Skip to main content
Top
Published in: Drugs 1/2019

Open Access 01-01-2019 | Review Article

Targeted Systemic Treatment of Neuroendocrine Tumors: Current Options and Future Perspectives

Authors: Aura D. Herrera-Martínez, Johannes Hofland, Leo J. Hofland, Tessa Brabander, Ferry A. L. M. Eskens, María A. Gálvez Moreno, Raúl M. Luque, Justo P. Castaño, Wouter W. de Herder, Richard A. Feelders

Published in: Drugs | Issue 1/2019

Login to get access

Abstract

Neuroendocrine tumors (NETs) originate from the neuroendocrine cell system in the bronchial and gastrointestinal tract and can produce hormones leading to distinct clinical syndromes. Systemic treatment of patients with unresectable NETs aims to control symptoms related to hormonal overproduction and tumor growth. In the last decades prognosis has improved as a result of increased detection of early stage disease and the introduction of somatostatin analogs (SSAs) as well as several new therapeutic options. SSAs are the first-line medical treatment of NETs and can control hormonal production and tumor growth. The development of next-generation multireceptor targeted and radiolabelled somatostatin analogs, as well as target-directed therapies (as second-line treatment options) further improve progression-free survival in NET patients. To date, however, a significant prolongation of overall survival with systemic treatment in NET has not been convincingly demonstrated. Several new medical options and treatment combinations will become available in the upcoming years, and although preliminary results of preclinical and clinical trials are encouraging, large, preferrably randomized clinical studies are required to provide definitive evidence of their effect on survival and symptom control.
Literature
1.
go back to reference Ameri P, Ferone D. Diffuse endocrine system, neuroendocrine tumors and immunity: what’s new? Neuroendocrinology. 2012;95(4):267–76.PubMed Ameri P, Ferone D. Diffuse endocrine system, neuroendocrine tumors and immunity: what’s new? Neuroendocrinology. 2012;95(4):267–76.PubMed
2.
go back to reference Rosai J. The origin of neuroendocrine tumors and the neural crest saga. Mod Pathol. 2011;24(Suppl 2):S53–7.PubMed Rosai J. The origin of neuroendocrine tumors and the neural crest saga. Mod Pathol. 2011;24(Suppl 2):S53–7.PubMed
3.
go back to reference Fraenkel M, et al. Incidence of gastroenteropancreatic neuroendocrine tumours: a systematic review of the literature. Endocr Relat Cancer. 2014;21(3):R153–63.PubMed Fraenkel M, et al. Incidence of gastroenteropancreatic neuroendocrine tumours: a systematic review of the literature. Endocr Relat Cancer. 2014;21(3):R153–63.PubMed
4.
go back to reference Kunz PL. Carcinoid and neuroendocrine tumors: building on success. J Clin Oncol. 2015;33(16):1855–63.PubMed Kunz PL. Carcinoid and neuroendocrine tumors: building on success. J Clin Oncol. 2015;33(16):1855–63.PubMed
5.
go back to reference Kim JY, Hong SM, Ro JY. Recent updates on grading and classification of neuroendocrine tumors. Ann Diagn Pathol. 2017;29:11–6.PubMed Kim JY, Hong SM, Ro JY. Recent updates on grading and classification of neuroendocrine tumors. Ann Diagn Pathol. 2017;29:11–6.PubMed
6.
go back to reference Dasari A, et al. Trends in the incidence, prevalence, and survival outcomes in patients with neuroendocrine tumors in the United States. JAMA Oncol. 2017;3(10):1335–42.PubMedCentralPubMed Dasari A, et al. Trends in the incidence, prevalence, and survival outcomes in patients with neuroendocrine tumors in the United States. JAMA Oncol. 2017;3(10):1335–42.PubMedCentralPubMed
7.
go back to reference Hofland J, et al. Recent developments in the diagnosis and therapy of well-differentiated neuroendocrine tumours. Neth J Med. 2018;76(3):100–8.PubMed Hofland J, et al. Recent developments in the diagnosis and therapy of well-differentiated neuroendocrine tumours. Neth J Med. 2018;76(3):100–8.PubMed
8.
go back to reference Colao A, Faggiano A, Pivonello R. Somatostatin analogues: treatment of pituitary and neuroendocrine tumors. Prog Brain Res. 2010;182:281–94.PubMed Colao A, Faggiano A, Pivonello R. Somatostatin analogues: treatment of pituitary and neuroendocrine tumors. Prog Brain Res. 2010;182:281–94.PubMed
9.
go back to reference Baldelli R, et al. Somatostatin analogs therapy in gastroenteropancreatic neuroendocrine tumors: current aspects and new perspectives. Front Endocrinol (Lausanne). 2014;5:7. Baldelli R, et al. Somatostatin analogs therapy in gastroenteropancreatic neuroendocrine tumors: current aspects and new perspectives. Front Endocrinol (Lausanne). 2014;5:7.
10.
go back to reference Cakir M, Dworakowska D, Grossman A. Somatostatin receptor biology in neuroendocrine and pituitary tumours: part 2–clinical implications. J Cell Mol Med. 2010;14(11):2585–91.PubMedCentralPubMed Cakir M, Dworakowska D, Grossman A. Somatostatin receptor biology in neuroendocrine and pituitary tumours: part 2–clinical implications. J Cell Mol Med. 2010;14(11):2585–91.PubMedCentralPubMed
11.
go back to reference Falconi M, et al. ENETS consensus guidelines update for the management of patients with functional pancreatic neuroendocrine tumors and non-functional pancreatic neuroendocrine tumors. Neuroendocrinology. 2016;103(2):153–71.PubMed Falconi M, et al. ENETS consensus guidelines update for the management of patients with functional pancreatic neuroendocrine tumors and non-functional pancreatic neuroendocrine tumors. Neuroendocrinology. 2016;103(2):153–71.PubMed
12.
go back to reference Veenstra MJ, et al. Targeting the somatostatin receptor in pituitary and neuroendocrine tumors. Expert Opin Ther Targets. 2013;17(11):1329–43.PubMed Veenstra MJ, et al. Targeting the somatostatin receptor in pituitary and neuroendocrine tumors. Expert Opin Ther Targets. 2013;17(11):1329–43.PubMed
13.
go back to reference van der Zwan WA, et al. GEPNETs update: radionuclide therapy in neuroendocrine tumors. Eur J Endocrinol. 2015;172(1):R1–8.PubMed van der Zwan WA, et al. GEPNETs update: radionuclide therapy in neuroendocrine tumors. Eur J Endocrinol. 2015;172(1):R1–8.PubMed
14.
go back to reference Raymond E, et al. Therapy innovations: tyrosine kinase inhibitors for the treatment of pancreatic neuroendocrine tumors. Cancer Metastasis Rev. 2011;30(Suppl 1):19–26.PubMed Raymond E, et al. Therapy innovations: tyrosine kinase inhibitors for the treatment of pancreatic neuroendocrine tumors. Cancer Metastasis Rev. 2011;30(Suppl 1):19–26.PubMed
15.
go back to reference Chan J, Kulke M. Targeting the mTOR signaling pathway in neuroendocrine tumors. Curr Treat Options Oncol. 2014;15(3):365–79.PubMedCentralPubMed Chan J, Kulke M. Targeting the mTOR signaling pathway in neuroendocrine tumors. Curr Treat Options Oncol. 2014;15(3):365–79.PubMedCentralPubMed
16.
go back to reference Garcia-Carbonero R, et al. ENETS consensus guidelines for the standards of care in neuroendocrine neoplasms. Systemic therapy 2: chemotherapy. Neuroendocrinology. 2017;105(3):281–94.PubMed Garcia-Carbonero R, et al. ENETS consensus guidelines for the standards of care in neuroendocrine neoplasms. Systemic therapy 2: chemotherapy. Neuroendocrinology. 2017;105(3):281–94.PubMed
17.
go back to reference Pavel M, et al. Telotristat ethyl in carcinoid syndrome: safety and efficacy in the TELECAST phase III trial. Endocr Relat Cancer. 2018;25(3):309–22.PubMedCentralPubMed Pavel M, et al. Telotristat ethyl in carcinoid syndrome: safety and efficacy in the TELECAST phase III trial. Endocr Relat Cancer. 2018;25(3):309–22.PubMedCentralPubMed
18.
go back to reference Pavel M, et al. Telotristat etiprate for carcinoid syndrome: a single-arm, multicenter trial. J Clin Endocrinol Metab. 2015;100(4):1511–9.PubMed Pavel M, et al. Telotristat etiprate for carcinoid syndrome: a single-arm, multicenter trial. J Clin Endocrinol Metab. 2015;100(4):1511–9.PubMed
19.
go back to reference Halperin DM, et al. Frequency of carcinoid syndrome at neuroendocrine tumour diagnosis: a population-based study. Lancet Oncol. 2017;18(4):525–34.PubMedCentralPubMed Halperin DM, et al. Frequency of carcinoid syndrome at neuroendocrine tumour diagnosis: a population-based study. Lancet Oncol. 2017;18(4):525–34.PubMedCentralPubMed
20.
go back to reference Zandee WT, de Herder WW, Jann H. Incidence and prognosis of carcinoid syndrome: hormones or tumour burden? Lancet Oncol. 2017;18(6):e299.PubMed Zandee WT, de Herder WW, Jann H. Incidence and prognosis of carcinoid syndrome: hormones or tumour burden? Lancet Oncol. 2017;18(6):e299.PubMed
21.
go back to reference Mougey AM, Adler DG. Neuroendocrine tumors: review and clinical update. Hosp Physician. 2007;51:12–20. Mougey AM, Adler DG. Neuroendocrine tumors: review and clinical update. Hosp Physician. 2007;51:12–20.
22.
go back to reference Niederle B, et al. ENETS consensus guidelines update for neuroendocrine neoplasms of the jejunum and ileum. Neuroendocrinology. 2016;103(2):125–38.PubMed Niederle B, et al. ENETS consensus guidelines update for neuroendocrine neoplasms of the jejunum and ileum. Neuroendocrinology. 2016;103(2):125–38.PubMed
23.
go back to reference Pavel M, et al. ENETS consensus guidelines update for the management of distant metastatic disease of intestinal, pancreatic, bronchial neuroendocrine neoplasms (NEN) and NEN of unknown primary site. Neuroendocrinology. 2016;103(2):172–85.PubMed Pavel M, et al. ENETS consensus guidelines update for the management of distant metastatic disease of intestinal, pancreatic, bronchial neuroendocrine neoplasms (NEN) and NEN of unknown primary site. Neuroendocrinology. 2016;103(2):172–85.PubMed
24.
go back to reference Ito T, Lee L, Jensen RT. Carcinoid-syndrome: recent advances, current status and controversies. Curr Opin Endocrinol Diabetes Obes. 2018;25(1):22–35.PubMedCentralPubMed Ito T, Lee L, Jensen RT. Carcinoid-syndrome: recent advances, current status and controversies. Curr Opin Endocrinol Diabetes Obes. 2018;25(1):22–35.PubMedCentralPubMed
25.
go back to reference Davis GR, et al. Effect of somatostatin infusion on jejunal water and electrolyte transport in a patient with secretory diarrhea due to malignant carcinoid syndrome. Gastroenterology. 1980;78(2):346–9.PubMed Davis GR, et al. Effect of somatostatin infusion on jejunal water and electrolyte transport in a patient with secretory diarrhea due to malignant carcinoid syndrome. Gastroenterology. 1980;78(2):346–9.PubMed
26.
go back to reference Dharmsathaphorn K, et al. Somatostatin inhibits diarrhea in the carcinoid syndrome. Ann Intern Med. 1980;92(1):68–9.PubMed Dharmsathaphorn K, et al. Somatostatin inhibits diarrhea in the carcinoid syndrome. Ann Intern Med. 1980;92(1):68–9.PubMed
27.
go back to reference Kvols LK, et al. Treatment of the malignant carcinoid syndrome. Evaluation of a long-acting somatostatin analogue. N Engl J Med. 1986;315(11):663–6.PubMed Kvols LK, et al. Treatment of the malignant carcinoid syndrome. Evaluation of a long-acting somatostatin analogue. N Engl J Med. 1986;315(11):663–6.PubMed
28.
go back to reference Rubin J, et al. Octreotide acetate long-acting formulation versus open-label subcutaneous octreotide acetate in malignant carcinoid syndrome. J Clin Oncol. 1999;17(2):600–6.PubMed Rubin J, et al. Octreotide acetate long-acting formulation versus open-label subcutaneous octreotide acetate in malignant carcinoid syndrome. J Clin Oncol. 1999;17(2):600–6.PubMed
29.
go back to reference O’Toole D, et al. Treatment of carcinoid syndrome: a prospective crossover evaluation of lanreotide versus octreotide in terms of efficacy, patient acceptability, and tolerance. Cancer. 2000;88(4):770–6.PubMed O’Toole D, et al. Treatment of carcinoid syndrome: a prospective crossover evaluation of lanreotide versus octreotide in terms of efficacy, patient acceptability, and tolerance. Cancer. 2000;88(4):770–6.PubMed
30.
go back to reference Ruszniewski P, et al. Rapid and sustained relief from the symptoms of carcinoid syndrome: results from an open 6-month study of the 28-day prolonged-release formulation of lanreotide. Neuroendocrinology. 2004;80(4):244–51.PubMed Ruszniewski P, et al. Rapid and sustained relief from the symptoms of carcinoid syndrome: results from an open 6-month study of the 28-day prolonged-release formulation of lanreotide. Neuroendocrinology. 2004;80(4):244–51.PubMed
31.
go back to reference Plockinger U, Wiedenmann B. Neuroendocrine tumors. Biotherapy. Best Pract Res Clin Endocrinol Metab. 2007;21(1):145–62.PubMed Plockinger U, Wiedenmann B. Neuroendocrine tumors. Biotherapy. Best Pract Res Clin Endocrinol Metab. 2007;21(1):145–62.PubMed
32.
go back to reference Janson ET, Oberg K. Long-term management of the carcinoid syndrome. Treatment with octreotide alone and in combination with alpha-interferon. Acta Oncol. 1993;32(2):225–9.PubMed Janson ET, Oberg K. Long-term management of the carcinoid syndrome. Treatment with octreotide alone and in combination with alpha-interferon. Acta Oncol. 1993;32(2):225–9.PubMed
33.
go back to reference Hofland LJ, Lamberts SW. The pathophysiological consequences of somatostatin receptor internalization and resistance. Endocr Rev. 2003;24(1):28–47.PubMed Hofland LJ, Lamberts SW. The pathophysiological consequences of somatostatin receptor internalization and resistance. Endocr Rev. 2003;24(1):28–47.PubMed
34.
go back to reference Strosberg JR, et al. Clinical benefits of above-standard dose of octreotide LAR in patients with neuroendocrine tumors for control of carcinoid syndrome symptoms: a multicenter retrospective chart review study. Oncologist. 2014;19(9):930–6.PubMedCentralPubMed Strosberg JR, et al. Clinical benefits of above-standard dose of octreotide LAR in patients with neuroendocrine tumors for control of carcinoid syndrome symptoms: a multicenter retrospective chart review study. Oncologist. 2014;19(9):930–6.PubMedCentralPubMed
35.
go back to reference Ferolla P, et al. Shortened interval of long-acting octreotide administration is effective in patients with well-differentiated neuroendocrine carcinomas in progression on standard doses. J Endocrinol Invest. 2012;35(3):326–31.PubMed Ferolla P, et al. Shortened interval of long-acting octreotide administration is effective in patients with well-differentiated neuroendocrine carcinomas in progression on standard doses. J Endocrinol Invest. 2012;35(3):326–31.PubMed
36.
go back to reference Wolin EM. The expanding role of somatostatin analogs in the management of neuroendocrine tumors. Gastrointest Cancer Res. 2012;5(5):161–8.PubMedCentralPubMed Wolin EM. The expanding role of somatostatin analogs in the management of neuroendocrine tumors. Gastrointest Cancer Res. 2012;5(5):161–8.PubMedCentralPubMed
37.
go back to reference Sandostatin LAR® depot (octreotide acetate for injectable suspension) [prescribing information]. East Hanover, NJ: Novartis Pharmaceutical Corporation; 2011. Sandostatin LAR® depot (octreotide acetate for injectable suspension) [prescribing information]. East Hanover, NJ: Novartis Pharmaceutical Corporation; 2011.
38.
go back to reference Teunissen JJ, Kwekkeboom DJ, Krenning EP. Quality of life in patients with gastroenteropancreatic tumors treated with [177Lu-DOTA0, Tyr3]octreotate. J Clin Oncol. 2004;22(13):2724–9.PubMed Teunissen JJ, Kwekkeboom DJ, Krenning EP. Quality of life in patients with gastroenteropancreatic tumors treated with [177Lu-DOTA0, Tyr3]octreotate. J Clin Oncol. 2004;22(13):2724–9.PubMed
39.
go back to reference Waldherr C, et al. Tumor response and clinical benefit in neuroendocrine tumors after 7.4 GBq (90)Y-DOTATOC. J Nucl Med. 2002;43(5):610–6. Waldherr C, et al. Tumor response and clinical benefit in neuroendocrine tumors after 7.4 GBq (90)Y-DOTATOC. J Nucl Med. 2002;43(5):610–6.
40.
go back to reference Kiesewetter B, Raderer M. Ondansetron for diarrhea associated with neuroendocrine tumors. N Engl J Med. 2013;368(20):1947–8.PubMed Kiesewetter B, Raderer M. Ondansetron for diarrhea associated with neuroendocrine tumors. N Engl J Med. 2013;368(20):1947–8.PubMed
41.
go back to reference Mota JM, Sousa LG, Riechelmann RP. Complications from carcinoid syndrome: review of the current evidence. Ecancermedicalscience. 2016;10:662.PubMedCentralPubMed Mota JM, Sousa LG, Riechelmann RP. Complications from carcinoid syndrome: review of the current evidence. Ecancermedicalscience. 2016;10:662.PubMedCentralPubMed
42.
go back to reference Kvols LK, et al. Pasireotide (SOM230) shows efficacy and tolerability in the treatment of patients with advanced neuroendocrine tumors refractory or resistant to octreotide LAR: results from a phase II study. Endocr Relat Cancer. 2012;19(5):657–66.PubMed Kvols LK, et al. Pasireotide (SOM230) shows efficacy and tolerability in the treatment of patients with advanced neuroendocrine tumors refractory or resistant to octreotide LAR: results from a phase II study. Endocr Relat Cancer. 2012;19(5):657–66.PubMed
43.
go back to reference Wolin EM, et al. Phase III study of pasireotide long-acting release in patients with metastatic neuroendocrine tumors and carcinoid symptoms refractory to available somatostatin analogues. Drug Des Dev Ther. 2015;9:5075–86. Wolin EM, et al. Phase III study of pasireotide long-acting release in patients with metastatic neuroendocrine tumors and carcinoid symptoms refractory to available somatostatin analogues. Drug Des Dev Ther. 2015;9:5075–86.
44.
go back to reference Frank M, et al. Combination therapy with octreotide and alpha-interferon: effect on tumor growth in metastatic endocrine gastroenteropancreatic tumors. Am J Gastroenterol. 1999;94(5):1381–7.PubMed Frank M, et al. Combination therapy with octreotide and alpha-interferon: effect on tumor growth in metastatic endocrine gastroenteropancreatic tumors. Am J Gastroenterol. 1999;94(5):1381–7.PubMed
45.
go back to reference Kulke MH, et al. Telotristat ethyl, a tryptophan hydroxylase inhibitor for the treatment of carcinoid syndrome. J Clin Oncol. 2017;35(1):14–23.PubMed Kulke MH, et al. Telotristat ethyl, a tryptophan hydroxylase inhibitor for the treatment of carcinoid syndrome. J Clin Oncol. 2017;35(1):14–23.PubMed
46.
go back to reference Kulke MH, et al. A randomized, open-label, phase II study of everolimus in combination with pasireotide LAR or everolimus alone in advanced, well-differentiated, progressive pancreatic neuroendocrine tumors: COOPERATE-2 trial. Ann Oncol. 2017;28(6):1309–15.PubMedPubMedCentral Kulke MH, et al. A randomized, open-label, phase II study of everolimus in combination with pasireotide LAR or everolimus alone in advanced, well-differentiated, progressive pancreatic neuroendocrine tumors: COOPERATE-2 trial. Ann Oncol. 2017;28(6):1309–15.PubMedPubMedCentral
47.
go back to reference Kulke MH, et al. Telotristat etiprate, a novel serotonin synthesis inhibitor, in patients with carcinoid syndrome and diarrhea not adequately controlled by octreotide. Endocr Relat Cancer. 2014;21(5):705–14.PubMedCentralPubMed Kulke MH, et al. Telotristat etiprate, a novel serotonin synthesis inhibitor, in patients with carcinoid syndrome and diarrhea not adequately controlled by octreotide. Endocr Relat Cancer. 2014;21(5):705–14.PubMedCentralPubMed
48.
go back to reference Hutcheson JD, et al. Serotonin receptors and heart valve disease–it was meant 2B. Pharmacol Ther. 2011;132(2):146–57.PubMedCentralPubMed Hutcheson JD, et al. Serotonin receptors and heart valve disease–it was meant 2B. Pharmacol Ther. 2011;132(2):146–57.PubMedCentralPubMed
49.
go back to reference Blazevic A, et al. Small intestinal neuroendocrine tumours and fibrosis: an entangled conundrum. Endocr Relat Cancer. 2018;25(3):R115–30.PubMed Blazevic A, et al. Small intestinal neuroendocrine tumours and fibrosis: an entangled conundrum. Endocr Relat Cancer. 2018;25(3):R115–30.PubMed
50.
go back to reference Grozinsky-Glasberg S, Grossman AB, Gross DJ. Carcinoid heart disease: from pathophysiology to treatment—’something in the way it moves’. Neuroendocrinology. 2015;101(4):263–73.PubMed Grozinsky-Glasberg S, Grossman AB, Gross DJ. Carcinoid heart disease: from pathophysiology to treatment—’something in the way it moves’. Neuroendocrinology. 2015;101(4):263–73.PubMed
51.
go back to reference Hassan SA, et al. Carcinoid heart disease. Heart. 2017;103(19):1488–95.PubMed Hassan SA, et al. Carcinoid heart disease. Heart. 2017;103(19):1488–95.PubMed
52.
go back to reference Davar J, et al. Diagnosing and managing carcinoid heart disease in patients with neuroendocrine tumors: an expert statement. J Am Coll Cardiol. 2017;69(10):1288–304.PubMed Davar J, et al. Diagnosing and managing carcinoid heart disease in patients with neuroendocrine tumors: an expert statement. J Am Coll Cardiol. 2017;69(10):1288–304.PubMed
53.
go back to reference Modlin IM, et al. Therapeutic options for gastrointestinal carcinoids. Clin Gastroenterol Hepatol. 2006;4(5):526–47.PubMed Modlin IM, et al. Therapeutic options for gastrointestinal carcinoids. Clin Gastroenterol Hepatol. 2006;4(5):526–47.PubMed
54.
go back to reference Moller JE, et al. Factors associated with progression of carcinoid heart disease. N Engl J Med. 2003;348(11):1005–15.PubMed Moller JE, et al. Factors associated with progression of carcinoid heart disease. N Engl J Med. 2003;348(11):1005–15.PubMed
55.
56.
go back to reference Kaltsas G, et al. ENETS consensus guidelines for the standards of care in neuroendocrine tumors: pre- and perioperative therapy in patients with neuroendocrine tumors. Neuroendocrinology. 2017;105(3):245–54.PubMed Kaltsas G, et al. ENETS consensus guidelines for the standards of care in neuroendocrine tumors: pre- and perioperative therapy in patients with neuroendocrine tumors. Neuroendocrinology. 2017;105(3):245–54.PubMed
57.
go back to reference Castillo JG, et al. Management of patients undergoing multivalvular surgery for carcinoid heart disease: the role of the anaesthetist. Br J Anaesth. 2008;101(5):618–26.PubMed Castillo JG, et al. Management of patients undergoing multivalvular surgery for carcinoid heart disease: the role of the anaesthetist. Br J Anaesth. 2008;101(5):618–26.PubMed
58.
go back to reference Goode PN, et al. Diazoxide in the management of patients with insulinoma. World J Surg. 1986;10(4):586–92.PubMed Goode PN, et al. Diazoxide in the management of patients with insulinoma. World J Surg. 1986;10(4):586–92.PubMed
59.
go back to reference Gill GV, Rauf O, MacFarlane IA. Diazoxide treatment for insulinoma: a national UK survey. Postgrad Med J. 1997;73(864):640–1.PubMedCentralPubMed Gill GV, Rauf O, MacFarlane IA. Diazoxide treatment for insulinoma: a national UK survey. Postgrad Med J. 1997;73(864):640–1.PubMedCentralPubMed
60.
go back to reference de Herder WW, et al. New therapeutic options for metastatic malignant insulinomas. Clin Endocrinol (Oxf). 2011;75(3):277–84. de Herder WW, et al. New therapeutic options for metastatic malignant insulinomas. Clin Endocrinol (Oxf). 2011;75(3):277–84.
61.
go back to reference Baudin E, et al. Malignant insulinoma: recommendations for characterisation and treatment. Ann Endocrinol (Paris). 2013;74(5–6):523–33. Baudin E, et al. Malignant insulinoma: recommendations for characterisation and treatment. Ann Endocrinol (Paris). 2013;74(5–6):523–33.
62.
go back to reference Altszuler N, Moraru E, Hampshire J. On the mechanism of diazoxide-induced hyperglycemia. Diabetes. 1977;26(10):931–5.PubMed Altszuler N, Moraru E, Hampshire J. On the mechanism of diazoxide-induced hyperglycemia. Diabetes. 1977;26(10):931–5.PubMed
63.
go back to reference Matej A, Bujwid H, Wronski J. Glycemic control in patients with insulinoma. Hormones (Athens). 2016;15(4):489–99. Matej A, Bujwid H, Wronski J. Glycemic control in patients with insulinoma. Hormones (Athens). 2016;15(4):489–99.
64.
go back to reference Oberg K. Pancreatic endocrine tumors. Semin Oncol. 2010;37(6):594–618.PubMed Oberg K. Pancreatic endocrine tumors. Semin Oncol. 2010;37(6):594–618.PubMed
65.
go back to reference Ferrer-Garcia JC, et al. Management of malignant insulinoma. Clin Transl Oncol. 2013;15(9):725–31.PubMed Ferrer-Garcia JC, et al. Management of malignant insulinoma. Clin Transl Oncol. 2013;15(9):725–31.PubMed
66.
go back to reference de Herder WW, et al. Well-differentiated pancreatic tumor/carcinoma: insulinoma. Neuroendocrinology. 2006;84(3):183–8.PubMed de Herder WW, et al. Well-differentiated pancreatic tumor/carcinoma: insulinoma. Neuroendocrinology. 2006;84(3):183–8.PubMed
67.
go back to reference Singh V, et al. Characterization of somatostatin receptor subtype-specific regulation of insulin and glucagon secretion: an in vitro study on isolated human pancreatic islets. J Clin Endocrinol Metab. 2007;92(2):673–80.PubMed Singh V, et al. Characterization of somatostatin receptor subtype-specific regulation of insulin and glucagon secretion: an in vitro study on isolated human pancreatic islets. J Clin Endocrinol Metab. 2007;92(2):673–80.PubMed
68.
go back to reference Bruns C, et al. SOM230: a novel somatostatin peptidomimetic with broad somatotropin release inhibiting factor (SRIF) receptor binding and a unique antisecretory profile. Eur J Endocrinol. 2002;146(5):707–16.PubMed Bruns C, et al. SOM230: a novel somatostatin peptidomimetic with broad somatotropin release inhibiting factor (SRIF) receptor binding and a unique antisecretory profile. Eur J Endocrinol. 2002;146(5):707–16.PubMed
69.
go back to reference Hendren NS, et al. Pasireotide for the treatment of refractory hypoglycaemia from malignant insulinoma. Clin Endocrinol (Oxf). 2018;88(2):341–3.PubMed Hendren NS, et al. Pasireotide for the treatment of refractory hypoglycaemia from malignant insulinoma. Clin Endocrinol (Oxf). 2018;88(2):341–3.PubMed
70.
go back to reference Tirosh A, et al. Pasireotide for malignant insulinoma. Hormones (Athens). 2016;15(2):271–6. Tirosh A, et al. Pasireotide for malignant insulinoma. Hormones (Athens). 2016;15(2):271–6.
71.
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.PubMed Kulke MH, Bergsland EK, Yao JC. Glycemic control in patients with insulinoma treated with everolimus. N Engl J Med. 2009;360(2):195–7.PubMed
72.
go back to reference Bourcier ME, et al. Successful control of intractable hypoglycemia using rapamycin in an 86-year-old man with a pancreatic insulin-secreting islet cell tumor and metastases. J Clin Endocrinol Metab. 2009;94(9):3157–62.PubMed Bourcier ME, et al. Successful control of intractable hypoglycemia using rapamycin in an 86-year-old man with a pancreatic insulin-secreting islet cell tumor and metastases. J Clin Endocrinol Metab. 2009;94(9):3157–62.PubMed
73.
go back to reference Fuhrer DK, Kobayashi M, Jiang H. Insulin release and suppression by tacrolimus, rapamycin and cyclosporin A are through regulation of the ATP-sensitive potassium channel. Diabetes Obes Metab. 2001;3(6):393–402.PubMed Fuhrer DK, Kobayashi M, Jiang H. Insulin release and suppression by tacrolimus, rapamycin and cyclosporin A are through regulation of the ATP-sensitive potassium channel. Diabetes Obes Metab. 2001;3(6):393–402.PubMed
74.
go back to reference Di Paolo S, et al. Chronic inhibition of mammalian target of rapamycin signaling downregulates insulin receptor substrates 1 and 2 and AKT activation: a crossroad between cancer and diabetes? J Am Soc Nephrol. 2006;17(8):2236–44.PubMed Di Paolo S, et al. Chronic inhibition of mammalian target of rapamycin signaling downregulates insulin receptor substrates 1 and 2 and AKT activation: a crossroad between cancer and diabetes? J Am Soc Nephrol. 2006;17(8):2236–44.PubMed
75.
go back to reference Fiebrich HB, 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.PubMedCentralPubMed Fiebrich HB, 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.PubMedCentralPubMed
76.
go back to reference van Schaik E, et al. Improved control of severe hypoglycemia in patients with malignant insulinomas by peptide receptor radionuclide therapy. J Clin Endocrinol Metab. 2011;96(11):3381–9.PubMed van Schaik E, et al. Improved control of severe hypoglycemia in patients with malignant insulinomas by peptide receptor radionuclide therapy. J Clin Endocrinol Metab. 2011;96(11):3381–9.PubMed
77.
go back to reference John AM, Schwartz RA. Glucagonoma syndrome: a review and update on treatment. J Eur Acad Dermatol Venereol. 2016;30(12):2016–22.PubMed John AM, Schwartz RA. Glucagonoma syndrome: a review and update on treatment. J Eur Acad Dermatol Venereol. 2016;30(12):2016–22.PubMed
78.
go back to reference Santangelo WC, et al. Somatostatin analog-induced remission of necrolytic migratory erythema without changes in plasma glucagon concentration. Pancreas. 1986;1(5):464–9.PubMed Santangelo WC, et al. Somatostatin analog-induced remission of necrolytic migratory erythema without changes in plasma glucagon concentration. Pancreas. 1986;1(5):464–9.PubMed
79.
go back to reference Schmid HA. Pasireotide (SOM230): development, mechanism of action and potential applications. Mol Cell Endocrinol. 2008;286(1–2):69–74.PubMed Schmid HA. Pasireotide (SOM230): development, mechanism of action and potential applications. Mol Cell Endocrinol. 2008;286(1–2):69–74.PubMed
80.
go back to reference Raymond E, et al. Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. N Engl J Med. 2011;364(6):501–13.PubMed Raymond E, et al. Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. N Engl J Med. 2011;364(6):501–13.PubMed
81.
82.
go back to reference Kwekkeboom DJ, et al. Treatment with the radiolabeled somatostatin analog [177 Lu-DOTA 0, Tyr3]octreotate: toxicity, efficacy, and survival. J Clin Oncol. 2008;26(13):2124–30.PubMed Kwekkeboom DJ, et al. Treatment with the radiolabeled somatostatin analog [177 Lu-DOTA 0, Tyr3]octreotate: toxicity, efficacy, and survival. J Clin Oncol. 2008;26(13):2124–30.PubMed
83.
go back to reference Jensen RT. Gastrinomas: advances in diagnosis and management. Neuroendocrinology. 2004;80(Suppl 1):23–7.PubMed Jensen RT. Gastrinomas: advances in diagnosis and management. Neuroendocrinology. 2004;80(Suppl 1):23–7.PubMed
84.
go back to reference Brandi ML, et al. Guidelines for diagnosis and therapy of MEN type 1 and type 2. J Clin Endocrinol Metab. 2001;86(12):5658–71.PubMed Brandi ML, et al. Guidelines for diagnosis and therapy of MEN type 1 and type 2. J Clin Endocrinol Metab. 2001;86(12):5658–71.PubMed
85.
go back to reference Auernhammer CJ, Goke B. Medical treatment of gastrinomas. Wien Klin Wochenschr. 2007;119(19–20):609–15.PubMed Auernhammer CJ, Goke B. Medical treatment of gastrinomas. Wien Klin Wochenschr. 2007;119(19–20):609–15.PubMed
86.
go back to reference Termanini B, et al. Effect of long-term gastric acid suppressive therapy on serum vitamin B12 levels in patients with Zollinger–Ellison syndrome. Am J Med. 1998;104(5):422–30.PubMed Termanini B, et al. Effect of long-term gastric acid suppressive therapy on serum vitamin B12 levels in patients with Zollinger–Ellison syndrome. Am J Med. 1998;104(5):422–30.PubMed
87.
go back to reference Jensen RT. Consequences of long-term proton pump blockade: insights from studies of patients with gastrinomas. Basic Clin Pharmacol Toxicol. 2006;98(1):4–19.PubMed Jensen RT. Consequences of long-term proton pump blockade: insights from studies of patients with gastrinomas. Basic Clin Pharmacol Toxicol. 2006;98(1):4–19.PubMed
88.
go back to reference Oberg K, et al. Consensus report on the use of somatostatin analogs for the management of neuroendocrine tumors of the gastroenteropancreatic system. Ann Oncol. 2004;15(6):966–73.PubMed Oberg K, et al. Consensus report on the use of somatostatin analogs for the management of neuroendocrine tumors of the gastroenteropancreatic system. Ann Oncol. 2004;15(6):966–73.PubMed
89.
go back to reference Burgess JR, et al. Octreotide improves biochemical, radiologic, and symptomatic indices of gastroenteropancreatic neoplasia in patients with multiple endocrine neoplasia type 1 (MEN-1). Implications for an integrated model of MEN-1 tumorigenesis. Cancer. 1999;86(10):2154–9.PubMed Burgess JR, et al. Octreotide improves biochemical, radiologic, and symptomatic indices of gastroenteropancreatic neoplasia in patients with multiple endocrine neoplasia type 1 (MEN-1). Implications for an integrated model of MEN-1 tumorigenesis. Cancer. 1999;86(10):2154–9.PubMed
90.
go back to reference Ricci S, et al. Long-acting depot lanreotide in the treatment of patients with advanced neuroendocrine tumors. Am J Clin Oncol. 2000;23(4):412–5.PubMed Ricci S, et al. Long-acting depot lanreotide in the treatment of patients with advanced neuroendocrine tumors. Am J Clin Oncol. 2000;23(4):412–5.PubMed
91.
go back to reference Wymenga AN, et al. Efficacy and safety of prolonged-release lanreotide in patients with gastrointestinal neuroendocrine tumors and hormone-related symptoms. J Clin Oncol. 1999;17(4):1111.PubMed Wymenga AN, et al. Efficacy and safety of prolonged-release lanreotide in patients with gastrointestinal neuroendocrine tumors and hormone-related symptoms. J Clin Oncol. 1999;17(4):1111.PubMed
92.
go back to reference Shojamanesh H, et al. Prospective study of the antitumor efficacy of long-term octreotide treatment in patients with progressive metastatic gastrinoma. Cancer. 2002;94(2):331–43.PubMed Shojamanesh H, et al. Prospective study of the antitumor efficacy of long-term octreotide treatment in patients with progressive metastatic gastrinoma. Cancer. 2002;94(2):331–43.PubMed
93.
go back to reference Hiraide S, Ono S, Kato S. Long-term efficacy of S-1 chemotherapy plus administration of octreotide for a patient with metastatic neuroendocrine tumor (Gastrinoma). Case Rep Oncol. 2017;10(2):420–7.PubMedCentralPubMed Hiraide S, Ono S, Kato S. Long-term efficacy of S-1 chemotherapy plus administration of octreotide for a patient with metastatic neuroendocrine tumor (Gastrinoma). Case Rep Oncol. 2017;10(2):420–7.PubMedCentralPubMed
94.
go back to reference Peghini PL, et al. Effect of chronic hypergastrinemia on human enterochromaffin-like cells: insights from patients with sporadic gastrinomas. Gastroenterology. 2002;123(1):68–85.PubMed Peghini PL, et al. Effect of chronic hypergastrinemia on human enterochromaffin-like cells: insights from patients with sporadic gastrinomas. Gastroenterology. 2002;123(1):68–85.PubMed
95.
go back to reference Krenning EP, et al. Somatostatin receptor scintigraphy in carcinoids, gastrinomas and Cushing’s syndrome. Digestion. 1994;55(Suppl 3):54–9.PubMed Krenning EP, et al. Somatostatin receptor scintigraphy in carcinoids, gastrinomas and Cushing’s syndrome. Digestion. 1994;55(Suppl 3):54–9.PubMed
96.
go back to reference O’Dorisio TM, Mekhjian HS, Gaginella TS. Medical therapy of VIPomas. Endocrinol Metab Clin N Am. 1989;18(2):545–56. O’Dorisio TM, Mekhjian HS, Gaginella TS. Medical therapy of VIPomas. Endocrinol Metab Clin N Am. 1989;18(2):545–56.
97.
98.
go back to reference Friesen SR. Update on the diagnosis and treatment of rare neuroendocrine tumors. Surg Clin N Am. 1987;67(2):379–93.PubMed Friesen SR. Update on the diagnosis and treatment of rare neuroendocrine tumors. Surg Clin N Am. 1987;67(2):379–93.PubMed
99.
go back to reference Oberg K, et al. Neuroendocrine gastro-entero-pancreatic tumors: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2012;23(Suppl 7):vii124–30.PubMed Oberg K, et al. Neuroendocrine gastro-entero-pancreatic tumors: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2012;23(Suppl 7):vii124–30.PubMed
100.
go back to reference Eriksson B, et al. High-dose treatment with lanreotide of patients with advanced neuroendocrine gastrointestinal tumors: clinical and biological effects. Ann Oncol. 1997;8(10):1041–4.PubMed Eriksson B, et al. High-dose treatment with lanreotide of patients with advanced neuroendocrine gastrointestinal tumors: clinical and biological effects. Ann Oncol. 1997;8(10):1041–4.PubMed
101.
go back to reference Delbaldo C, et al. Sunitinib in advanced pancreatic neuroendocrine tumors: latest evidence and clinical potential. Ther Adv Med Oncol. 2012;4(1):9–18.PubMedCentralPubMed Delbaldo C, et al. Sunitinib in advanced pancreatic neuroendocrine tumors: latest evidence and clinical potential. Ther Adv Med Oncol. 2012;4(1):9–18.PubMedCentralPubMed
102.
go back to reference Faivre S, et al. Sunitinib in pancreatic neuroendocrine tumors: updated progression-free survival and final overall survival from a phase III randomized study. Ann Oncol. 2017;28(2):339–43.PubMed Faivre S, et al. Sunitinib in pancreatic neuroendocrine tumors: updated progression-free survival and final overall survival from a phase III randomized study. Ann Oncol. 2017;28(2):339–43.PubMed
103.
go back to reference Hubold C, Brabant G. [Ectopic hormone secretion by neuroendocrine tumors] Ektope Hormonsekretion bei neuroendokrinen Tumoren. Internist (Berl). 2012;53(2):145–51. Hubold C, Brabant G. [Ectopic hormone secretion by neuroendocrine tumors] Ektope Hormonsekretion bei neuroendokrinen Tumoren. Internist (Berl). 2012;53(2):145–51.
104.
go back to reference Kamp K, et al. Parathyroid hormone-related peptide (PTHrP) secretion by gastroenteropancreatic neuroendocrine tumors (GEP-NETs): clinical features, diagnosis, management, and follow-up. J Clin Endocrinol Metab. 2014;99(9):3060–9.PubMed Kamp K, et al. Parathyroid hormone-related peptide (PTHrP) secretion by gastroenteropancreatic neuroendocrine tumors (GEP-NETs): clinical features, diagnosis, management, and follow-up. J Clin Endocrinol Metab. 2014;99(9):3060–9.PubMed
105.
go back to reference Herrera-Martinez AD, Padillo Cuenca JC, Bahamondes Opazo R, Barrera Martín A, Rebollo Roman A, Díaz Iglesia C, Gálvez Moreno MA. ACTH producing pancreatic neuroendocrine tumors in multiple endocrine neoplasia type 1. J Pancreas. 2017;02:175–80. Herrera-Martinez AD, Padillo Cuenca JC, Bahamondes Opazo R, Barrera Martín A, Rebollo Roman A, Díaz Iglesia C, Gálvez Moreno MA. ACTH producing pancreatic neuroendocrine tumors in multiple endocrine neoplasia type 1. J Pancreas. 2017;02:175–80.
106.
go back to reference Ilias I, et al. Cushing’s syndrome due to ectopic corticotropin secretion: twenty years’ experience at the National Institutes of Health. J Clin Endocrinol Metab. 2005;90(8):4955–62.PubMed Ilias I, et al. Cushing’s syndrome due to ectopic corticotropin secretion: twenty years’ experience at the National Institutes of Health. J Clin Endocrinol Metab. 2005;90(8):4955–62.PubMed
107.
go back to reference Kamp K, et al. Prevalence and clinical features of the ectopic ACTH syndrome in patients with gastroenteropancreatic and thoracic neuroendocrine tumors. Eur J Endocrinol. 2016;174(3):271–80.PubMed Kamp K, et al. Prevalence and clinical features of the ectopic ACTH syndrome in patients with gastroenteropancreatic and thoracic neuroendocrine tumors. Eur J Endocrinol. 2016;174(3):271–80.PubMed
108.
go back to reference van der Pas R, et al. New developments in the medical treatment of Cushing’s syndrome. Endocr Relat Cancer. 2012;19(6):R205–23.PubMed van der Pas R, et al. New developments in the medical treatment of Cushing’s syndrome. Endocr Relat Cancer. 2012;19(6):R205–23.PubMed
109.
go back to reference Aniszewski JP, et al. Cushing syndrome due to ectopic adrenocorticotropic hormone secretion. World J Surg. 2001;25(7):934–40.PubMed Aniszewski JP, et al. Cushing syndrome due to ectopic adrenocorticotropic hormone secretion. World J Surg. 2001;25(7):934–40.PubMed
110.
go back to reference Lamberts SW, et al. A role of (labeled) somatostatin analogs in the differential diagnosis and treatment of Cushing’s syndrome. J Clin Endocrinol Metab. 1994;78(1):17–9.PubMed Lamberts SW, et al. A role of (labeled) somatostatin analogs in the differential diagnosis and treatment of Cushing’s syndrome. J Clin Endocrinol Metab. 1994;78(1):17–9.PubMed
111.
go back to reference Phlipponneau M, et al. Somatostatin analogs for the localization and preoperative treatment of an adrenocorticotropin-secreting bronchial carcinoid tumor. J Clin Endocrinol Metab. 1994;78(1):20–4.PubMed Phlipponneau M, et al. Somatostatin analogs for the localization and preoperative treatment of an adrenocorticotropin-secreting bronchial carcinoid tumor. J Clin Endocrinol Metab. 1994;78(1):20–4.PubMed
112.
go back to reference Pivonello R, et al. Dopamine receptor expression and function in corticotroph ectopic tumors. J Clin Endocrinol Metab. 2007;92(1):65–9.PubMed Pivonello R, et al. Dopamine receptor expression and function in corticotroph ectopic tumors. J Clin Endocrinol Metab. 2007;92(1):65–9.PubMed
113.
go back to reference Pivonello R, et al. Cabergoline plus lanreotide for ectopic Cushing’s syndrome. N Engl J Med. 2005;352(23):2457–8.PubMed Pivonello R, et al. Cabergoline plus lanreotide for ectopic Cushing’s syndrome. N Engl J Med. 2005;352(23):2457–8.PubMed
114.
go back to reference de Herder WW, et al. Somatostatin receptor scintigraphy: its value in tumor localization in patients with Cushing’s syndrome caused by ectopic corticotropin or corticotropin-releasing hormone secretion. Am J Med. 1994;96(4):305–12.PubMed de Herder WW, et al. Somatostatin receptor scintigraphy: its value in tumor localization in patients with Cushing’s syndrome caused by ectopic corticotropin or corticotropin-releasing hormone secretion. Am J Med. 1994;96(4):305–12.PubMed
115.
go back to reference Baudry C, Paepegaey AC, Groussin L. Reversal of Cushing’s syndrome by vandetanib in medullary thyroid carcinoma. N Engl J Med. 2013;369(6):584–6.PubMed Baudry C, Paepegaey AC, Groussin L. Reversal of Cushing’s syndrome by vandetanib in medullary thyroid carcinoma. N Engl J Med. 2013;369(6):584–6.PubMed
116.
go back to reference Barroso-Sousa R, et al. Complete resolution of hypercortisolism with sorafenib in a patient with advanced medullary thyroid carcinoma and ectopic ACTH (adrenocorticotropic hormone) syndrome. Thyroid. 2014;24(6):1062–6.PubMed Barroso-Sousa R, et al. Complete resolution of hypercortisolism with sorafenib in a patient with advanced medullary thyroid carcinoma and ectopic ACTH (adrenocorticotropic hormone) syndrome. Thyroid. 2014;24(6):1062–6.PubMed
117.
go back to reference Kamenicky P, et al. Mitotane, metyrapone, and ketoconazole combination therapy as an alternative to rescue adrenalectomy for severe ACTH-dependent Cushing’s syndrome. J Clin Endocrinol Metab. 2011;96(9):2796–804.PubMed Kamenicky P, et al. Mitotane, metyrapone, and ketoconazole combination therapy as an alternative to rescue adrenalectomy for severe ACTH-dependent Cushing’s syndrome. J Clin Endocrinol Metab. 2011;96(9):2796–804.PubMed
118.
go back to reference Schulte HM, et al. Infusion of low dose etomidate: correction of hypercortisolemia in patients with Cushing’s syndrome and dose-response relationship in normal subjects. J Clin Endocrinol Metab. 1990;70(5):1426–30.PubMed Schulte HM, et al. Infusion of low dose etomidate: correction of hypercortisolemia in patients with Cushing’s syndrome and dose-response relationship in normal subjects. J Clin Endocrinol Metab. 1990;70(5):1426–30.PubMed
119.
go back to reference Nieman LK, et al. Treatment of Cushing’s syndrome: an endocrine society clinical practice guideline. J Clin Endocrinol Metab. 2015;100(8):2807–31.PubMedCentralPubMed Nieman LK, et al. Treatment of Cushing’s syndrome: an endocrine society clinical practice guideline. J Clin Endocrinol Metab. 2015;100(8):2807–31.PubMedCentralPubMed
120.
go back to reference Athanassiadi K, et al. Acromegaly caused by ectopic growth hormone-releasing hormone secretion by a carcinoid bronchial tumor: a rare entity. J Thorac Cardiovasc Surg. 2004;128(4):631–2.PubMed Athanassiadi K, et al. Acromegaly caused by ectopic growth hormone-releasing hormone secretion by a carcinoid bronchial tumor: a rare entity. J Thorac Cardiovasc Surg. 2004;128(4):631–2.PubMed
121.
go back to reference Dayal Y, et al. Immunocytochemical demonstration of growth hormone-releasing factor in gastrointestinal and pancreatic endocrine tumors. Am J Clin Pathol. 1986;85(1):13–20.PubMed Dayal Y, et al. Immunocytochemical demonstration of growth hormone-releasing factor in gastrointestinal and pancreatic endocrine tumors. Am J Clin Pathol. 1986;85(1):13–20.PubMed
122.
go back to reference Gola M, et al. Neuroendocrine tumors secreting growth hormone-releasing hormone: pathophysiological and clinical aspects. Pituitary. 2006;9(3):221–9.PubMed Gola M, et al. Neuroendocrine tumors secreting growth hormone-releasing hormone: pathophysiological and clinical aspects. Pituitary. 2006;9(3):221–9.PubMed
123.
go back to reference Giustina A, et al. Diagnosis and treatment of acromegaly complications. J Endocrinol Invest. 2003;26(12):1242–7.PubMed Giustina A, et al. Diagnosis and treatment of acromegaly complications. J Endocrinol Invest. 2003;26(12):1242–7.PubMed
124.
go back to reference van Hoek M, et al. Effects of somatostatin analogs on a growth hormone-releasing hormone secreting bronchial carcinoid, in vivo and in vitro studies. J Clin Endocrinol Metab. 2009;94(2):428–33.PubMed van Hoek M, et al. Effects of somatostatin analogs on a growth hormone-releasing hormone secreting bronchial carcinoid, in vivo and in vitro studies. J Clin Endocrinol Metab. 2009;94(2):428–33.PubMed
125.
go back to reference Van den Bruel A, et al. Hormonal and volumetric long term control of a growth hormone-releasing hormone-producing carcinoid tumor. J Clin Endocrinol Metab. 1999;84(9):3162–9.PubMed Van den Bruel A, et al. Hormonal and volumetric long term control of a growth hormone-releasing hormone-producing carcinoid tumor. J Clin Endocrinol Metab. 1999;84(9):3162–9.PubMed
126.
go back to reference Rindi G, et al. TNM staging of neoplasms of the endocrine pancreas: results from a large international cohort study. J Natl Cancer Inst. 2012;104(10):764–77.PubMed Rindi G, et al. TNM staging of neoplasms of the endocrine pancreas: results from a large international cohort study. J Natl Cancer Inst. 2012;104(10):764–77.PubMed
127.
go back to reference Green JR. Bisphosphonates: preclinical review. Oncologist. 2004;9(Suppl 4):3–13.PubMed Green JR. Bisphosphonates: preclinical review. Oncologist. 2004;9(Suppl 4):3–13.PubMed
128.
go back to reference Loftus LS, Edwards-Bennett S, Sokol GH. Systemic therapy for bone metastases. Cancer Control. 2012;19(2):145–53.PubMed Loftus LS, Edwards-Bennett S, Sokol GH. Systemic therapy for bone metastases. Cancer Control. 2012;19(2):145–53.PubMed
129.
go back to reference Jensen RT. Carcinoid and pancreatic endocrine tumors: recent advances in molecular pathogenesis, localization, and treatment. Curr Opin Oncol. 2000;12(4):368–77.PubMed Jensen RT. Carcinoid and pancreatic endocrine tumors: recent advances in molecular pathogenesis, localization, and treatment. Curr Opin Oncol. 2000;12(4):368–77.PubMed
130.
go back to reference Wangberg B, et al. Somatostatin receptors. A new way to diagnosis and therapy of neuroendocrine tumors. Lakartidningen. 1997;94(10):829–30, 835–8. Wangberg B, et al. Somatostatin receptors. A new way to diagnosis and therapy of neuroendocrine tumors. Lakartidningen. 1997;94(10):829–30, 835–8.
131.
go back to reference Delle Fave G, et al. ENETS consensus guidelines update for gastroduodenal neuroendocrine neoplasms. Neuroendocrinology. 2016;103(2):119–24.PubMed Delle Fave G, et al. ENETS consensus guidelines update for gastroduodenal neuroendocrine neoplasms. Neuroendocrinology. 2016;103(2):119–24.PubMed
132.
go back to reference Rinke A, et al. Placebo-controlled, double-blind, prospective, randomized study on the effect of octreotide LAR in the control of tumor growth in patients with metastatic neuroendocrine midgut tumors: a report from the PROMID Study Group. J Clin Oncol. 2009;27(28):4656–63.PubMed Rinke A, et al. Placebo-controlled, double-blind, prospective, randomized study on the effect of octreotide LAR in the control of tumor growth in patients with metastatic neuroendocrine midgut tumors: a report from the PROMID Study Group. J Clin Oncol. 2009;27(28):4656–63.PubMed
133.
go back to reference Rinke A, et al. Placebo-controlled, double-blind, prospective, randomized study on the effect of octreotide LAR in the control of tumor growth in patients with metastatic neuroendocrine midgut tumors (PROMID): results of long-term survival. Neuroendocrinology. 2017;104(1):26–32.PubMed Rinke A, et al. Placebo-controlled, double-blind, prospective, randomized study on the effect of octreotide LAR in the control of tumor growth in patients with metastatic neuroendocrine midgut tumors (PROMID): results of long-term survival. Neuroendocrinology. 2017;104(1):26–32.PubMed
134.
go back to reference Caplin ME, Pavel M, Ruszniewski P. Lanreotide in metastatic enteropancreatic neuroendocrine tumors. N Engl J Med. 2014;371(16):1556–7.PubMed Caplin ME, Pavel M, Ruszniewski P. Lanreotide in metastatic enteropancreatic neuroendocrine tumors. N Engl J Med. 2014;371(16):1556–7.PubMed
135.
go back to reference Zomerhuis MT, et al. Octreotide exerts only acute, but no sustained, effects on MRI enhancement of liver metastases in carcinoid syndrome. Neuroendocrinology. 2005;82(1):41–8.PubMed Zomerhuis MT, et al. Octreotide exerts only acute, but no sustained, effects on MRI enhancement of liver metastases in carcinoid syndrome. Neuroendocrinology. 2005;82(1):41–8.PubMed
136.
go back to reference Imam H, et al. Induction of apoptosis in neuroendocrine tumors of the digestive system during treatment with somatostatin analogs. Acta Oncol. 1997;36(6):607–14.PubMed Imam H, et al. Induction of apoptosis in neuroendocrine tumors of the digestive system during treatment with somatostatin analogs. Acta Oncol. 1997;36(6):607–14.PubMed
137.
go back to reference Welin SV, et al. High-dose treatment with a long-acting somatostatin analogue in patients with advanced midgut carcinoid tumours. Eur J Endocrinol. 2004;151(1):107–12.PubMed Welin SV, et al. High-dose treatment with a long-acting somatostatin analogue in patients with advanced midgut carcinoid tumours. Eur J Endocrinol. 2004;151(1):107–12.PubMed
138.
go back to reference Yao JC, et al. Phase I dose-escalation study of long-acting pasireotide in patients with neuroendocrine tumors. Onco Targets Ther. 2017;10:3177–86.PubMedCentralPubMed Yao JC, et al. Phase I dose-escalation study of long-acting pasireotide in patients with neuroendocrine tumors. Onco Targets Ther. 2017;10:3177–86.PubMedCentralPubMed
139.
go back to reference Cives M, et al. Phase II clinical trial of pasireotide long-acting repeatable in patients with metastatic neuroendocrine tumors. Endocr Relat Cancer. 2015;22(1):1–9.PubMed Cives M, et al. Phase II clinical trial of pasireotide long-acting repeatable in patients with metastatic neuroendocrine tumors. Endocr Relat Cancer. 2015;22(1):1–9.PubMed
140.
go back to reference Ferolla P, et al. Efficacy and safety of long-acting pasireotide or everolimus alone or in combination in patients with advanced carcinoids of the lung and thymus (LUNA): an open-label, multicentre, randomised, phase II trial. Lancet Oncol. 2017;18(12):1652–64.PubMed Ferolla P, et al. Efficacy and safety of long-acting pasireotide or everolimus alone or in combination in patients with advanced carcinoids of the lung and thymus (LUNA): an open-label, multicentre, randomised, phase II trial. Lancet Oncol. 2017;18(12):1652–64.PubMed
141.
go back to reference Gillis JC, Noble S, Goa KL. Octreotide long-acting release (LAR). A review of its pharmacological properties and therapeutic use in the management of acromegaly. Drugs. 1997;53(4):681–99. Gillis JC, Noble S, Goa KL. Octreotide long-acting release (LAR). A review of its pharmacological properties and therapeutic use in the management of acromegaly. Drugs. 1997;53(4):681–99.
142.
go back to reference Oberg K, Eriksson B. The role of interferons in the management of carcinoid tumours. Br J Haematol. 1991;79(Suppl 1):74–7.PubMed Oberg K, Eriksson B. The role of interferons in the management of carcinoid tumours. Br J Haematol. 1991;79(Suppl 1):74–7.PubMed
143.
go back to reference Oberg K, Funa K, Alm G. Effects of leukocyte interferon on clinical symptoms and hormone levels in patients with mid-gut carcinoid tumors and carcinoid syndrome. N Engl J Med. 1983;309(3):129–33.PubMed Oberg K, Funa K, Alm G. Effects of leukocyte interferon on clinical symptoms and hormone levels in patients with mid-gut carcinoid tumors and carcinoid syndrome. N Engl J Med. 1983;309(3):129–33.PubMed
144.
go back to reference Plockinger U, et al. Guidelines for the diagnosis and treatment of neuroendocrine gastrointestinal tumours. A consensus statement on behalf of the European Neuroendocrine Tumour Society (ENETS). Neuroendocrinology. 2004;80(6):394–424. Plockinger U, et al. Guidelines for the diagnosis and treatment of neuroendocrine gastrointestinal tumours. A consensus statement on behalf of the European Neuroendocrine Tumour Society (ENETS). Neuroendocrinology. 2004;80(6):394–424.
145.
go back to reference Kolby L, et al. Randomized clinical trial of the effect of interferon alpha on survival in patients with disseminated midgut carcinoid tumours. Br J Surg. 2003;90(6):687–93.PubMed Kolby L, et al. Randomized clinical trial of the effect of interferon alpha on survival in patients with disseminated midgut carcinoid tumours. Br J Surg. 2003;90(6):687–93.PubMed
146.
go back to reference Oberg K. Interferon-alpha versus somatostatin or the combination of both in gastro-enteropancreatic tumours. Digestion. 1996;57(Suppl 1):81–3.PubMed Oberg K. Interferon-alpha versus somatostatin or the combination of both in gastro-enteropancreatic tumours. Digestion. 1996;57(Suppl 1):81–3.PubMed
147.
go back to reference Yao JC, et al. Phase III prospective randomized comparison trial of depot octreotide plus interferon Alfa-2b versus depot octreotide plus bevacizumab in patients with advanced carcinoid tumors: SWOG S0518. J Clin Oncol. 2017;35(15):1695–703.PubMedCentralPubMed Yao JC, et al. Phase III prospective randomized comparison trial of depot octreotide plus interferon Alfa-2b versus depot octreotide plus bevacizumab in patients with advanced carcinoid tumors: SWOG S0518. J Clin Oncol. 2017;35(15):1695–703.PubMedCentralPubMed
148.
go back to reference Pavel ME, et al. Efficacy and tolerability of pegylated IFN-alpha in patients with neuroendocrine gastroenteropancreatic carcinomas. J Interferon Cytokine Res. 2006;26(1):8–13.PubMed Pavel ME, et al. Efficacy and tolerability of pegylated IFN-alpha in patients with neuroendocrine gastroenteropancreatic carcinomas. J Interferon Cytokine Res. 2006;26(1):8–13.PubMed
149.
go back to reference Zandee WT, de Herder WW. The evolution of neuroendocrine tumor treatment reflected by ENETS guidelines. Neuroendocrinology. 2018;106(4):357–65.PubMed Zandee WT, de Herder WW. The evolution of neuroendocrine tumor treatment reflected by ENETS guidelines. Neuroendocrinology. 2018;106(4):357–65.PubMed
150.
go back to reference Cives M, Strosberg J. Radionuclide therapy for neuroendocrine tumors. Curr Oncol Rep. 2017;19(2):9.PubMed Cives M, Strosberg J. Radionuclide therapy for neuroendocrine tumors. Curr Oncol Rep. 2017;19(2):9.PubMed
151.
go back to reference Pool SE, et al. Preclinical and clinical studies of peptide receptor radionuclide therapy. Semin Nucl Med. 2010;40(3):209–18.PubMed Pool SE, et al. Preclinical and clinical studies of peptide receptor radionuclide therapy. Semin Nucl Med. 2010;40(3):209–18.PubMed
152.
go back to reference Kwekkeboom DJ, et al. Somatostatin-receptor-based imaging and therapy of gastroenteropancreatic neuroendocrine tumors. Endocr Relat Cancer. 2010;17(1):R53–73.PubMed Kwekkeboom DJ, et al. Somatostatin-receptor-based imaging and therapy of gastroenteropancreatic neuroendocrine tumors. Endocr Relat Cancer. 2010;17(1):R53–73.PubMed
153.
go back to reference van Adrichem RC, et al. Is there an additional value of using somatostatin receptor subtype 2a immunohistochemistry compared to somatostatin receptor scintigraphy uptake in predicting gastroenteropancreatic neuroendocrine tumor response? Neuroendocrinology. 2016;103(5):560–6.PubMed van Adrichem RC, et al. Is there an additional value of using somatostatin receptor subtype 2a immunohistochemistry compared to somatostatin receptor scintigraphy uptake in predicting gastroenteropancreatic neuroendocrine tumor response? Neuroendocrinology. 2016;103(5):560–6.PubMed
154.
go back to reference Severi S, et al. Peptide receptor radionuclide therapy in the management of gastrointestinal neuroendocrine tumors: efficacy profile, safety, and quality of life. Onco Targets Ther. 2017;10:551–7.PubMedCentralPubMed Severi S, et al. Peptide receptor radionuclide therapy in the management of gastrointestinal neuroendocrine tumors: efficacy profile, safety, and quality of life. Onco Targets Ther. 2017;10:551–7.PubMedCentralPubMed
155.
go back to reference Brabander T, et al. Long-term efficacy, survival, and safety of [(177)Lu-DOTA(0), Tyr(3)]octreotate in patients with gastroenteropancreatic and bronchial neuroendocrine tumors. Clin Cancer Res. 2017;23(16):4617–24.PubMed Brabander T, et al. Long-term efficacy, survival, and safety of [(177)Lu-DOTA(0), Tyr(3)]octreotate in patients with gastroenteropancreatic and bronchial neuroendocrine tumors. Clin Cancer Res. 2017;23(16):4617–24.PubMed
156.
go back to reference Kwekkeboom DJ, et al. Radiolabeled somatostatin analog [177Lu-DOTA0, Tyr3]octreotate in patients with endocrine gastroenteropancreatic tumors. J Clin Oncol. 2005;23(12):2754–62.PubMed Kwekkeboom DJ, et al. Radiolabeled somatostatin analog [177Lu-DOTA0, Tyr3]octreotate in patients with endocrine gastroenteropancreatic tumors. J Clin Oncol. 2005;23(12):2754–62.PubMed
157.
go back to reference Ezziddin S, et al. Predictors of long-term outcome in patients with well-differentiated gastroenteropancreatic neuroendocrine tumors after peptide receptor radionuclide therapy with 177Lu-octreotate. J Nucl Med. 2014;55(2):183–90.PubMed Ezziddin S, et al. Predictors of long-term outcome in patients with well-differentiated gastroenteropancreatic neuroendocrine tumors after peptide receptor radionuclide therapy with 177Lu-octreotate. J Nucl Med. 2014;55(2):183–90.PubMed
158.
go back to reference Valkema R, et al. Survival and response after peptide receptor radionuclide therapy with [90Y-DOTA0, Tyr3]octreotide in patients with advanced gastroenteropancreatic neuroendocrine tumors. Semin Nucl Med. 2006;36(2):147–56.PubMed Valkema R, et al. Survival and response after peptide receptor radionuclide therapy with [90Y-DOTA0, Tyr3]octreotide in patients with advanced gastroenteropancreatic neuroendocrine tumors. Semin Nucl Med. 2006;36(2):147–56.PubMed
159.
go back to reference Bushnell DL Jr, et al. 90Y-edotreotide for metastatic carcinoid refractory to octreotide. J Clin Oncol. 2010;28(10):1652–9.PubMedCentralPubMed Bushnell DL Jr, et al. 90Y-edotreotide for metastatic carcinoid refractory to octreotide. J Clin Oncol. 2010;28(10):1652–9.PubMedCentralPubMed
160.
go back to reference Imhof A, et al. Response, survival, and long-term toxicity after therapy with the radiolabeled somatostatin analogue [90Y-DOTA]-TOC in metastasized neuroendocrine cancers. J Clin Oncol. 2011;29(17):2416–23.PubMed Imhof A, et al. Response, survival, and long-term toxicity after therapy with the radiolabeled somatostatin analogue [90Y-DOTA]-TOC in metastasized neuroendocrine cancers. J Clin Oncol. 2011;29(17):2416–23.PubMed
161.
go back to reference Bodei L, et al. Peptide receptor radionuclide therapy with (1)(7)(7)Lu-DOTATATE: the IEO phase I-II study. Eur J Nucl Med Mol Imaging. 2011;38(12):2125–35.PubMed Bodei L, et al. Peptide receptor radionuclide therapy with (1)(7)(7)Lu-DOTATATE: the IEO phase I-II study. Eur J Nucl Med Mol Imaging. 2011;38(12):2125–35.PubMed
162.
go back to reference Sabet A, et al. Specific efficacy of peptide receptor radionuclide therapy with (177)Lu-octreotate in advanced neuroendocrine tumours of the small intestine. Eur J Nucl Med Mol Imaging. 2015;42(8):1238–46.PubMed Sabet A, et al. Specific efficacy of peptide receptor radionuclide therapy with (177)Lu-octreotate in advanced neuroendocrine tumours of the small intestine. Eur J Nucl Med Mol Imaging. 2015;42(8):1238–46.PubMed
163.
go back to reference Paganelli G, et al. 177 Lu-Dota-octreotate radionuclide therapy of advanced gastrointestinal neuroendocrine tumors: results from a phase II study. Eur J Nucl Med Mol Imaging. 2014;41(10):1845–51.PubMed Paganelli G, et al. 177 Lu-Dota-octreotate radionuclide therapy of advanced gastrointestinal neuroendocrine tumors: results from a phase II study. Eur J Nucl Med Mol Imaging. 2014;41(10):1845–51.PubMed
164.
go back to reference Kim SJ, et al. The efficacy of (177)Lu-labelled peptide receptor radionuclide therapy in patients with neuroendocrine tumours: a meta-analysis. Eur J Nucl Med Mol Imaging. 2015;42(13):1964–70.PubMed Kim SJ, et al. The efficacy of (177)Lu-labelled peptide receptor radionuclide therapy in patients with neuroendocrine tumours: a meta-analysis. Eur J Nucl Med Mol Imaging. 2015;42(13):1964–70.PubMed
165.
go back to reference Khan S, et al. Quality of life in 265 patients with gastroenteropancreatic or bronchial neuroendocrine tumors treated with [177Lu-DOTA0, Tyr3]octreotate. J Nucl Med. 2011;52(9):1361–8.PubMed Khan S, et al. Quality of life in 265 patients with gastroenteropancreatic or bronchial neuroendocrine tumors treated with [177Lu-DOTA0, Tyr3]octreotate. J Nucl Med. 2011;52(9):1361–8.PubMed
166.
go back to reference Strosberg J, et al. Phase 3 trial of (177)Lu-dotatate for midgut neuroendocrine tumors. N Engl J Med. 2017;376(2):125–35.PubMedCentralPubMed Strosberg J, et al. Phase 3 trial of (177)Lu-dotatate for midgut neuroendocrine tumors. N Engl J Med. 2017;376(2):125–35.PubMedCentralPubMed
167.
go back to reference Ianniello A, et al. Peptide receptor radionuclide therapy with (177)Lu-DOTATATE in advanced bronchial carcinoids: prognostic role of thyroid transcription factor 1 and (18)F-FDG PET. Eur J Nucl Med Mol Imaging. 2016;43(6):1040–6.PubMed Ianniello A, et al. Peptide receptor radionuclide therapy with (177)Lu-DOTATATE in advanced bronchial carcinoids: prognostic role of thyroid transcription factor 1 and (18)F-FDG PET. Eur J Nucl Med Mol Imaging. 2016;43(6):1040–6.PubMed
168.
go back to reference Severi S, et al. Feasibility and utility of re-treatment with (177)Lu-DOTATATE in GEP-NENs relapsed after treatment with (90)Y-DOTATOC. Eur J Nucl Med Mol Imaging. 2015;42(13):1955–63.PubMed Severi S, et al. Feasibility and utility of re-treatment with (177)Lu-DOTATATE in GEP-NENs relapsed after treatment with (90)Y-DOTATOC. Eur J Nucl Med Mol Imaging. 2015;42(13):1955–63.PubMed
169.
go back to reference Forrer F, et al. Treatment with 177Lu-DOTATOC of patients with relapse of neuroendocrine tumors after treatment with 90Y-DOTATOC. J Nucl Med. 2005;46(8):1310–6.PubMed Forrer F, et al. Treatment with 177Lu-DOTATOC of patients with relapse of neuroendocrine tumors after treatment with 90Y-DOTATOC. J Nucl Med. 2005;46(8):1310–6.PubMed
170.
go back to reference Claringbold PG, et al. Phase II study of radiopeptide 177Lu-octreotate and capecitabine therapy of progressive disseminated neuroendocrine tumours. Eur J Nucl Med Mol Imaging. 2011;38(2):302–11.PubMed Claringbold PG, et al. Phase II study of radiopeptide 177Lu-octreotate and capecitabine therapy of progressive disseminated neuroendocrine tumours. Eur J Nucl Med Mol Imaging. 2011;38(2):302–11.PubMed
171.
go back to reference Claringbold PG, Price RA, Turner JH. Phase I-II study of radiopeptide 177Lu-octreotate in combination with capecitabine and temozolomide in advanced low-grade neuroendocrine tumors. Cancer Biother Radiopharm. 2012;27(9):561–9.PubMed Claringbold PG, Price RA, Turner JH. Phase I-II study of radiopeptide 177Lu-octreotate in combination with capecitabine and temozolomide in advanced low-grade neuroendocrine tumors. Cancer Biother Radiopharm. 2012;27(9):561–9.PubMed
172.
go back to reference Kong G, et al. Assessment of predictors of response and long-term survival of patients with neuroendocrine tumour treated with peptide receptor chemoradionuclide therapy (PRCRT). Eur J Nucl Med Mol Imaging. 2014;41(10):1831–44.PubMedCentralPubMed Kong G, et al. Assessment of predictors of response and long-term survival of patients with neuroendocrine tumour treated with peptide receptor chemoradionuclide therapy (PRCRT). Eur J Nucl Med Mol Imaging. 2014;41(10):1831–44.PubMedCentralPubMed
173.
go back to reference Claringbold PG, Turner JH. NeuroEndocrine tumor therapy with lutetium-177-octreotate and everolimus (NETTLE): a phase I study. Cancer Biother Radiopharm. 2015;30(6):261–9.PubMed Claringbold PG, Turner JH. NeuroEndocrine tumor therapy with lutetium-177-octreotate and everolimus (NETTLE): a phase I study. Cancer Biother Radiopharm. 2015;30(6):261–9.PubMed
174.
go back to reference Barber TW, et al. The potential for induction peptide receptor chemoradionuclide therapy to render inoperable pancreatic and duodenal neuroendocrine tumours resectable. Eur J Surg Oncol. 2012;38(1):64–71.PubMed Barber TW, et al. The potential for induction peptide receptor chemoradionuclide therapy to render inoperable pancreatic and duodenal neuroendocrine tumours resectable. Eur J Surg Oncol. 2012;38(1):64–71.PubMed
175.
go back to reference Ezziddin S, et al. Neoadjuvant downsizing by internal radiation: a case for preoperative peptide receptor radionuclide therapy in patients with pancreatic neuroendocrine tumors. Clin Nucl Med. 2012;37(1):102–4.PubMed Ezziddin S, et al. Neoadjuvant downsizing by internal radiation: a case for preoperative peptide receptor radionuclide therapy in patients with pancreatic neuroendocrine tumors. Clin Nucl Med. 2012;37(1):102–4.PubMed
176.
go back to reference van Vliet EI, et al. Neoadjuvant treatment of nonfunctioning pancreatic neuroendocrine tumors with [177Lu-DOTA0, Tyr3]octreotate. J Nucl Med. 2015;56(11):1647–53.PubMed van Vliet EI, et al. Neoadjuvant treatment of nonfunctioning pancreatic neuroendocrine tumors with [177Lu-DOTA0, Tyr3]octreotate. J Nucl Med. 2015;56(11):1647–53.PubMed
177.
go back to reference Brabander T, et al. Pitfalls in the response evaluation after peptide receptor radionuclide therapy with [(177)Lu-DOTA(0), Tyr(3)]octreotate. Endocr Relat Cancer. 2017;24(5):243–51.PubMed Brabander T, et al. Pitfalls in the response evaluation after peptide receptor radionuclide therapy with [(177)Lu-DOTA(0), Tyr(3)]octreotate. Endocr Relat Cancer. 2017;24(5):243–51.PubMed
178.
go back to reference Ginj M, et al. Radiolabeled somatostatin receptor antagonists are preferable to agonists for in vivo peptide receptor targeting of tumors. Proc Natl Acad Sci USA. 2006;103(44):16436–41.PubMedPubMedCentral Ginj M, et al. Radiolabeled somatostatin receptor antagonists are preferable to agonists for in vivo peptide receptor targeting of tumors. Proc Natl Acad Sci USA. 2006;103(44):16436–41.PubMedPubMedCentral
179.
go back to reference Nonnekens J, et al. Potentiation of peptide receptor radionuclide therapy by the PARP inhibitor olaparib. Theranostics. 2016;6(11):1821–32.PubMedCentralPubMed Nonnekens J, et al. Potentiation of peptide receptor radionuclide therapy by the PARP inhibitor olaparib. Theranostics. 2016;6(11):1821–32.PubMedCentralPubMed
180.
go back to reference Bodei L, et al. Measurement of circulating transcripts and gene cluster analysis predicts and defines therapeutic efficacy of peptide receptor radionuclide therapy (PRRT) in neuroendocrine tumors. Eur J Nucl Med Mol Imaging. 2016;43(5):839–51.PubMed Bodei L, et al. Measurement of circulating transcripts and gene cluster analysis predicts and defines therapeutic efficacy of peptide receptor radionuclide therapy (PRRT) in neuroendocrine tumors. Eur J Nucl Med Mol Imaging. 2016;43(5):839–51.PubMed
181.
go back to reference Kidd M, Modlin IM. Therapy: the role of liquid biopsies to manage and predict PRRT for NETs. Nat Rev Gastroenterol Hepatol. 2017;14(6):331–2.PubMed Kidd M, Modlin IM. Therapy: the role of liquid biopsies to manage and predict PRRT for NETs. Nat Rev Gastroenterol Hepatol. 2017;14(6):331–2.PubMed
182.
go back to reference Bodei L, et al. PRRT genomic signature in blood for prediction of (177)Lu-octreotate efficacy. Eur J Nucl Med Mol Imaging. 2018;45(7):1155–69.PubMedPubMedCentral Bodei L, et al. PRRT genomic signature in blood for prediction of (177)Lu-octreotate efficacy. Eur J Nucl Med Mol Imaging. 2018;45(7):1155–69.PubMedPubMedCentral
183.
go back to reference Bodei L, Pepe G, Paganelli G. Peptide receptor radionuclide therapy (PRRT) of neuroendocrine tumors with somatostatin analogues. Eur Rev Med Pharmacol Sci. 2010;14(4):347–51.PubMed Bodei L, Pepe G, Paganelli G. Peptide receptor radionuclide therapy (PRRT) of neuroendocrine tumors with somatostatin analogues. Eur Rev Med Pharmacol Sci. 2010;14(4):347–51.PubMed
184.
go back to reference Yao JC, et al. Everolimus for the treatment of advanced pancreatic neuroendocrine tumors: overall survival and circulating biomarkers from the randomized, phase III RADIANT-3 study. J Clin Oncol. 2016;34(32):3906–13.PubMedCentralPubMed Yao JC, et al. Everolimus for the treatment of advanced pancreatic neuroendocrine tumors: overall survival and circulating biomarkers from the randomized, phase III RADIANT-3 study. J Clin Oncol. 2016;34(32):3906–13.PubMedCentralPubMed
185.
go back to reference Lombard-Bohas C, et al. Impact of prior chemotherapy use on the efficacy of everolimus in patients with advanced pancreatic neuroendocrine tumors: a subgroup analysis of the phase III RADIANT-3 trial. Pancreas. 2015;44(2):181–9.PubMedCentralPubMed Lombard-Bohas C, et al. Impact of prior chemotherapy use on the efficacy of everolimus in patients with advanced pancreatic neuroendocrine tumors: a subgroup analysis of the phase III RADIANT-3 trial. Pancreas. 2015;44(2):181–9.PubMedCentralPubMed
186.
go back to reference Anthony LB, et al. Impact of previous somatostatin analogue use on the activity of everolimus in patients with advanced neuroendocrine tumors: analysis from the phase III RADIANT-2 trial. Neuroendocrinology. 2015;102(1–2):18–25.PubMed Anthony LB, et al. Impact of previous somatostatin analogue use on the activity of everolimus in patients with advanced neuroendocrine tumors: analysis from the phase III RADIANT-2 trial. Neuroendocrinology. 2015;102(1–2):18–25.PubMed
187.
go back to reference Yao JC, et al. Everolimus for the treatment of advanced, non-functional neuroendocrine tumours of the lung or gastrointestinal tract (RADIANT-4): a randomised, placebo-controlled, phase III study. Lancet. 2016;387(10022):968–77.PubMed Yao JC, et al. Everolimus for the treatment of advanced, non-functional neuroendocrine tumours of the lung or gastrointestinal tract (RADIANT-4): a randomised, placebo-controlled, phase III study. Lancet. 2016;387(10022):968–77.PubMed
188.
go back to reference Pavel ME, 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 III study. Lancet. 2011;378(9808):2005–12.PubMed Pavel ME, 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 III study. Lancet. 2011;378(9808):2005–12.PubMed
189.
go back to reference Fazio N, et al. Everolimus plus octreotide long-acting repeatable in patients with advanced lung neuroendocrine tumors: analysis of the phase III, randomized, placebo-controlled RADIANT-2 study. Chest. 2013;143(4):955–62.PubMed Fazio N, et al. Everolimus plus octreotide long-acting repeatable in patients with advanced lung neuroendocrine tumors: analysis of the phase III, randomized, placebo-controlled RADIANT-2 study. Chest. 2013;143(4):955–62.PubMed
190.
go back to reference Castellano D, 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.PubMed Castellano D, 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.PubMed
191.
go back to reference Ferolla P. Everolimus and pasireotide LAR alone or in combination significantly improved outcome in patients with advanced lung and thymic carcinoids. In: ESMO conference 2016. Abstract. 2016. Ferolla P. Everolimus and pasireotide LAR alone or in combination significantly improved outcome in patients with advanced lung and thymic carcinoids. In: ESMO conference 2016. Abstract. 2016.
192.
go back to reference Bago-Horvath Z, et al. Synergistic effects of erlotinib and everolimus on bronchial carcinoids and large-cell neuroendocrine carcinomas with activated EGFR/AKT/mTOR pathway. Neuroendocrinology. 2012;96(3):228–37.PubMed Bago-Horvath Z, et al. Synergistic effects of erlotinib and everolimus on bronchial carcinoids and large-cell neuroendocrine carcinomas with activated EGFR/AKT/mTOR pathway. Neuroendocrinology. 2012;96(3):228–37.PubMed
193.
go back to reference Wiedmann MW, Mossner J. Safety and efficacy of sunitinib in patients with unresectable pancreatic neuroendocrine tumors. Clin Med Insights Oncol. 2012;6:381–93.PubMedCentralPubMed Wiedmann MW, Mossner J. Safety and efficacy of sunitinib in patients with unresectable pancreatic neuroendocrine tumors. Clin Med Insights Oncol. 2012;6:381–93.PubMedCentralPubMed
194.
go back to reference Kulke MH, et al. Activity of sunitinib in patients with advanced neuroendocrine tumors. J Clin Oncol. 2008;26(20):3403–10.PubMed Kulke MH, et al. Activity of sunitinib in patients with advanced neuroendocrine tumors. J Clin Oncol. 2008;26(20):3403–10.PubMed
195.
go back to reference Faivre S, et al. Sunitinib in pancreatic neuroendocrine tumors: updated progression-free survival and final overall survival from a phase III randomized study. Ann Oncol. 2017;28(2):339–43.PubMed Faivre S, et al. Sunitinib in pancreatic neuroendocrine tumors: updated progression-free survival and final overall survival from a phase III randomized study. Ann Oncol. 2017;28(2):339–43.PubMed
196.
go back to reference Faivre S, et al. Imaging response in neuroendocrine tumors treated with targeted therapies: the experience of sunitinib. Target Oncol. 2012;7(2):127–33.PubMed Faivre S, et al. Imaging response in neuroendocrine tumors treated with targeted therapies: the experience of sunitinib. Target Oncol. 2012;7(2):127–33.PubMed
197.
go back to reference Valle JW, et al. A systematic review of non-surgical treatments for pancreatic neuroendocrine tumours. Cancer Treat Rev. 2014;40(3):376–89.PubMed Valle JW, et al. A systematic review of non-surgical treatments for pancreatic neuroendocrine tumours. Cancer Treat Rev. 2014;40(3):376–89.PubMed
198.
go back to reference Castellano D, et al. Sorafenib and bevacizumab combination targeted therapy in advanced neuroendocrine tumour: a phase II study of Spanish Neuroendocrine Tumour Group (GETNE0801). Eur J Cancer. 2013;49(18):3780–7.PubMed Castellano D, et al. Sorafenib and bevacizumab combination targeted therapy in advanced neuroendocrine tumour: a phase II study of Spanish Neuroendocrine Tumour Group (GETNE0801). Eur J Cancer. 2013;49(18):3780–7.PubMed
199.
go back to reference Kulke MH, Niedzwiecki D, Foster NR, et al. Randomized phase II study of everolimus (E) versus everolimus plus bevacizumab (E + B) in patients (Pts) with locally advanced or metastatic pancreatic neuroendocrine tumors (pNET), CALGB 80701 (Alliance). J Clin Oncol. 2015;33(15) (suppl; abstr 4005). Kulke MH, Niedzwiecki D, Foster NR, et al. Randomized phase II study of everolimus (E) versus everolimus plus bevacizumab (E + B) in patients (Pts) with locally advanced or metastatic pancreatic neuroendocrine tumors (pNET), CALGB 80701 (Alliance). J Clin Oncol. 2015;33(15) (suppl; abstr 4005).
200.
go back to reference Chan JA, et al. Phase I study of sorafenib in combination with everolimus (RAD001) in patients with advanced neuroendocrine tumors. Cancer Chemother Pharmacol. 2013;71(5):1241–6.PubMedCentralPubMed Chan JA, et al. Phase I study of sorafenib in combination with everolimus (RAD001) in patients with advanced neuroendocrine tumors. Cancer Chemother Pharmacol. 2013;71(5):1241–6.PubMedCentralPubMed
201.
go back to reference Phan AT, et al. Pazopanib and depot octreotide in advanced, well-differentiated neuroendocrine tumours: a multicentre, single-group, phase II study. Lancet Oncol. 2015;16(6):695–703.PubMedCentralPubMed Phan AT, et al. Pazopanib and depot octreotide in advanced, well-differentiated neuroendocrine tumours: a multicentre, single-group, phase II study. Lancet Oncol. 2015;16(6):695–703.PubMedCentralPubMed
202.
go back to reference Fazio N, Spada F, Giovannini M. Chemotherapy in gastroenteropancreatic (GEP) neuroendocrine carcinomas (NEC): a critical view. Cancer Treat Rev. 2013;39(3):270–4.PubMed Fazio N, Spada F, Giovannini M. Chemotherapy in gastroenteropancreatic (GEP) neuroendocrine carcinomas (NEC): a critical view. Cancer Treat Rev. 2013;39(3):270–4.PubMed
203.
go back to reference Auernhammer CJ, et al. Advanced neuroendocrine tumours of the small intestine and pancreas: clinical developments, controversies, and future strategies. Lancet Diabetes Endocrinol. 2018;6(5):404–15.PubMed Auernhammer CJ, et al. Advanced neuroendocrine tumours of the small intestine and pancreas: clinical developments, controversies, and future strategies. Lancet Diabetes Endocrinol. 2018;6(5):404–15.PubMed
204.
go back to reference Kunz PL, et al. Oxaliplatin-fluoropyrimidine chemotherapy plus bevacizumab in advanced neuroendocrine tumors: an analysis of 2 phase II trials. Pancreas. 2016;45(10):1394–400.PubMed Kunz PL, et al. Oxaliplatin-fluoropyrimidine chemotherapy plus bevacizumab in advanced neuroendocrine tumors: an analysis of 2 phase II trials. Pancreas. 2016;45(10):1394–400.PubMed
205.
go back to reference Aristizabal Prada ET, Auernhammer CJ. Targeted therapy of gastroenteropancreatic neuroendocrine tumours: preclinical strategies and future targets. Endocr Connect. 2018;7(1):R1–25.PubMed Aristizabal Prada ET, Auernhammer CJ. Targeted therapy of gastroenteropancreatic neuroendocrine tumours: preclinical strategies and future targets. Endocr Connect. 2018;7(1):R1–25.PubMed
206.
go back to reference Pennock GK, Chow LQ. The evolving role of immune checkpoint inhibitors in cancer treatment. Oncologist. 2015;20(7):812–22.PubMedCentralPubMed Pennock GK, Chow LQ. The evolving role of immune checkpoint inhibitors in cancer treatment. Oncologist. 2015;20(7):812–22.PubMedCentralPubMed
207.
go back to reference Chauhan A, et al. Immune checkpoint inhibitors in neuroendocrine tumors: a single institution experience with review of literature. Oncotarget. 2018;9(10):8801–9.PubMed Chauhan A, et al. Immune checkpoint inhibitors in neuroendocrine tumors: a single institution experience with review of literature. Oncotarget. 2018;9(10):8801–9.PubMed
208.
209.
go back to reference Herbst RS, et al. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature. 2014;515(7528):563–7.PubMedCentralPubMed Herbst RS, et al. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature. 2014;515(7528):563–7.PubMedCentralPubMed
210.
go back to reference Nomi T, et al. Clinical significance and therapeutic potential of the programmed death-1 ligand/programmed death-1 pathway in human pancreatic cancer. Clin Cancer Res. 2007;13(7):2151–7.PubMed Nomi T, et al. Clinical significance and therapeutic potential of the programmed death-1 ligand/programmed death-1 pathway in human pancreatic cancer. Clin Cancer Res. 2007;13(7):2151–7.PubMed
211.
go back to reference Badoual C, et al. PD-1-expressing tumor-infiltrating T cells are a favorable prognostic biomarker in HPV-associated head and neck cancer. Cancer Res. 2013;73(1):128–38.PubMed Badoual C, et al. PD-1-expressing tumor-infiltrating T cells are a favorable prognostic biomarker in HPV-associated head and neck cancer. Cancer Res. 2013;73(1):128–38.PubMed
212.
go back to reference Droeser RA, et al. Clinical impact of programmed cell death ligand 1 expression in colorectal cancer. Eur J Cancer. 2013;49(9):2233–42.PubMed Droeser RA, et al. Clinical impact of programmed cell death ligand 1 expression in colorectal cancer. Eur J Cancer. 2013;49(9):2233–42.PubMed
213.
go back to reference Tsuruoka K, et al. PD-L1 expression in neuroendocrine tumors of the lung. Lung Cancer. 2017;108:115–20.PubMed Tsuruoka K, et al. PD-L1 expression in neuroendocrine tumors of the lung. Lung Cancer. 2017;108:115–20.PubMed
214.
go back to reference Lamarca A, et al. PD-L1 expression and presence of TILs in small intestinal neuroendocrine tumours. Oncotarget. 2018;9(19):14922–38.PubMedCentralPubMed Lamarca A, et al. PD-L1 expression and presence of TILs in small intestinal neuroendocrine tumours. Oncotarget. 2018;9(19):14922–38.PubMedCentralPubMed
215.
go back to reference Pinato D, Brown M, White S, Zhang H, Toloue S, Dina R, Trivedi R, Stebbing J, Mauri F, Sharma R. Programmed cell death (PD-1) ligands expression in gastro-entero-pancreatic neuroendocrine tumours (GEP-NETs): relationship with angiogenesis and clinical outcome. J Clin Oncol. 2016;34(15_suppl):e15658. Pinato D, Brown M, White S, Zhang H, Toloue S, Dina R, Trivedi R, Stebbing J, Mauri F, Sharma R. Programmed cell death (PD-1) ligands expression in gastro-entero-pancreatic neuroendocrine tumours (GEP-NETs): relationship with angiogenesis and clinical outcome. J Clin Oncol. 2016;34(15_suppl):e15658.
216.
go back to reference Fan Y, et al. Prognostic value of PD-L1 and PD-1 expression in pulmonary neuroendocrine tumors. Onco Targets Ther. 2016;9:6075–82.PubMedCentralPubMed Fan Y, et al. Prognostic value of PD-L1 and PD-1 expression in pulmonary neuroendocrine tumors. Onco Targets Ther. 2016;9:6075–82.PubMedCentralPubMed
217.
go back to reference Reck M, et al. Ipilimumab in combination with paclitaxel and carboplatin as first-line therapy in extensive-disease-small-cell lung cancer: results from a randomized, double-blind, multicenter phase II trial. Ann Oncol. 2013;24(1):75–83.PubMed Reck M, et al. Ipilimumab in combination with paclitaxel and carboplatin as first-line therapy in extensive-disease-small-cell lung cancer: results from a randomized, double-blind, multicenter phase II trial. Ann Oncol. 2013;24(1):75–83.PubMed
218.
go back to reference Tarhini A, Lo E, Minor DR. Releasing the brake on the immune system: ipilimumab in melanoma and other tumors. Cancer Biother Radiopharm. 2010;25(6):601–13.PubMedCentralPubMed Tarhini A, Lo E, Minor DR. Releasing the brake on the immune system: ipilimumab in melanoma and other tumors. Cancer Biother Radiopharm. 2010;25(6):601–13.PubMedCentralPubMed
219.
go back to reference Katz SC, et al. T cell infiltrate and outcome following resection of intermediate-grade primary neuroendocrine tumours and liver metastases. HPB (Oxford). 2010;12(10):674–83. Katz SC, et al. T cell infiltrate and outcome following resection of intermediate-grade primary neuroendocrine tumours and liver metastases. HPB (Oxford). 2010;12(10):674–83.
220.
go back to reference Cavalcanti E, et al. Role of PD-L1 expression as a biomarker for GEP neuroendocrine neoplasm grading. Cell Death Dis. 2017;8(8):e3004.PubMedCentralPubMed Cavalcanti E, et al. Role of PD-L1 expression as a biomarker for GEP neuroendocrine neoplasm grading. Cell Death Dis. 2017;8(8):e3004.PubMedCentralPubMed
221.
go back to reference Culler MD. Somatostatin-dopamine chimeras: a novel approach to treatment of neuroendocrine tumors. Horm Metab Res. 2011;43(12):854–7.PubMed Culler MD. Somatostatin-dopamine chimeras: a novel approach to treatment of neuroendocrine tumors. Horm Metab Res. 2011;43(12):854–7.PubMed
222.
go back to reference Rocheville M, et al. Subtypes of the somatostatin receptor assemble as functional homo- and heterodimers. J Biol Chem. 2000;275(11):7862–9.PubMed Rocheville M, et al. Subtypes of the somatostatin receptor assemble as functional homo- and heterodimers. J Biol Chem. 2000;275(11):7862–9.PubMed
223.
go back to reference Rocheville M, et al. Receptors for dopamine and somatostatin: formation of hetero-oligomers with enhanced functional activity. Science. 2000;288(5463):154–7.PubMed Rocheville M, et al. Receptors for dopamine and somatostatin: formation of hetero-oligomers with enhanced functional activity. Science. 2000;288(5463):154–7.PubMed
224.
go back to reference Couvelard A, et al. Antisecretory effects of chimeric somatostatin/dopamine receptor ligands on gastroenteropancreatic neuroendocrine tumors. Pancreas. 2017;46(5):631–8.PubMed Couvelard A, et al. Antisecretory effects of chimeric somatostatin/dopamine receptor ligands on gastroenteropancreatic neuroendocrine tumors. Pancreas. 2017;46(5):631–8.PubMed
Metadata
Title
Targeted Systemic Treatment of Neuroendocrine Tumors: Current Options and Future Perspectives
Authors
Aura D. Herrera-Martínez
Johannes Hofland
Leo J. Hofland
Tessa Brabander
Ferry A. L. M. Eskens
María A. Gálvez Moreno
Raúl M. Luque
Justo P. Castaño
Wouter W. de Herder
Richard A. Feelders
Publication date
01-01-2019
Publisher
Springer International Publishing
Published in
Drugs / Issue 1/2019
Print ISSN: 0012-6667
Electronic ISSN: 1179-1950
DOI
https://doi.org/10.1007/s40265-018-1033-0

Other articles of this Issue 1/2019

Drugs 1/2019 Go to the issue