Skip to main content
Top
Published in: Cancer and Metastasis Reviews 3-4/2013

01-12-2013 | NON-THEMATIC REVIEW

The targeted therapy revolution in neuroendocrine tumors: in search of biomarkers for patient selection and response evaluation

Authors: Sara De Dosso, Enrique Grande, Jorge Barriuso, Daniel Castellano, Josep Tabernero, Jaume Capdevila

Published in: Cancer and Metastasis Reviews | Issue 3-4/2013

Login to get access

Abstract

The molecular events of tumorigenesis in neuroendocrine tumors are poorly understood. Understanding of the molecular alterations will lead to the identification of molecular markers, providing new targets for therapeutics. The purpose of this review was to critically analyze the genetic abnormalities in neuroendocrine tumors, with the aim of identifying biomarkers that indicate a response to agents developed against these targets and to serve as an understanding for the combinations of different active compounds. Human epidermal growth factor receptor 1/2 (EGFR and HER2), vascular endothelial growth factor receptors, hepatocyte growth factor receptor (c-Met), platelet-derived growth factor receptor, insulin-like growth factor, phosphatidylinositol 3-kinase–Akt–mammalian target of rapamycin pathway, and heat shock proteins are all interesting candidate biomarkers with involvement in carcinogenesis and tumor evolution of several neoplasms, including neuroendocrine tumors. Some of them have already been evaluated both as targets and also as biomarkers in clinical trials conducted in advanced neuroendocrine tumor settings, and others should encourage further investigations into innovative therapeutic opportunities.
Literature
1.
go back to reference Yao, J. C., Hassan, M., Phan, A., Dagohoy, C., Leary, C., Mares, J. E., Abdalla, E. K., et al. (2008). One hundred years after “carcinoid”: epidemiology of and prognostic factors for neuroendocrine tumors in 35,825 cases in the United States. Journal of Clinical Oncology, 26, 3063–3072.PubMedCrossRef Yao, J. C., Hassan, M., Phan, A., Dagohoy, C., Leary, C., Mares, J. E., Abdalla, E. K., et al. (2008). One hundred years after “carcinoid”: epidemiology of and prognostic factors for neuroendocrine tumors in 35,825 cases in the United States. Journal of Clinical Oncology, 26, 3063–3072.PubMedCrossRef
2.
go back to reference Modlin, I. M., Lye, K. D., & Kidd, M. (2003). A 5-decade analysis of 13715 carcinoid tumors. Cancer, 97, 934–959.PubMedCrossRef Modlin, I. M., Lye, K. D., & Kidd, M. (2003). A 5-decade analysis of 13715 carcinoid tumors. Cancer, 97, 934–959.PubMedCrossRef
3.
go back to reference DeLellis, R. A. (2001). The neuroendocrine system and its tumors. American Journal of Clinical Pathology, 115, S5.PubMed DeLellis, R. A. (2001). The neuroendocrine system and its tumors. American Journal of Clinical Pathology, 115, S5.PubMed
4.
go back to reference Grande, E., Capdevila, J., Barriuso, J., Anton-Aparicio, J., & Castellano, D. (2012). Gastroenteropancreatic neuroendocrine tumor cancer stem cells: do they exist? Cancer Metastasis Reviews, 31(1-2), 47–53.PubMedCrossRef Grande, E., Capdevila, J., Barriuso, J., Anton-Aparicio, J., & Castellano, D. (2012). Gastroenteropancreatic neuroendocrine tumor cancer stem cells: do they exist? Cancer Metastasis Reviews, 31(1-2), 47–53.PubMedCrossRef
5.
go back to reference Castellano, D., Salazar, R., & Raymond, E. (2011). Future perspectives on neuroendocrine tumors. Cancer Metastasis Reviews, 30(Suppl 1), 35–40.PubMed Castellano, D., Salazar, R., & Raymond, E. (2011). Future perspectives on neuroendocrine tumors. Cancer Metastasis Reviews, 30(Suppl 1), 35–40.PubMed
6.
go back to reference Grozinsky-Glasberg, S., Shimon, I., & Rubinfeld, H. (2012). The role of cell lines in the study of neuroendocrine tumors. Neuroendocrinology, 96, 173–187.PubMedCrossRef Grozinsky-Glasberg, S., Shimon, I., & Rubinfeld, H. (2012). The role of cell lines in the study of neuroendocrine tumors. Neuroendocrinology, 96, 173–187.PubMedCrossRef
7.
go back to reference Arany, I., Rady, P., Evers, B. M., Tyring, S. K., & Townsend, C. M. (1994). Analysis of multiple molecular changes in human endocrine tumors. Surgical Oncology, 3(3), 153–159.PubMedCrossRef Arany, I., Rady, P., Evers, B. M., Tyring, S. K., & Townsend, C. M. (1994). Analysis of multiple molecular changes in human endocrine tumors. Surgical Oncology, 3(3), 153–159.PubMedCrossRef
8.
go back to reference Zitzmann, K., De Toni, E. N., Brand, S., Göke, B., Meinecke, J., Spöttl, G., et al. (2007). The novel mTOR inhibitor RAD001 (everolimus) induces antiproliferative effects in human pancreatic neuroendocrine tumor cells. Neuroendocrinology, 85(1), 54–60.PubMedCrossRef Zitzmann, K., De Toni, E. N., Brand, S., Göke, B., Meinecke, J., Spöttl, G., et al. (2007). The novel mTOR inhibitor RAD001 (everolimus) induces antiproliferative effects in human pancreatic neuroendocrine tumor cells. Neuroendocrinology, 85(1), 54–60.PubMedCrossRef
9.
go back to reference Pitt, S. C., Chen, H., & Kunnimalaiyaan, M. (2009). Inhibition of phosphatidylinositol 3-kinase/Akt signaling suppresses tumor cell proliferation and neuroendocrine marker expression in GI carcinoid tumors. Annals of Surgical Oncology, 16(10), 2936–2942.PubMedCrossRef Pitt, S. C., Chen, H., & Kunnimalaiyaan, M. (2009). Inhibition of phosphatidylinositol 3-kinase/Akt signaling suppresses tumor cell proliferation and neuroendocrine marker expression in GI carcinoid tumors. Annals of Surgical Oncology, 16(10), 2936–2942.PubMedCrossRef
10.
go back to reference Kölby, L., Bernhardt, P., Ahlman, H., Wängberg, B., Johanson, V., Wigander, A., et al. (2001). A transplantable human carcinoid as model for somatostatin receptor-mediated and amine transporter-mediated radionuclide uptake. American Journal of Pathology, 158(2), 745–755.PubMedCrossRef Kölby, L., Bernhardt, P., Ahlman, H., Wängberg, B., Johanson, V., Wigander, A., et al. (2001). A transplantable human carcinoid as model for somatostatin receptor-mediated and amine transporter-mediated radionuclide uptake. American Journal of Pathology, 158(2), 745–755.PubMedCrossRef
11.
go back to reference Zitzmann, K., Rüden, J., Brand, S., Göke, B., Lichtl, J., Spöttl, G., & Auernhammer, C. J. (2010). Compensatory activation of Akt in response to mTOR and Raf inhibitors—a rationale for dual-targeted therapy approaches in neuroendocrine tumor disease. Cancer Letter, 295(1), 100–109.CrossRef Zitzmann, K., Rüden, J., Brand, S., Göke, B., Lichtl, J., Spöttl, G., & Auernhammer, C. J. (2010). Compensatory activation of Akt in response to mTOR and Raf inhibitors—a rationale for dual-targeted therapy approaches in neuroendocrine tumor disease. Cancer Letter, 295(1), 100–109.CrossRef
12.
go back to reference Hanahan, D. (1985). Heritable formation of pancreatic beta-cell tumors in trasngenic mice expressing recombinant insulin/simian virus 40 oncogenes. Nature, 315, 115–122.PubMedCrossRef Hanahan, D. (1985). Heritable formation of pancreatic beta-cell tumors in trasngenic mice expressing recombinant insulin/simian virus 40 oncogenes. Nature, 315, 115–122.PubMedCrossRef
13.
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. Journal of Clinical Investigation, 111(9), 1287–1295.PubMed 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. Journal of Clinical Investigation, 111(9), 1287–1295.PubMed
14.
go back to reference Kitadai, Y., Sasaki, T., Nakamura, T., Bucana, C. D., & Fidler, I. J. (2006). Targeting the expression of platelet-derived growth factor receptor by reactive stroma inhibits growth ad metastasis of human colon carcinoma. American Journal of Pathology, 169, 2054–2065.PubMedCrossRef Kitadai, Y., Sasaki, T., Nakamura, T., Bucana, C. D., & Fidler, I. J. (2006). Targeting the expression of platelet-derived growth factor receptor by reactive stroma inhibits growth ad metastasis of human colon carcinoma. American Journal of Pathology, 169, 2054–2065.PubMedCrossRef
15.
go back to reference Kaplan, C. D., Kruger, J. A., Zhou, H., Luo, Y., Xiang, R., & Reisteld, R. A. (2006). A novel DNA vaccine encoding PDGFRbeta suppresses growth and dissemination of murine colon, lung and breast carcinoma. Vaccine, 24, 6994–7002.PubMedCrossRef Kaplan, C. D., Kruger, J. A., Zhou, H., Luo, Y., Xiang, R., & Reisteld, R. A. (2006). A novel DNA vaccine encoding PDGFRbeta suppresses growth and dissemination of murine colon, lung and breast carcinoma. Vaccine, 24, 6994–7002.PubMedCrossRef
16.
go back to reference Chiu, C. W., 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. Journal of Clinical Oncology, 28(29), 4425–4433.PubMedCrossRef Chiu, C. W., 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. Journal of Clinical Oncology, 28(29), 4425–4433.PubMedCrossRef
17.
go back to reference Rinke, A., Müller, H. H., Schade-Brittinger, C., Klose, K. J., 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. Journal of Clinical Oncology, 27(28), 4656–4663.PubMedCrossRef Rinke, A., Müller, H. H., Schade-Brittinger, C., Klose, K. J., 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. Journal of Clinical Oncology, 27(28), 4656–4663.PubMedCrossRef
18.
go back to reference Yao, J. C., Shah, M. H., Ito, T., Bohas, C. L., Wolin, E. M., Van Cutsem, E., et al. (2011). Everolimus for advanced pancreatic neuroendocrine tumors. The New England Journal of Medicine, 364(6), 514–523.PubMedCrossRef Yao, J. C., Shah, M. H., Ito, T., Bohas, C. L., Wolin, E. M., Van Cutsem, E., et al. (2011). Everolimus for advanced pancreatic neuroendocrine tumors. The New England Journal of Medicine, 364(6), 514–523.PubMedCrossRef
19.
go back to reference Raymond, E., Dahan, L., Raoul, J. L., Bang, Y. J., Borbath, I., Lombard-Bohas, C., et al. (2011). Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. The New England Journal of Medicine, 364(6), 501–513.PubMedCrossRef Raymond, E., Dahan, L., Raoul, J. L., Bang, Y. J., Borbath, I., Lombard-Bohas, C., et al. (2011). Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. The New England Journal of Medicine, 364(6), 501–513.PubMedCrossRef
20.
go back to reference Naraev, B., Strosberg, J. R., & Halfdanarson, T. R. (2012). Current status and perspectives of targeted therapy in well differentiated neuroendocrine tumors. Oncology, 83, 117–127.PubMedCrossRef Naraev, B., Strosberg, J. R., & Halfdanarson, T. R. (2012). Current status and perspectives of targeted therapy in well differentiated neuroendocrine tumors. Oncology, 83, 117–127.PubMedCrossRef
21.
go back to reference Kulke, M. H., Hornick, J. L., Frauenhoffer, C., Hooshmand, S., Ryan, D. P., Enzinger, P. C., et al. (2009). O 6-methylguanine DNA methyltransferase deficiency and response to temozolomide-based therapy in patients with neuroendocrine tumors. Clinical Cancer Research, 15(1), 338–345.PubMedCrossRef Kulke, M. H., Hornick, J. L., Frauenhoffer, C., Hooshmand, S., Ryan, D. P., Enzinger, P. C., et al. (2009). O 6-methylguanine DNA methyltransferase deficiency and response to temozolomide-based therapy in patients with neuroendocrine tumors. Clinical Cancer Research, 15(1), 338–345.PubMedCrossRef
22.
go back to reference Shah, T., Hochhauser, D., Frow, R., Quaglia, A., Dhillon, A. P., & Caplin, M. E. (2006). Epidermal growth factor receptor expression and activation in neuroendocrine tumours. Journal of Neuroendocrinology, 18(5), 355–360.PubMedCrossRef Shah, T., Hochhauser, D., Frow, R., Quaglia, A., Dhillon, A. P., & Caplin, M. E. (2006). Epidermal growth factor receptor expression and activation in neuroendocrine tumours. Journal of Neuroendocrinology, 18(5), 355–360.PubMedCrossRef
23.
go back to reference Srivastava, A., Alexander, J., Lomakin, I., & Dayal, Y. (2001). Immunohistochemical expression of transforming growth factor alpha and epidermal growth factor receptor in pancreatic endocrine tumors. Human Pathology, 32(11), 1184–1189.PubMedCrossRef Srivastava, A., Alexander, J., Lomakin, I., & Dayal, Y. (2001). Immunohistochemical expression of transforming growth factor alpha and epidermal growth factor receptor in pancreatic endocrine tumors. Human Pathology, 32(11), 1184–1189.PubMedCrossRef
24.
go back to reference Gilbert, J. A., Adhikari, L. J., Lloyd, R. V., Rubin, J., Haluska, P., Carboni, J. M., et al. (2010). Molecular markers for novel therapies in neuroendocrine (carcinoid) tumors. Endocrine-Related Cancer, 17(3), 623–636.PubMedCrossRef Gilbert, J. A., Adhikari, L. J., Lloyd, R. V., Rubin, J., Haluska, P., Carboni, J. M., et al. (2010). Molecular markers for novel therapies in neuroendocrine (carcinoid) tumors. Endocrine-Related Cancer, 17(3), 623–636.PubMedCrossRef
25.
go back to reference Rickman, O. B., Vohra, P. K., Sanyal, B., Vrana, J. A., Aubry, M. C., Wigle, D. A., et al. (2009). Analysis of ErbB receptors in pulmonary carcinoid tumors. Clinical Cancer Research, 15(10), 3315–3324.PubMedCrossRef Rickman, O. B., Vohra, P. K., Sanyal, B., Vrana, J. A., Aubry, M. C., Wigle, D. A., et al. (2009). Analysis of ErbB receptors in pulmonary carcinoid tumors. Clinical Cancer Research, 15(10), 3315–3324.PubMedCrossRef
26.
go back to reference Tannapfel, A., Vomschloss, S., Karhoff, D., Markwarth, A., Hengge, U. R., Wittekind, C., et al. (2005). BRAF gene mutations are rare events in gastroenteropancreatic neuroendocrine tumors. American Journal of Clinical Pathology, 123(2), 256–260.PubMedCrossRef Tannapfel, A., Vomschloss, S., Karhoff, D., Markwarth, A., Hengge, U. R., Wittekind, C., et al. (2005). BRAF gene mutations are rare events in gastroenteropancreatic neuroendocrine tumors. American Journal of Clinical Pathology, 123(2), 256–260.PubMedCrossRef
27.
go back to reference Hobday, T. J., Holen, K., Donehower, R., Camoriano, J., Kim, G., Picus, J., et al. (2006). A phase II trial of gefitinib in patients (pts) with progressive metastatic neuroendocrine tumors (NET): a Phase II Consortium (P2C) study. ASCO Meeting Abstracts, 24(18), 4043. Hobday, T. J., Holen, K., Donehower, R., Camoriano, J., Kim, G., Picus, J., et al. (2006). A phase II trial of gefitinib in patients (pts) with progressive metastatic neuroendocrine tumors (NET): a Phase II Consortium (P2C) study. ASCO Meeting Abstracts, 24(18), 4043.
28.
go back to reference Azzoni, C., Bottarelli, L., Cecchini, S., Lagrasta, C., Pizzi, S., D’Adda, T., et al. (2011). Involvement of HER-2/neu and metastasis-related proteins in the development of ileal neuroendocrine tumors. Virchows Archiv, 458(5), 525–536.PubMedCrossRef Azzoni, C., Bottarelli, L., Cecchini, S., Lagrasta, C., Pizzi, S., D’Adda, T., et al. (2011). Involvement of HER-2/neu and metastasis-related proteins in the development of ileal neuroendocrine tumors. Virchows Archiv, 458(5), 525–536.PubMedCrossRef
29.
go back to reference Hansel, D. E., Rahman, A., House, M., Ashfaq, R., Berg, K., Yeo, C. J., et al. (2004). Met proto-oncogene and insulin-like growth factor binding protein 3 overexpression correlates with metastatic ability in well-differentiated pancreatic endocrine neoplasms. Clinical Cancer Research, 10(18 Pt 1), 6152–6158.PubMedCrossRef Hansel, D. E., Rahman, A., House, M., Ashfaq, R., Berg, K., Yeo, C. J., et al. (2004). Met proto-oncogene and insulin-like growth factor binding protein 3 overexpression correlates with metastatic ability in well-differentiated pancreatic endocrine neoplasms. Clinical Cancer Research, 10(18 Pt 1), 6152–6158.PubMedCrossRef
30.
go back to reference Peghini, P. L., Iwamoto, M., Raffeld, M., Chen, Y. J., Goebel, S. U., Serrano, J., et al. (2002). Overexpression of epidermal growth factor and hepatocyte growth factor receptors in a proportion of gastrinomas correlates with aggressive growth and lower curability. Clinical Cancer Research, 8(7), 2273–2285.PubMed Peghini, P. L., Iwamoto, M., Raffeld, M., Chen, Y. J., Goebel, S. U., Serrano, J., et al. (2002). Overexpression of epidermal growth factor and hepatocyte growth factor receptors in a proportion of gastrinomas correlates with aggressive growth and lower curability. Clinical Cancer Research, 8(7), 2273–2285.PubMed
31.
go back to reference Sennino, B., Ishiguro-Oonuma, T., Wei, Y., Naylor, R. M., Williamson, C. W., Bhagwandin, V., et al. (2012). Suppression of tumor invasion and metastasis by concurrent inhibition of c-Met and VEGF signaling in pancreatic neuroendocrine tumors. Cancer Discovery, 2(3), 270–287.PubMedCrossRef Sennino, B., Ishiguro-Oonuma, T., Wei, Y., Naylor, R. M., Williamson, C. W., Bhagwandin, V., et al. (2012). Suppression of tumor invasion and metastasis by concurrent inhibition of c-Met and VEGF signaling in pancreatic neuroendocrine tumors. Cancer Discovery, 2(3), 270–287.PubMedCrossRef
32.
go back to reference Wulbrand, U., Remmert, G., Zofel, P., Wied, M., Arnold, R., & Fehmann, H. C. (2000). mRNA expression patterns of insulin-like growth factor system components in human neuroendocrine tumours. European Journal of Clinical Investigation, 30(8), 729–739.PubMedCrossRef Wulbrand, U., Remmert, G., Zofel, P., Wied, M., Arnold, R., & Fehmann, H. C. (2000). mRNA expression patterns of insulin-like growth factor system components in human neuroendocrine tumours. European Journal of Clinical Investigation, 30(8), 729–739.PubMedCrossRef
33.
go back to reference Hopfner, M., Baradari, V., Huether, A., Schofl, C., & Scherubl, H. (2006). The insulin-like growth factor receptor 1 is a promising target for novel treatment approaches in neuroendocrine gastrointestinal tumours. Endocrine-Related Cancer, 13(1), 135–149.PubMedCrossRef Hopfner, M., Baradari, V., Huether, A., Schofl, C., & Scherubl, H. (2006). The insulin-like growth factor receptor 1 is a promising target for novel treatment approaches in neuroendocrine gastrointestinal tumours. Endocrine-Related Cancer, 13(1), 135–149.PubMedCrossRef
34.
go back to reference Tolcher, A. W., Sarantopoulos, J., Patnaik, A., Papadopoulos, K., Lin, C. C., Rodon, J., et al. (2009). Phase I, pharmacokinetic, and pharmacodynamic study of AMG 479, a fully human monoclonal antibody to insulin-like growth factor receptor 1. Journal of Clinical Oncology, 27(34), 5800–5807.PubMedCrossRef Tolcher, A. W., Sarantopoulos, J., Patnaik, A., Papadopoulos, K., Lin, C. C., Rodon, J., et al. (2009). Phase I, pharmacokinetic, and pharmacodynamic study of AMG 479, a fully human monoclonal antibody to insulin-like growth factor receptor 1. Journal of Clinical Oncology, 27(34), 5800–5807.PubMedCrossRef
35.
go back to reference Fjallskog, M. L., Lejonklou, M. H., Oberg, K. E., Eriksson, B. K., & Janson, E. T. (2003). Expression of molecular targets for tyrosine kinase receptor antagonists in malignant endocrine pancreatic tumors. Clinical Cancer Research, 9(4), 1469–1473.PubMed Fjallskog, M. L., Lejonklou, M. H., Oberg, K. E., Eriksson, B. K., & Janson, E. T. (2003). Expression of molecular targets for tyrosine kinase receptor antagonists in malignant endocrine pancreatic tumors. Clinical Cancer Research, 9(4), 1469–1473.PubMed
36.
go back to reference Corbo, V., Beghelli, S., Bersani, S., Antonello, D., Talamini, G., Brunelli, M., et al. (2012). Pancreatic endocrine tumours: mutational and immunohistochemical survey of protein kinases reveals alterations in targetable kinases in cancer cell lines and rare primaries. Annals of Oncology, 23(1), 127–134.PubMedCrossRef Corbo, V., Beghelli, S., Bersani, S., Antonello, D., Talamini, G., Brunelli, M., et al. (2012). Pancreatic endocrine tumours: mutational and immunohistochemical survey of protein kinases reveals alterations in targetable kinases in cancer cell lines and rare primaries. Annals of Oncology, 23(1), 127–134.PubMedCrossRef
37.
go back to reference Bukowski, R. M., Eisen, T., Szczylik, C., Stadler, W. M., Simantov, R., Shan, M., et al. (2007). Final results of the randomized phase III trial of sorafenib in advanced renal cell carcinoma: survival and biomarker analysis. ASCO Meeting Abstracts, 25(18), 5023. Bukowski, R. M., Eisen, T., Szczylik, C., Stadler, W. M., Simantov, R., Shan, M., et al. (2007). Final results of the randomized phase III trial of sorafenib in advanced renal cell carcinoma: survival and biomarker analysis. ASCO Meeting Abstracts, 25(18), 5023.
38.
go back to reference Mass, R. D., Sarkar, S., Holden, S. N., & Hurwitz, H. (2005). Clinical benefit from bevacizumab (BV) in responding (R) and non-responding (NR) patients (pts) with metastatic colorectal cancer (mCRC). ASCO Meeting Abstracts, 23(16), 3514. Mass, R. D., Sarkar, S., Holden, S. N., & Hurwitz, H. (2005). Clinical benefit from bevacizumab (BV) in responding (R) and non-responding (NR) patients (pts) with metastatic colorectal cancer (mCRC). ASCO Meeting Abstracts, 23(16), 3514.
39.
go back to reference Terris, B., Scoazec, J. Y., Rubbia, L., Bregeaud, L., Pepper, M. S., Ruszniewski, P., et al. (1998). Expression of vascular endothelial growth factor in digestive neuroendocrine tumours. Histopathology, 32(2), 133–138.PubMedCrossRef Terris, B., Scoazec, J. Y., Rubbia, L., Bregeaud, L., Pepper, M. S., Ruszniewski, P., et al. (1998). Expression of vascular endothelial growth factor in digestive neuroendocrine tumours. Histopathology, 32(2), 133–138.PubMedCrossRef
40.
go back to reference La Rosa, S., Uccella, S., Finzi, G., Albarello, L., Sessa, F., & Capella, C. (2003). Localization of vascular endothelial growth factor and its receptors in digestive endocrine tumors: correlation with microvessel density and clinicopathologic features. Human Pathology, 34(1), 18–27.PubMedCrossRef La Rosa, S., Uccella, S., Finzi, G., Albarello, L., Sessa, F., & Capella, C. (2003). Localization of vascular endothelial growth factor and its receptors in digestive endocrine tumors: correlation with microvessel density and clinicopathologic features. Human Pathology, 34(1), 18–27.PubMedCrossRef
41.
go back to reference Oxboel, J., Binderup, T., Knigge, U., & Kjaer, A. (2009). Quantitative gene-expression of the tumor angiogenesis markers vascular endothelial growth factor, integrin alphaV and integrin beta3 in human neuroendocrine tumors. Oncology Reports, 21(3), 769–775.PubMed Oxboel, J., Binderup, T., Knigge, U., & Kjaer, A. (2009). Quantitative gene-expression of the tumor angiogenesis markers vascular endothelial growth factor, integrin alphaV and integrin beta3 in human neuroendocrine tumors. Oncology Reports, 21(3), 769–775.PubMed
42.
go back to reference Kulke, M. H., Lenz, H. J., Meropol, N. J., Posey, J., Ryan, D. P., Picus, J., et al. (2008). Activity of sunitinib in patients with advanced neuroendocrine tumors. Journal of Clinical Oncology, 26(20), 3403–3410.PubMedCrossRef Kulke, M. H., Lenz, H. J., Meropol, N. J., Posey, J., Ryan, D. P., Picus, J., et al. (2008). Activity of sunitinib in patients with advanced neuroendocrine tumors. Journal of Clinical Oncology, 26(20), 3403–3410.PubMedCrossRef
43.
go back to reference Bello, C., Deprimo, S. E., Friece, C., Smeraglia, J., Sherman, L., Tye, L., et al. (2006). Analysis of circulating biomarkers of sunitinib malate in patients with unresectable neuroendocrine tumors (NET): VEGF, IL-8, and soluble VEGF receptors 2 and 3. ASCO Meeting Abstracts, 24(18), 4045. Bello, C., Deprimo, S. E., Friece, C., Smeraglia, J., Sherman, L., Tye, L., et al. (2006). Analysis of circulating biomarkers of sunitinib malate in patients with unresectable neuroendocrine tumors (NET): VEGF, IL-8, and soluble VEGF receptors 2 and 3. ASCO Meeting Abstracts, 24(18), 4045.
44.
go back to reference Garcia-Donas, J., Esteban, E., Leandro-Garcia, L. J., Castellano, D. E., del Alba, A. G., Climent, M. A., et al. (2011). Single nucleotide polymorphism associations with response and toxic effects in patients with advanced renal-cell carcinoma treated with first-line sunitinib: a multicentre, observational, prospective study. The Lancet Oncology, 12(12), 1143–1150.PubMedCrossRef Garcia-Donas, J., Esteban, E., Leandro-Garcia, L. J., Castellano, D. E., del Alba, A. G., Climent, M. A., et al. (2011). Single nucleotide polymorphism associations with response and toxic effects in patients with advanced renal-cell carcinoma treated with first-line sunitinib: a multicentre, observational, prospective study. The Lancet Oncology, 12(12), 1143–1150.PubMedCrossRef
45.
go back to reference Yao, J. C. (2007). Neuroendocrine tumors. Molecular targeted therapy for carcinoid and islet-cell carcinoma. Best Practice & Research. Clinical Endocrinology & Metabolism, 21(1), 163–172.CrossRef Yao, J. C. (2007). Neuroendocrine tumors. Molecular targeted therapy for carcinoid and islet-cell carcinoma. Best Practice & Research. Clinical Endocrinology & Metabolism, 21(1), 163–172.CrossRef
46.
go back to reference Missiaglia, E., Dalai, I., Barbi, S., Beghelli, S., Falconi, M., della Peruta, M., et al. (2010). Pancreatic endocrine tumors: expression profiling evidences a role for AKT–mTOR pathway. Journal of Clinical Oncology, 28(2), 245–255.PubMedCrossRef Missiaglia, E., Dalai, I., Barbi, S., Beghelli, S., Falconi, M., della Peruta, M., et al. (2010). Pancreatic endocrine tumors: expression profiling evidences a role for AKT–mTOR pathway. Journal of Clinical Oncology, 28(2), 245–255.PubMedCrossRef
47.
go back to reference Corbo, V., Dalai, I., Scardoni, M., Barbi, S., Beghelli, S., Bersani, S., et al. (2010). MEN1 in pancreatic endocrine tumors: analysis of gene and protein status in 169 sporadic neoplasms reveals alterations in the vast majority of cases. Endocrine-Related Cancer, 17(3), 771–783.PubMedCrossRef Corbo, V., Dalai, I., Scardoni, M., Barbi, S., Beghelli, S., Bersani, S., et al. (2010). MEN1 in pancreatic endocrine tumors: analysis of gene and protein status in 169 sporadic neoplasms reveals alterations in the vast majority of cases. Endocrine-Related Cancer, 17(3), 771–783.PubMedCrossRef
48.
go back to reference Jiao, Y., Shi, C., Edil, B. H., de Wilde, R. F., Klimstra, D. S., Maitra, A., et al. (2011). DAXX/ATRX, MEN1, and mTOR pathway genes are frequently altered in pancreatic neuroendocrine tumors. Science, 331(6021), 1199–1203.PubMedCrossRef Jiao, Y., Shi, C., Edil, B. H., de Wilde, R. F., Klimstra, D. S., Maitra, A., et al. (2011). DAXX/ATRX, MEN1, and mTOR pathway genes are frequently altered in pancreatic neuroendocrine tumors. Science, 331(6021), 1199–1203.PubMedCrossRef
49.
go back to reference Shida, T., Kishimoto, T., Furuya, M., Nikaido, T., Koda, K., Takano, S., et al. (2010). Expression of an activated mammalian target of rapamycin (mTOR) in gastroenteropancreatic neuroendocrine tumors. Cancer Chemotherapy and Pharmacology, 65(5), 889–893.PubMedCrossRef Shida, T., Kishimoto, T., Furuya, M., Nikaido, T., Koda, K., Takano, S., et al. (2010). Expression of an activated mammalian target of rapamycin (mTOR) in gastroenteropancreatic neuroendocrine tumors. Cancer Chemotherapy and Pharmacology, 65(5), 889–893.PubMedCrossRef
50.
go back to reference Righi, L., Volante, M., Rapa, I., Tavaglione, V., Inzani, F., Pelosi, G., et al. (2010). Mammalian target of rapamycin signaling activation patterns in neuroendocrine tumors of the lung. Endocrine-Related Cancer, 17(4), 977–987.PubMedCrossRef Righi, L., Volante, M., Rapa, I., Tavaglione, V., Inzani, F., Pelosi, G., et al. (2010). Mammalian target of rapamycin signaling activation patterns in neuroendocrine tumors of the lung. Endocrine-Related Cancer, 17(4), 977–987.PubMedCrossRef
51.
go back to reference Pavel, M. E., Hainsworth, J. D., Baudin, E., Peeters, M., Horsch, D., Winkler, R. 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.PubMedCrossRef Pavel, M. E., Hainsworth, J. D., Baudin, E., Peeters, M., Horsch, D., Winkler, R. 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.PubMedCrossRef
52.
go back to reference Duran, I., Kortmansky, J., Singh, D., Hirte, H., Kocha, W., Goss, G., et al. (2006). A phase II clinical and pharmacodynamic study of temsirolimus in advanced neuroendocrine carcinomas. British Journal of Cancer, 95(9), 1148–1154.PubMedCrossRef Duran, I., Kortmansky, J., Singh, D., Hirte, H., Kocha, W., Goss, G., et al. (2006). A phase II clinical and pharmacodynamic study of temsirolimus in advanced neuroendocrine carcinomas. British Journal of Cancer, 95(9), 1148–1154.PubMedCrossRef
53.
go back to reference Cho, D., Signoretti, S., Dabora, S., Regan, M., Seeley, A., Mariotti, M., et al. (2007). Potential histologic and molecular predictors of response to temsirolimus in patients with advanced renal cell carcinoma. Clinical Genitourinary Cancer, 5(6), 379–385.PubMedCrossRef Cho, D., Signoretti, S., Dabora, S., Regan, M., Seeley, A., Mariotti, M., et al. (2007). Potential histologic and molecular predictors of response to temsirolimus in patients with advanced renal cell carcinoma. Clinical Genitourinary Cancer, 5(6), 379–385.PubMedCrossRef
54.
go back to reference Delbaldo, C., Albert, S., Dreyer, C., Sablin, M. P., Serova, M., Raymond, E., et al. (2011). Predictive biomarkers for the activity of mammalian target of rapamycin (mTOR) inhibitors. Targeted Oncology, 6(2), 119–124.PubMedCrossRef Delbaldo, C., Albert, S., Dreyer, C., Sablin, M. P., Serova, M., Raymond, E., et al. (2011). Predictive biomarkers for the activity of mammalian target of rapamycin (mTOR) inhibitors. Targeted Oncology, 6(2), 119–124.PubMedCrossRef
55.
go back to reference Roldo, C., Missiaglia, E., Hagan, J. P., Falconi, M., Capelli, P., Bersani, S., et al. (2006). MicroRNA expression abnormalities in pancreatic endocrine and acinar tumors are associated with distinctive pathologic features and clinical behavior. Journal of Clinical Oncology, 24(29), 4677–4684.PubMedCrossRef Roldo, C., Missiaglia, E., Hagan, J. P., Falconi, M., Capelli, P., Bersani, S., et al. (2006). MicroRNA expression abnormalities in pancreatic endocrine and acinar tumors are associated with distinctive pathologic features and clinical behavior. Journal of Clinical Oncology, 24(29), 4677–4684.PubMedCrossRef
56.
go back to reference Volinia, S., Calin, G. A., Liu, C. G., Ambs, S., Cimmino, A., Petrocca, F., et al. (2006). A microRNA expression signature of human solid tumors defines cancer gene targets. Proceedings of the National Academy of Sciences of the United States of America, 103(7), 2257–2261.PubMedCrossRef Volinia, S., Calin, G. A., Liu, C. G., Ambs, S., Cimmino, A., Petrocca, F., et al. (2006). A microRNA expression signature of human solid tumors defines cancer gene targets. Proceedings of the National Academy of Sciences of the United States of America, 103(7), 2257–2261.PubMedCrossRef
57.
go back to reference Svejda, B., Kidd, M., Kazberouk, A., Lawrence, B., Pfragner, R., & Modlin, I. M. (2011). Limitations in small intestinal neuroendocrine tumor therapy by mTor kinase inhibition reflect growth factor-mediated PI3K feedback loop activation via ERK1/2 and AKT. Cancer, 117(18), 4141–4154.PubMedCrossRef Svejda, B., Kidd, M., Kazberouk, A., Lawrence, B., Pfragner, R., & Modlin, I. M. (2011). Limitations in small intestinal neuroendocrine tumor therapy by mTor kinase inhibition reflect growth factor-mediated PI3K feedback loop activation via ERK1/2 and AKT. Cancer, 117(18), 4141–4154.PubMedCrossRef
58.
go back to reference Vilar, E., Salazar, R., Perez-Garcia, J., Cortes, J., Oberg, K., & Tabernero, J. (2007). Chemotherapy and role of the proliferation marker Ki-67 in digestive neuroendocrine tumors. Endocrine-Related Cancer, 14(2), 221–232.PubMedCrossRef Vilar, E., Salazar, R., Perez-Garcia, J., Cortes, J., Oberg, K., & Tabernero, J. (2007). Chemotherapy and role of the proliferation marker Ki-67 in digestive neuroendocrine tumors. Endocrine-Related Cancer, 14(2), 221–232.PubMedCrossRef
59.
go back to reference Tabernero, J., Rojo, F., Calvo, E., Burris, H., Judson, I., Hazell, K., et al. (2008). Dose- and schedule-dependent inhibition of the mammalian target of rapamycin pathway with everolimus: a phase I tumor pharmacodynamic study in patients with advanced solid tumors. Journal of Clinical Oncology, 26(10), 1603–1610.PubMedCrossRef Tabernero, J., Rojo, F., Calvo, E., Burris, H., Judson, I., Hazell, K., et al. (2008). Dose- and schedule-dependent inhibition of the mammalian target of rapamycin pathway with everolimus: a phase I tumor pharmacodynamic study in patients with advanced solid tumors. Journal of Clinical Oncology, 26(10), 1603–1610.PubMedCrossRef
60.
go back to reference Yao, J. C., Phan, A. T., Chang, D. Z., Wolff, R. A., Hess, K., Gupta, S., et al. (2008). Efficacy of RAD001 (everolimus) and octreotide LAR in advanced low- to intermediate-grade neuroendocrine tumors: results of a phase II study. Journal of Clinical Oncology, 26(26), 4311–4318.PubMedCrossRef Yao, J. C., Phan, A. T., Chang, D. Z., Wolff, R. A., Hess, K., Gupta, S., et al. (2008). Efficacy of RAD001 (everolimus) and octreotide LAR in advanced low- to intermediate-grade neuroendocrine tumors: results of a phase II study. Journal of Clinical Oncology, 26(26), 4311–4318.PubMedCrossRef
61.
go back to reference Yao, J. C., Lombard-Bohas, C., Baudin, E., Kvols, L. K., Rougier, P., Ruszniewski, P., et al. (2010). Daily oral everolimus activity in patients with metastatic pancreatic neuroendocrine tumors after failure of cytotoxic chemotherapy: a phase II trial. Journal of Clinical Oncology, 28(1), 69–76.PubMedCrossRef Yao, J. C., Lombard-Bohas, C., Baudin, E., Kvols, L. K., Rougier, P., Ruszniewski, P., et al. (2010). Daily oral everolimus activity in patients with metastatic pancreatic neuroendocrine tumors after failure of cytotoxic chemotherapy: a phase II trial. Journal of Clinical Oncology, 28(1), 69–76.PubMedCrossRef
62.
go back to reference De Vries, E., Anthony, L. B., Sideris, L., Chen, L., Lebrec, J., Tsuchihashi, Z., et al. (2011). Effect of everolimus treatment on chromogranin A, neuron-specific enolase, gastrin, and glucagon levels in patients with advanced pancreatic neuroendocrine tumors (pNET): phase III RADIANT-3 Study results. ASCO Meeting Abstracts, 29(15_Suppl), 10624. De Vries, E., Anthony, L. B., Sideris, L., Chen, L., Lebrec, J., Tsuchihashi, Z., et al. (2011). Effect of everolimus treatment on chromogranin A, neuron-specific enolase, gastrin, and glucagon levels in patients with advanced pancreatic neuroendocrine tumors (pNET): phase III RADIANT-3 Study results. ASCO Meeting Abstracts, 29(15_Suppl), 10624.
63.
go back to reference Yao, J. C., Ricci, S., Winkler, R. E., Jehl, V., & Pavel, M. E. (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. ASCO Meeting Abstracts, 29(15_Suppl), 4011. Yao, J. C., Ricci, S., Winkler, R. E., Jehl, V., & Pavel, M. E. (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. ASCO Meeting Abstracts, 29(15_Suppl), 4011.
64.
go back to reference Yao, J. C., Phan, A. T., Fogleman, D., Ng, C. S., Jacobs, C. B., Dagohoy, C. D., et al. (2010). Randomized run-in study of bevacizumab (B) and everolimus (E) in low- to intermediate-grade neuroendocrine tumors (LGNETs) using perfusion CT as functional biomarker. ASCO Meeting Abstracts, 28(15_Suppl), 4002. Yao, J. C., Phan, A. T., Fogleman, D., Ng, C. S., Jacobs, C. B., Dagohoy, C. D., et al. (2010). Randomized run-in study of bevacizumab (B) and everolimus (E) in low- to intermediate-grade neuroendocrine tumors (LGNETs) using perfusion CT as functional biomarker. ASCO Meeting Abstracts, 28(15_Suppl), 4002.
65.
go back to reference Yao, J. C., Phan, A., Hoff, P. M., Chen, H. X., Charnsangavej, C., Yeung, S. C., et al. (2008). 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. Journal of Clinical Oncology, 26(8), 1316–1323.PubMedCrossRef Yao, J. C., Phan, A., Hoff, P. M., Chen, H. X., Charnsangavej, C., Yeung, S. C., et al. (2008). 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. Journal of Clinical Oncology, 26(8), 1316–1323.PubMedCrossRef
66.
go back to reference Ng, C. S., Charnsangavej, C., Wei, W., & Yao, J. C. (2011). Perfusion CT findings in patients with metastatic carcinoid tumors undergoing bevacizumab and interferon therapy. AJR. American Journal of Roentgenology, 196(3), 569–576.PubMedCrossRef Ng, C. S., Charnsangavej, C., Wei, W., & Yao, J. C. (2011). Perfusion CT findings in patients with metastatic carcinoid tumors undergoing bevacizumab and interferon therapy. AJR. American Journal of Roentgenology, 196(3), 569–576.PubMedCrossRef
67.
go back to reference Ng, C. S., Wang, X., Faria, S. C., Lin, E., Charnsangavej, C., & Tannir, N. M. (2010). Perfusion CT in patients with metastatic renal cell carcinoma treated with interferon. AJR. American Journal of Roentgenology, 194(1), 166–171.PubMedCrossRef Ng, C. S., Wang, X., Faria, S. C., Lin, E., Charnsangavej, C., & Tannir, N. M. (2010). Perfusion CT in patients with metastatic renal cell carcinoma treated with interferon. AJR. American Journal of Roentgenology, 194(1), 166–171.PubMedCrossRef
68.
go back to reference Phan, A. T., Yao, J. C., Fogelman, D. R., Hess, K. R., Ng, C. S., Bullock, S. A., et al. (2010). A prospective, multi-institutional phase II study of GW786034 (pazopanib) and depot octreotide (sandostatin LAR) in advanced low-grade neuroendocrine carcinoma (LGNEC). ASCO Meeting Abstracts, 28(15_Suppl), 4001. Phan, A. T., Yao, J. C., Fogelman, D. R., Hess, K. R., Ng, C. S., Bullock, S. A., et al. (2010). A prospective, multi-institutional phase II study of GW786034 (pazopanib) and depot octreotide (sandostatin LAR) in advanced low-grade neuroendocrine carcinoma (LGNEC). ASCO Meeting Abstracts, 28(15_Suppl), 4001.
69.
go back to reference Cui, T., Hurtig, M., Elgue, G., Li, S. C., Veronesi, G., Essaghir, A., et al. (2010). Paraneoplastic antigen Ma2 autoantibodies as specific blood biomarkers for detection of early recurrence of small intestine neuroendocrine tumors. PLoS One, 5(12), e16010.PubMedCrossRef Cui, T., Hurtig, M., Elgue, G., Li, S. C., Veronesi, G., Essaghir, A., et al. (2010). Paraneoplastic antigen Ma2 autoantibodies as specific blood biomarkers for detection of early recurrence of small intestine neuroendocrine tumors. PLoS One, 5(12), e16010.PubMedCrossRef
70.
go back to reference Khan, M. S., Tsigani, T., Rashid, M., Rabouhans, J. S., Yu, D., Luong, T. V., et al. (2011). Circulating tumor cells and EpCAM expression in neuroendocrine tumors. Clinical Cancer Research, 17(2), 337–345.PubMedCrossRef Khan, M. S., Tsigani, T., Rashid, M., Rabouhans, J. S., Yu, D., Luong, T. V., et al. (2011). Circulating tumor cells and EpCAM expression in neuroendocrine tumors. Clinical Cancer Research, 17(2), 337–345.PubMedCrossRef
Metadata
Title
The targeted therapy revolution in neuroendocrine tumors: in search of biomarkers for patient selection and response evaluation
Authors
Sara De Dosso
Enrique Grande
Jorge Barriuso
Daniel Castellano
Josep Tabernero
Jaume Capdevila
Publication date
01-12-2013
Publisher
Springer US
Published in
Cancer and Metastasis Reviews / Issue 3-4/2013
Print ISSN: 0167-7659
Electronic ISSN: 1573-7233
DOI
https://doi.org/10.1007/s10555-013-9421-0

Other articles of this Issue 3-4/2013

Cancer and Metastasis Reviews 3-4/2013 Go to the issue

EditorialNotes

Preface

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