Skip to main content
Top
Published in: Cancer and Metastasis Reviews 1/2011

01-03-2011

Somatostatin analogs for the treatment of neuroendocrine tumors

Authors: Michael D. Culler, Kjell Öberg, Rudolf Arnold, Eric P. Krenning, Isabel Sevilla, José Ángel Díaz

Published in: Cancer and Metastasis Reviews | Special Issue 1/2011

Login to get access

Abstract

Somatostatin is an important regulator of endocrine and exocrine secretion, affecting the release of many hormones. The effects of somatostatin are mediated through its interaction with one of five somatostatin receptors. Gastroenteropancreatic neuroendocrine tumors (GEP-NETs) express multiple somatostatin receptors, making them excellent potential therapeutic targets. Many trials have shown that treatment with somatostatin analogs is associated with disease stabilization and prolonged survival. More recently, somatostatin analogs have been shown to have antiproliferative effects, thus broadening the scope of their uses. In this review, we update the current data on the treatment of GEP-NETs with somatostatin analogs, with particular emphasis on the results of the PROMID study. In addition, we discuss the current state of knowledge of novel therapies against GEP-NETs, including the use of somatostatin analogs with broader receptor binding profiles, chimeric somatostatin–dopamine molecules, combinations of somatostatin analogs with other active chemotherapy agents, and peptide receptor-targeted radionuclide therapy.
Literature
1.
go back to reference Brazeau, P., Vale, W., Burgus, R., et al. (1973). Hypothalamic polypeptide that inhibits the secretion of immunoreactive pituitary growth hormone. Science, 179(68), 77–79.PubMedCrossRef Brazeau, P., Vale, W., Burgus, R., et al. (1973). Hypothalamic polypeptide that inhibits the secretion of immunoreactive pituitary growth hormone. Science, 179(68), 77–79.PubMedCrossRef
2.
3.
go back to reference Serri, O., Brazeau, P., Kachra, Z., et al. (1992). Octreotide inhibits insulin-like growth factor-I hepatic gene expression in the hypophysectomized rat: Evidence for a direct and indirect mechanism of action. Endocrinology, 130(4), 1816–1821.PubMedCrossRef Serri, O., Brazeau, P., Kachra, Z., et al. (1992). Octreotide inhibits insulin-like growth factor-I hepatic gene expression in the hypophysectomized rat: Evidence for a direct and indirect mechanism of action. Endocrinology, 130(4), 1816–1821.PubMedCrossRef
4.
go back to reference Epelbaum, J. (1986). Somatostatin in the central nervous system: Physiology and pathological modifications. Progress in Neurobiology, 27(1), 63–100.PubMedCrossRef Epelbaum, J. (1986). Somatostatin in the central nervous system: Physiology and pathological modifications. Progress in Neurobiology, 27(1), 63–100.PubMedCrossRef
6.
go back to reference Bruns, C., Raulf, F., Hoyer, D., et al. (1996). Binding properties of somatostatin receptor subtypes. Metabolism, 45(8 Suppl 1), 17–20.PubMedCrossRef Bruns, C., Raulf, F., Hoyer, D., et al. (1996). Binding properties of somatostatin receptor subtypes. Metabolism, 45(8 Suppl 1), 17–20.PubMedCrossRef
7.
go back to reference Hoyer, D., Lubbert, H., & Bruns, C. (1994). Molecular pharmacology of somatostatin receptors. Naunyn Schmiedebergs Arch Pharmacol, 350(5), 441–453.PubMedCrossRef Hoyer, D., Lubbert, H., & Bruns, C. (1994). Molecular pharmacology of somatostatin receptors. Naunyn Schmiedebergs Arch Pharmacol, 350(5), 441–453.PubMedCrossRef
8.
go back to reference Oberg, K. E., Reubi, J. C., Kwekkeboom, D. J., et al. (2010). Role of somatostatins in gastroenteropancreatic neuroendocrine tumor development and therapy. Gastroenterology, 139(3), 742–753. 753 e1.PubMedCrossRef Oberg, K. E., Reubi, J. C., Kwekkeboom, D. J., et al. (2010). Role of somatostatins in gastroenteropancreatic neuroendocrine tumor development and therapy. Gastroenterology, 139(3), 742–753. 753 e1.PubMedCrossRef
9.
go back to reference Bauer, W., Briner, U., Doepfner, W., et al. (1982). SMS 201-995: A very potent and selective octapeptide analogue of somatostatin with prolonged action. Life Sciences, 31(11), 1133–1140.PubMedCrossRef Bauer, W., Briner, U., Doepfner, W., et al. (1982). SMS 201-995: A very potent and selective octapeptide analogue of somatostatin with prolonged action. Life Sciences, 31(11), 1133–1140.PubMedCrossRef
10.
go back to reference Murphy, W. A., Lance, V. A., Moreau, S., et al. (1987). Inhibition of rat prostate tumor growth by an octapeptide analog of somatostatin. Life Sciences, 40(26), 2515–2522.PubMedCrossRef Murphy, W. A., Lance, V. A., Moreau, S., et al. (1987). Inhibition of rat prostate tumor growth by an octapeptide analog of somatostatin. Life Sciences, 40(26), 2515–2522.PubMedCrossRef
11.
go back to reference Karashima, T., Cai, R. Z., & Schally, A. V. (1987). Effects of highly potent octapeptide analogs of somatostatin on growth hormone, insulin and glucagon release. Life Sciences, 41(8), 1011–1019.PubMedCrossRef Karashima, T., Cai, R. Z., & Schally, A. V. (1987). Effects of highly potent octapeptide analogs of somatostatin on growth hormone, insulin and glucagon release. Life Sciences, 41(8), 1011–1019.PubMedCrossRef
12.
go back to reference Mazziotti, G., Floriani, I., Bonadonna, S., et al. (2009). Effects of somatostatin analogs on glucose homeostasis: A metaanalysis of acromegaly studies. The Journal of Clinical Endocrinology and Metabolism, 94(5), 1500–1508.PubMedCrossRef Mazziotti, G., Floriani, I., Bonadonna, S., et al. (2009). Effects of somatostatin analogs on glucose homeostasis: A metaanalysis of acromegaly studies. The Journal of Clinical Endocrinology and Metabolism, 94(5), 1500–1508.PubMedCrossRef
13.
go back to reference Strosberg, J., Gardner, N., & Kvols, L. (2009). Survival and prognostic factor analysis of 146 metastatic neuroendocrine tumors of the mid-gut. Neuroendocrinology, 89(4), 471–476.PubMedCrossRef Strosberg, J., Gardner, N., & Kvols, L. (2009). Survival and prognostic factor analysis of 146 metastatic neuroendocrine tumors of the mid-gut. Neuroendocrinology, 89(4), 471–476.PubMedCrossRef
14.
go back to reference Arnold, R., Trautmann, M. E., Creutzfeldt, W., et al. (1996). Somatostatin analogue octreotide and inhibition of tumour growth in metastatic endocrine gastroenteropancreatic tumours. Gut, 38(3), 430–438.PubMedCrossRef Arnold, R., Trautmann, M. E., Creutzfeldt, W., et al. (1996). Somatostatin analogue octreotide and inhibition of tumour growth in metastatic endocrine gastroenteropancreatic tumours. Gut, 38(3), 430–438.PubMedCrossRef
15.
go back to reference Saltz, L., Trochanowski, B., Buckley, M., et al. (1993). Octreotide as an antineoplastic agent in the treatment of functional and nonfunctional neuroendocrine tumors. Cancer, 72(1), 244–248.PubMedCrossRef Saltz, L., Trochanowski, B., Buckley, M., et al. (1993). Octreotide as an antineoplastic agent in the treatment of functional and nonfunctional neuroendocrine tumors. Cancer, 72(1), 244–248.PubMedCrossRef
16.
go back to reference Ducreux, M., Ruszniewski, P., Chayvialle, J. A., et al. (2000). The antitumoral effect of the long-acting somatostatin analog lanreotide in neuroendocrine tumors. The American Journal of Gastroenterology, 95(11), 3276–3281.PubMedCrossRef Ducreux, M., Ruszniewski, P., Chayvialle, J. A., et al. (2000). The antitumoral effect of the long-acting somatostatin analog lanreotide in neuroendocrine tumors. The American Journal of Gastroenterology, 95(11), 3276–3281.PubMedCrossRef
17.
go back to reference Wymenga, A. N., Eriksson, B., Salmela, P. I., et al. (1999). Efficacy and safety of prolonged-release lanreotide in patients with gastrointestinal neuroendocrine tumors and hormone-related symptoms. Journal of Clinical Oncology, 17(4), 1111.PubMed Wymenga, A. N., Eriksson, B., Salmela, P. I., et al. (1999). Efficacy and safety of prolonged-release lanreotide in patients with gastrointestinal neuroendocrine tumors and hormone-related symptoms. Journal of Clinical Oncology, 17(4), 1111.PubMed
18.
go back to reference Susini, C., & Buscail, L. (2006). Rationale for the use of somatostatin analogs as antitumor agents. Annals of Oncology, 17(12), 1733–1742.PubMedCrossRef Susini, C., & Buscail, L. (2006). Rationale for the use of somatostatin analogs as antitumor agents. Annals of Oncology, 17(12), 1733–1742.PubMedCrossRef
19.
go back to reference Weckbecker, G., Lewis, I., Albert, R., et al. (2003). Opportunities in somatostatin research: Biological, chemical and therapeutic aspects. Nature Reviews. Drug Discovery, 2(12), 999–1017.PubMedCrossRef Weckbecker, G., Lewis, I., Albert, R., et al. (2003). Opportunities in somatostatin research: Biological, chemical and therapeutic aspects. Nature Reviews. Drug Discovery, 2(12), 999–1017.PubMedCrossRef
20.
go back to reference Lattuada, D., Casnici, C., Venuto, A., et al. (2002). The apoptotic effect of somatostatin analogue SMS 201-995 on human lymphocytes. Journal of Neuroimmunology, 133(1–2), 211–216.PubMedCrossRef Lattuada, D., Casnici, C., Venuto, A., et al. (2002). The apoptotic effect of somatostatin analogue SMS 201-995 on human lymphocytes. Journal of Neuroimmunology, 133(1–2), 211–216.PubMedCrossRef
21.
go back to reference Florio, T. (2008). Somatostatin/somatostatin receptor signalling: Phosphotyrosine phosphatases. Molecular and Cellular Endocrinology, 286(1–2), 40–48.PubMedCrossRef Florio, T. (2008). Somatostatin/somatostatin receptor signalling: Phosphotyrosine phosphatases. Molecular and Cellular Endocrinology, 286(1–2), 40–48.PubMedCrossRef
22.
go back to reference Florio, T., Morini, M., Villa, V., et al. (2003). Somatostatin inhibits tumor angiogenesis and growth via somatostatin receptor-3-mediated regulation of endothelial nitric oxide synthase and mitogen-activated protein kinase activities. Endocrinology, 144(4), 1574–1584.PubMedCrossRef Florio, T., Morini, M., Villa, V., et al. (2003). Somatostatin inhibits tumor angiogenesis and growth via somatostatin receptor-3-mediated regulation of endothelial nitric oxide synthase and mitogen-activated protein kinase activities. Endocrinology, 144(4), 1574–1584.PubMedCrossRef
23.
go back to reference Hofland, L. J., & Lamberts, S. W. (2003). The pathophysiological consequences of somatostatin receptor internalization and resistance. Endocrine Reviews, 24(1), 28–47.PubMedCrossRef Hofland, L. J., & Lamberts, S. W. (2003). The pathophysiological consequences of somatostatin receptor internalization and resistance. Endocrine Reviews, 24(1), 28–47.PubMedCrossRef
24.
go back to reference Bruns, C., Lewis, I., Briner, U., et al. (2002). SOM230: A novel somatostatin peptidomimetic with broad somatotropin release inhibiting factor (SRIF) receptor binding and a unique antisecretory profile. European Journal of Endocrinology, 146(5), 707–716.PubMedCrossRef Bruns, C., Lewis, I., Briner, U., et al. (2002). SOM230: A novel somatostatin peptidomimetic with broad somatotropin release inhibiting factor (SRIF) receptor binding and a unique antisecretory profile. European Journal of Endocrinology, 146(5), 707–716.PubMedCrossRef
25.
go back to reference Imtiaz, K. E., Monteith, P., & Khaleeli, A. (2000). Complete histological regression of metastatic carcinoid tumour after treatment with octreotide. Clinical Endocrinology (Oxford), 53(6), 755–758.CrossRef Imtiaz, K. E., Monteith, P., & Khaleeli, A. (2000). Complete histological regression of metastatic carcinoid tumour after treatment with octreotide. Clinical Endocrinology (Oxford), 53(6), 755–758.CrossRef
26.
go back to reference Joensuu, H., Katka, K., & Kujari, H. (1992). Dramatic response of a metastatic carcinoid tumour to a combination of interferon and octreotide. Acta Endocrinologica (Copenhagen), 126(2), 184–185. Joensuu, H., Katka, K., & Kujari, H. (1992). Dramatic response of a metastatic carcinoid tumour to a combination of interferon and octreotide. Acta Endocrinologica (Copenhagen), 126(2), 184–185.
27.
go back to reference Arnold, R., Rinke, A., Klose, K. J., et al. (2005). Octreotide versus octreotide plus interferon-alpha in endocrine gastroenteropancreatic tumors: A randomized trial. Clinical Gastroenterology and Hepatology, 3(8), 761–771.PubMedCrossRef Arnold, R., Rinke, A., Klose, K. J., et al. (2005). Octreotide versus octreotide plus interferon-alpha in endocrine gastroenteropancreatic tumors: A randomized trial. Clinical Gastroenterology and Hepatology, 3(8), 761–771.PubMedCrossRef
28.
go back to reference Faiss, S., Pape, U. F., Bohmig, M., et al. (2003). Prospective, randomized, multicenter trial on the antiproliferative effect of lanreotide, interferon alfa, and their combination for therapy of metastatic neuroendocrine gastroenteropancreatic tumors—the International Lanreotide and Interferon Alfa Study Group. Journal of Clinical Oncology, 21(14), 2689–2696.PubMedCrossRef Faiss, S., Pape, U. F., Bohmig, M., et al. (2003). Prospective, randomized, multicenter trial on the antiproliferative effect of lanreotide, interferon alfa, and their combination for therapy of metastatic neuroendocrine gastroenteropancreatic tumors—the International Lanreotide and Interferon Alfa Study Group. Journal of Clinical Oncology, 21(14), 2689–2696.PubMedCrossRef
29.
go back to reference Aparicio, T., Ducreux, M., Baudin, E., et al. (2001). Antitumour activity of somatostatin analogues in progressive metastatic neuroendocrine tumours. European Journal of Cancer, 37(8), 1014–1019.PubMedCrossRef Aparicio, T., Ducreux, M., Baudin, E., et al. (2001). Antitumour activity of somatostatin analogues in progressive metastatic neuroendocrine tumours. European Journal of Cancer, 37(8), 1014–1019.PubMedCrossRef
30.
go back to reference di Bartolomeo, M., Bajetta, E., Buzzoni, R., et al. (1996). Clinical efficacy of octreotide in the treatment of metastatic neuroendocrine tumors. A study by the Italian trials in medical oncology group. Cancer, 77(2), 402–408.PubMedCrossRef di Bartolomeo, M., Bajetta, E., Buzzoni, R., et al. (1996). Clinical efficacy of octreotide in the treatment of metastatic neuroendocrine tumors. A study by the Italian trials in medical oncology group. Cancer, 77(2), 402–408.PubMedCrossRef
31.
go back to reference Rinke, A., Müller, H.-H., Schade-Brittinger, C., 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., 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
32.
go back to reference Blumberg J, Gómez-Panzani E, Latapie-Martínez S, et al. Somatuline® Autogel® 120 mg (lanreotide) evaluation of tumor progression-free survival in patients with non-functioning entero-pancreatic endocrine tumors: An ongoing, double-blind, randomized, placebo-controlled, multicenter study (the CLARINET Study). NANETS—2010 Neuroendocrine Tumor Symposium (abstr. 726). Blumberg J, Gómez-Panzani E, Latapie-Martínez S, et al. Somatuline® Autogel® 120 mg (lanreotide) evaluation of tumor progression-free survival in patients with non-functioning entero-pancreatic endocrine tumors: An ongoing, double-blind, randomized, placebo-controlled, multicenter study (the CLARINET Study). NANETS—2010 Neuroendocrine Tumor Symposium (abstr. 726).
33.
go back to reference Rocheville, M., Lange, D. C., Kumar, U., et al. (2000). Receptors for dopamine and somatostatin: Formation of hetero-oligomers with enhanced functional activity. Science, 288(5463), 154–157.PubMedCrossRef Rocheville, M., Lange, D. C., Kumar, U., et al. (2000). Receptors for dopamine and somatostatin: Formation of hetero-oligomers with enhanced functional activity. Science, 288(5463), 154–157.PubMedCrossRef
34.
go back to reference Schmid, H. A. (2008). Pasireotide (SOM230): Development, mechanism of action and potential applications. Molecular and Cellular Endocrinology, 286(1–2), 69–74.PubMedCrossRef Schmid, H. A. (2008). Pasireotide (SOM230): Development, mechanism of action and potential applications. Molecular and Cellular Endocrinology, 286(1–2), 69–74.PubMedCrossRef
35.
go back to reference Van Vugt, H. H., Schmid, H. A., Sailer, A. W. (2008) Ligand-dependent internalization of somatostatin receptors. In: Endocrine abstract. p. 654. Van Vugt, H. H., Schmid, H. A., Sailer, A. W. (2008) Ligand-dependent internalization of somatostatin receptors. In: Endocrine abstract. p. 654.
36.
go back to reference Kvols, L., Wiedenmann, B., Oberg, K., et al. (2006). Safety and efficacy of pasireotide (SOM230) in patients with metastatic carcinoid tumors refractory or resistant to octreotide LAR: Results of a phase II study. Journal of Clinical Oncology (Meeting Abstracts), 24(18_suppl), 4082-. Kvols, L., Wiedenmann, B., Oberg, K., et al. (2006). Safety and efficacy of pasireotide (SOM230) in patients with metastatic carcinoid tumors refractory or resistant to octreotide LAR: Results of a phase II study. Journal of Clinical Oncology (Meeting Abstracts), 24(18_suppl), 4082-.
37.
go back to reference Lamberts, S. W., van der Lely, A. J., de Herder, W. W., et al. (1996). Octreotide. The New England Journal of Medicine, 334(4), 246–254.PubMedCrossRef Lamberts, S. W., van der Lely, A. J., de Herder, W. W., et al. (1996). Octreotide. The New England Journal of Medicine, 334(4), 246–254.PubMedCrossRef
38.
go back to reference Rocheville, M., Lange, D. C., Kumar, U., et al. (2000). Subtypes of the somatostatin receptor assemble as functional homo- and heterodimers. The Journal of Biological Chemistry, 275(11), 7862–7869.PubMedCrossRef Rocheville, M., Lange, D. C., Kumar, U., et al. (2000). Subtypes of the somatostatin receptor assemble as functional homo- and heterodimers. The Journal of Biological Chemistry, 275(11), 7862–7869.PubMedCrossRef
39.
go back to reference Saveanu, A., Gunz, G., Dufour, H., et al. (2001). Bim-23244, a somatostatin receptor subtype 2- and 5-selective analog with enhanced efficacy in suppressing growth hormone (GH) from octreotide-resistant human GH-secreting adenomas. The Journal of Clinical Endocrinology and Metabolism, 86(1), 140–145.PubMedCrossRef Saveanu, A., Gunz, G., Dufour, H., et al. (2001). Bim-23244, a somatostatin receptor subtype 2- and 5-selective analog with enhanced efficacy in suppressing growth hormone (GH) from octreotide-resistant human GH-secreting adenomas. The Journal of Clinical Endocrinology and Metabolism, 86(1), 140–145.PubMedCrossRef
40.
go back to reference Minniti, G., Jaffrain-Rea, M. L., Baldelli, R., et al. (1997). Acute effects of octreotide, cabergoline and a combination of both drugs on GH secretion in acromegalic patients. La Clinica Terapeutica, 148(12), 601–607.PubMed Minniti, G., Jaffrain-Rea, M. L., Baldelli, R., et al. (1997). Acute effects of octreotide, cabergoline and a combination of both drugs on GH secretion in acromegalic patients. La Clinica Terapeutica, 148(12), 601–607.PubMed
41.
go back to reference Flogstad, A. K., Halse, J., Grass, P., et al. (1994). A comparison of octreotide, bromocriptine, or a combination of both drugs in acromegaly. The Journal of Clinical Endocrinology and Metabolism, 79(2), 461–465.PubMedCrossRef Flogstad, A. K., Halse, J., Grass, P., et al. (1994). A comparison of octreotide, bromocriptine, or a combination of both drugs in acromegaly. The Journal of Clinical Endocrinology and Metabolism, 79(2), 461–465.PubMedCrossRef
42.
go back to reference Saveanu, A., Lavaque, E., Gunz, G., et al. (2002). Demonstration of enhanced potency of a chimeric somatostatin–dopamine molecule, BIM-23A387, in suppressing growth hormone and prolactin secretion from human pituitary somatotroph adenoma cells. The Journal of Clinical Endocrinology and Metabolism, 87(12), 5545–5552.PubMedCrossRef Saveanu, A., Lavaque, E., Gunz, G., et al. (2002). Demonstration of enhanced potency of a chimeric somatostatin–dopamine molecule, BIM-23A387, in suppressing growth hormone and prolactin secretion from human pituitary somatotroph adenoma cells. The Journal of Clinical Endocrinology and Metabolism, 87(12), 5545–5552.PubMedCrossRef
43.
go back to reference Kidd, M., Modlin, I. M., Black, J. W., et al. (2007). A comparison of the effects of gastrin, somatostatin and dopamine receptor ligands on rat gastric enterochromaffin-like cell secretion and proliferation. Regulatory Peptides, 143(1–3), 109–117.PubMedCrossRef Kidd, M., Modlin, I. M., Black, J. W., et al. (2007). A comparison of the effects of gastrin, somatostatin and dopamine receptor ligands on rat gastric enterochromaffin-like cell secretion and proliferation. Regulatory Peptides, 143(1–3), 109–117.PubMedCrossRef
44.
go back to reference O’Toole, D., Saveanu, A., Couvelard, A., et al. (2006). The analysis of quantitative expression of somatostatin and dopamine receptors in gastro-entero-pancreatic tumours opens new therapeutic strategies. European Journal of Endocrinology, 155(6), 849–857.PubMedCrossRef O’Toole, D., Saveanu, A., Couvelard, A., et al. (2006). The analysis of quantitative expression of somatostatin and dopamine receptors in gastro-entero-pancreatic tumours opens new therapeutic strategies. European Journal of Endocrinology, 155(6), 849–857.PubMedCrossRef
45.
go back to reference Hebert, T. E., & Bouvier, M. (1998). Structural and functional aspects of G protein-coupled receptor oligomerization. Biochemistry and Cell Biology, 76(1), 1–11.PubMedCrossRef Hebert, T. E., & Bouvier, M. (1998). Structural and functional aspects of G protein-coupled receptor oligomerization. Biochemistry and Cell Biology, 76(1), 1–11.PubMedCrossRef
46.
go back to reference Tiensuu Janson, E. M., Ahlstrom, H., Andersson, T., et al. (1992). Octreotide and interferon alfa: A new combination for the treatment of malignant carcinoid tumours. European Journal of Cancer, 28A(10), 1647–1650.PubMedCrossRef Tiensuu Janson, E. M., Ahlstrom, H., Andersson, T., et al. (1992). Octreotide and interferon alfa: A new combination for the treatment of malignant carcinoid tumours. European Journal of Cancer, 28A(10), 1647–1650.PubMedCrossRef
47.
go back to reference Kolby, L., Persson, G., Franzen, S., et al. (2003). Randomized clinical trial of the effect of interferon alpha on survival in patients with disseminated midgut carcinoid tumours. The British Journal of Surgery, 90(6), 687–693.PubMedCrossRef Kolby, L., Persson, G., Franzen, S., et al. (2003). Randomized clinical trial of the effect of interferon alpha on survival in patients with disseminated midgut carcinoid tumours. The British Journal of Surgery, 90(6), 687–693.PubMedCrossRef
48.
go back to reference Terris, B., Scoazec, J. Y., Rubbia, L., 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., et al. (1998). Expression of vascular endothelial growth factor in digestive neuroendocrine tumours. Histopathology, 32(2), 133–138.PubMedCrossRef
49.
go back to reference Konno, H., Arai, T., Tanaka, T., et al. (1998). Antitumor effect of a neutralizing antibody to vascular endothelial growth factor on liver metastasis of endocrine neoplasm. Japanese Journal of Cancer Research, 89(9), 933–939.PubMed Konno, H., Arai, T., Tanaka, T., et al. (1998). Antitumor effect of a neutralizing antibody to vascular endothelial growth factor on liver metastasis of endocrine neoplasm. Japanese Journal of Cancer Research, 89(9), 933–939.PubMed
50.
go back to reference Yao, J. C., Phan, A., Hoff, P. M., 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., 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
51.
go back to reference Yuan, R., Kay, A., Berg, W. J., et al. (2009). Targeting tumorigenesis: Development and use of mTOR inhibitors in cancer therapy. J Hematol Oncol, 2, 45.PubMedCrossRef Yuan, R., Kay, A., Berg, W. J., et al. (2009). Targeting tumorigenesis: Development and use of mTOR inhibitors in cancer therapy. J Hematol Oncol, 2, 45.PubMedCrossRef
52.
go back to reference von Wichert, G., Jehle, P. M., Hoeflich, A., et al. (2000). Insulin-like growth factor-I is an autocrine regulator of chromogranin A secretion and growth in human neuroendocrine tumor cells. Cancer Research, 60(16), 4573–4581. von Wichert, G., Jehle, P. M., Hoeflich, A., et al. (2000). Insulin-like growth factor-I is an autocrine regulator of chromogranin A secretion and growth in human neuroendocrine tumor cells. Cancer Research, 60(16), 4573–4581.
53.
go back to reference Moreno, A., Akcakanat, A., Munsell, M. F., et al. (2008). Antitumor activity of rapamycin and octreotide as single agents or in combination in neuroendocrine tumors. Endocrine-Related Cancer, 15(1), 257–266.PubMedCrossRef Moreno, A., Akcakanat, A., Munsell, M. F., et al. (2008). Antitumor activity of rapamycin and octreotide as single agents or in combination in neuroendocrine tumors. Endocrine-Related Cancer, 15(1), 257–266.PubMedCrossRef
54.
go back to reference Yao, J. C., Phan, A. T., Chang, D. Z., 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., 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
55.
go back to reference Yao, J. C., Lombard-Bohas, C., Baudin, E., 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., 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
56.
go back to reference Behr, T. M., Memtsoudis, S., Sharkey, R. M., et al. (1998). Experimental studies on the role of antibody fragments in cancer radio-immunotherapy: Influence of radiation dose and dose rate on toxicity and anti-tumor efficacy. International Journal of Cancer, 77(5), 787–795.CrossRef Behr, T. M., Memtsoudis, S., Sharkey, R. M., et al. (1998). Experimental studies on the role of antibody fragments in cancer radio-immunotherapy: Influence of radiation dose and dose rate on toxicity and anti-tumor efficacy. International Journal of Cancer, 77(5), 787–795.CrossRef
57.
go back to reference Krenning, E. P., Bakker, W. H., Breeman, W. A., et al. (1989). Localisation of endocrine-related tumours with radioiodinated analogue of somatostatin. Lancet, 1(8632), 242–244.PubMedCrossRef Krenning, E. P., Bakker, W. H., Breeman, W. A., et al. (1989). Localisation of endocrine-related tumours with radioiodinated analogue of somatostatin. Lancet, 1(8632), 242–244.PubMedCrossRef
58.
go back to reference Waldherr, C., Pless, M., Maecke, H. R., et al. (2002). Tumor response and clinical benefit in neuroendocrine tumors after 7.4 GBq (90)Y-DOTATOC. Journal of Nuclear Medicine, 43(5), 610–616.PubMed Waldherr, C., Pless, M., Maecke, H. R., et al. (2002). Tumor response and clinical benefit in neuroendocrine tumors after 7.4 GBq (90)Y-DOTATOC. Journal of Nuclear Medicine, 43(5), 610–616.PubMed
59.
go back to reference Appetecchia, M., & Baldelli, R. (2010). Somatostatin analogues in the treatment of gastroenteropancreatic neuroendocrine tumours, current aspects and new perspectives. Journal of Experimental & Clinical Cancer Research, 29, 19.CrossRef Appetecchia, M., & Baldelli, R. (2010). Somatostatin analogues in the treatment of gastroenteropancreatic neuroendocrine tumours, current aspects and new perspectives. Journal of Experimental & Clinical Cancer Research, 29, 19.CrossRef
60.
go back to reference Heppeler, A., Froidevaux, S., Eberle, A. N., et al. (2000). Receptor targeting for tumor localisation and therapy with radiopeptides. Current Medicinal Chemistry, 7(9), 971–994.PubMed Heppeler, A., Froidevaux, S., Eberle, A. N., et al. (2000). Receptor targeting for tumor localisation and therapy with radiopeptides. Current Medicinal Chemistry, 7(9), 971–994.PubMed
61.
go back to reference Reubi, J. C. (2003). Peptide receptors as molecular targets for cancer diagnosis and therapy. Endocrine Reviews, 24(4), 389–427.PubMedCrossRef Reubi, J. C. (2003). Peptide receptors as molecular targets for cancer diagnosis and therapy. Endocrine Reviews, 24(4), 389–427.PubMedCrossRef
62.
go back to reference Valkema, R., De Jong, M., Bakker, W. H., et al. (2002). Phase I study of peptide receptor radionuclide therapy with [In-DTPA]octreotide: The Rotterdam experience. Seminars in Nuclear Medicine, 32(2), 110–122.PubMedCrossRef Valkema, R., De Jong, M., Bakker, W. H., et al. (2002). Phase I study of peptide receptor radionuclide therapy with [In-DTPA]octreotide: The Rotterdam experience. Seminars in Nuclear Medicine, 32(2), 110–122.PubMedCrossRef
63.
go back to reference Henze, M., Schuhmacher, J., Hipp, P., et al. (2001). PET imaging of somatostatin receptors using [68GA]DOTA-D-Phe1-Tyr3-octreotide: First results in patients with meningiomas. Journal of Nuclear Medicine, 42(7), 1053–1056.PubMed Henze, M., Schuhmacher, J., Hipp, P., et al. (2001). PET imaging of somatostatin receptors using [68GA]DOTA-D-Phe1-Tyr3-octreotide: First results in patients with meningiomas. Journal of Nuclear Medicine, 42(7), 1053–1056.PubMed
64.
go back to reference Kwekkeboom, D. J., de Herder, W. W., Kam, B. L., et al. (2008). Treatment with the radiolabeled somatostatin analog [177 Lu-DOTA 0, Tyr3]octreotate: Toxicity, efficacy, and survival. Journal of Clinical Oncology, 26(13), 2124–2130.PubMedCrossRef Kwekkeboom, D. J., de Herder, W. W., Kam, B. L., et al. (2008). Treatment with the radiolabeled somatostatin analog [177 Lu-DOTA 0, Tyr3]octreotate: Toxicity, efficacy, and survival. Journal of Clinical Oncology, 26(13), 2124–2130.PubMedCrossRef
65.
go back to reference Teunissen, J. J., Kwekkeboom, D. J., & Krenning, E. P. (2004). Quality of life in patients with gastroenteropancreatic tumors treated with [177Lu-DOTA0, Tyr3]octreotate. Journal of Clinical Oncology, 22(13), 2724–2729.PubMedCrossRef Teunissen, J. J., Kwekkeboom, D. J., & Krenning, E. P. (2004). Quality of life in patients with gastroenteropancreatic tumors treated with [177Lu-DOTA0, Tyr3]octreotate. Journal of Clinical Oncology, 22(13), 2724–2729.PubMedCrossRef
66.
go back to reference van Essen, M., Krenning, E. P., Kam, B. L., et al. (2008). Report on short-term side effects of treatments with 177Lu-octreotate in combination with capecitabine in seven patients with gastroenteropancreatic neuroendocrine tumours. European Journal of Nuclear Medicine and Molecular Imaging, 35(4), 743–748.PubMedCrossRef van Essen, M., Krenning, E. P., Kam, B. L., et al. (2008). Report on short-term side effects of treatments with 177Lu-octreotate in combination with capecitabine in seven patients with gastroenteropancreatic neuroendocrine tumours. European Journal of Nuclear Medicine and Molecular Imaging, 35(4), 743–748.PubMedCrossRef
67.
go back to reference Arnold, R., Benning, R., Neuhaus, C., et al. (1993). Gastroenteropancreatic endocrine tumours: Effect of Sandostatin® on tumour growth. Digestion, 54(Suppl. 1), 72.PubMedCrossRef Arnold, R., Benning, R., Neuhaus, C., et al. (1993). Gastroenteropancreatic endocrine tumours: Effect of Sandostatin® on tumour growth. Digestion, 54(Suppl. 1), 72.PubMedCrossRef
68.
go back to reference Eriksson, B., Renstrup, J., Imam, H., et al. (1997). High-dose treatment with lanreotide of patients with advanced neuroendocrine gastrointestinal tumors: Clinical and biological effects. Annals of Oncology, 8(10), 1041–1044.PubMedCrossRef Eriksson, B., Renstrup, J., Imam, H., et al. (1997). High-dose treatment with lanreotide of patients with advanced neuroendocrine gastrointestinal tumors: Clinical and biological effects. Annals of Oncology, 8(10), 1041–1044.PubMedCrossRef
69.
go back to reference Ricci, S., Antonuzzo, A., Galli, L., et al. (2000). Long-acting depot lanreotide in the treatment of patients with advanced neuroendocrine tumors. American Journal of Clinical Oncology, 23(4), 412–415.PubMedCrossRef Ricci, S., Antonuzzo, A., Galli, L., et al. (2000). Long-acting depot lanreotide in the treatment of patients with advanced neuroendocrine tumors. American Journal of Clinical Oncology, 23(4), 412–415.PubMedCrossRef
Metadata
Title
Somatostatin analogs for the treatment of neuroendocrine tumors
Authors
Michael D. Culler
Kjell Öberg
Rudolf Arnold
Eric P. Krenning
Isabel Sevilla
José Ángel Díaz
Publication date
01-03-2011
Publisher
Springer US
Published in
Cancer and Metastasis Reviews / Issue Special Issue 1/2011
Print ISSN: 0167-7659
Electronic ISSN: 1573-7233
DOI
https://doi.org/10.1007/s10555-011-9293-0

Other articles of this Special Issue 1/2011

Cancer and Metastasis Reviews 1/2011 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