527-2729
Electronic ISSN: 1534-6277
DOI
https://doi.org/10.1007/s11864-014-0294-4

Other articles of this Issue 3/2014

Current Treatment Options in Oncology 3/2014 Go to the issue

Sarcoma (SH Okuno, Section Editor)

Surgical Treatment of Sarcomas of the Spine

Upper Gastrointestinal Cancers (L Rajdev, Section Editor)

HER2 Directed Therapy for Gastric/Esophageal Cancers

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