258
Views
4
CrossRef citations to date
0
Altmetric
Review

Comparative safety review of the current therapies for gastroenteropancreatic neuroendocrine tumors

&
Pages 321-334 | Received 28 Aug 2020, Accepted 17 Dec 2020, Published online: 27 Dec 2020

References

  • Ameri P, Ferone D. Diffuse endocrine system, neuroendocrine tumors and immunity: what’s new? Neuroendocrinology. 2012;95(4):267–276.
  • Lawrence B, BI G, Chan A, et al. The epidemiology of gastroenteropancreatic neuroendocrine tumors. Endocrinol Metab Clin North Am. 2011;40(1):1–18.
  • Fraenkel M, Kim M, Faggiano A, et al. Incidence of gastroenteropancreatic neuroendocrine tumours: a systematic review of the literature. Endocr Relat Cancer. 2014;21(3):R153–63.
  • Dasari A, Shen C, Halperin D, et al. Trends in the incidence, prevalence, and survival outcomes in patients with neuroendocrine tumors in the United States. JAMA Oncol. 2017;3(10):1335–1342.
  • O’Toole D, Kianmanesh R, ENETS CM. Consensus guidelines for the management of patients with digestive neuroendocrine tumors: an update. Neuroendocrinology. 2016;103(2):117–118.
  • Pape UF, Perren A, Niederle B, et al. Barcelona consensus conference participants. ENETS consensus guidelines for the management of patients with neuroendocrine neoplasms from the jejuno-ileum and the appendix including goblet cell carcinomas. Neuroendocrinology 2012;95:135–156.
  • Niederle B, UF P, Costa F, et al. ENETS consensus guidelines update for neuroendocrine neoplasms of the jejunum and ileum. Neuroendocrinology. 2016;103(2):125–138.
  • Falconi M, Eriksson B, Kaltsas G, et al. Consensus guidelines update for the management of functional p-NETs (F-p-NETs) and non-functional p-NETs (NF-p-NETs). Neuroendocrinology. 2016;103(2):153–171.
  • Herrera‑Martinez AD, Hofland J, Hofland LJ, et al. Targeted systemic treatment of neuroendocrine tumors: current options and future perspectives. Drugs. 2019;79:21–42.
  • Mougey AM, Adler DG. Neuroendocrine tumors: review and clinical update. Hosp Physician. 2007;51:12–20.
  • Rindi G, Falconi M, Klersy C, et al. TNM staging of neoplasms of the endocrine pancreas: results from a large international cohort study. J Natl Cancer Inst. 2012;104(10):764–777.
  • Rindi G, Petrone G, Inzani F. The 2010 WHO classification of digestive neuroendocrine neoplasms: a critical appraisal four years after its introduction. Endocr Pathol. 2014;25(2):186–192.
  • Lloyd RV, Osamura RY, Kloppel G, et al. World health organization, international agency for research on cancer. who classification of tumours of endocrine organs. 4. Geneva: World Health Organization; 2017.
  • Nagtegaal ID, Odze RD, Klimstra D, et al. The 2019 WHO classification of tumours of the digestive system. Histopathology. 2020;76(2):182–188. Jan.
  • Kruljac I, Pape UF. The classification of neuroendocrine neoplasms: “Neuroendocrine carcinomas” revisited – a 2017 update and future perspectives. Endocr Oncol Metab. 2017;3(2):37–42.
  • Rindi G, Kloppel G, Couvelard A, et al. TNM staging of midgut and hindgut (neuro) endocrine tumors: a consensus proposal including a grading system. Virchows Arch. 2007;451(4):757–762.
  • Rindi G, Kloppel G, Alhman H, et al. TNM staging of foregut (neuro)endocrine tumors: a consensus proposal including a grading system. Virchows Arch. 2006;449(4):395–401.
  • Araujo PB, Cheng S, Mete O, et al. Evaluation of the WHO 2010 grading and AJCC/UICC staging systems in prognostic behavior of intestinal neuroendocrine tumors. PLoS One. 2013 Apr 19;8(4):e61538.
  • Elvebakken H, Perren A, Scoazec JY, et al. A consensus developed morphological re-evaluation of 196 high-grade gastroenteropancreatic neuroendocrine neoplasms and its clinical correlations. Neuroendocrinology .2020 Online ahead of print.
  • Capurso G, Rinzivillo M, Bettini R, et al. Systematic review of resection of primary midgut carcinoid tumour in patients with unresectable liver metastases. Br J Surg. 2012;99(11):1480–1486.
  • Howe JR, Cardona K, Fraker DL, et al. The surgical management of small bowel neuroendocrine tumors: consensus guidelines of the North American Neuroendocrine Tumor Society (NANETS). Pancreas. 2017;46(6):715–731.
  • Mitchell WD, Findlay JM, Prescott RJ, et al. Bile acids in the diarrhoea of ileal resection. Gut. 1973 May;14(5):348–353.
  • BartoliniI, Bencini L, Risaliti M, et al. Current management of pancreatic neuroendocrine tumors: from demolitive surgery to observation. Gastroenterol Res. 2018;Pract.2018:9647247.
  • Panzuto F, Massironi S, Partelli S, et al. Gastro-entero-pancreatic neuroendocrine neoplasia: the rules for non-operative management. Surg Oncol. 2020;35:141–148.
  • Rinke A, Müller HH, Schade-Brittinger C, 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;27:4656–4663.
  • Caplin ME, Pavel M, Ćwikła JB, et al., Lanreotide in metastatic enteropancreatic neuroendocrine tumors. N Engl J Med. 2014;371(3): 224–233.
  • Modlin IM, Pavel M, Kidd M, et al. Review article: somatostatin analogues in the treatment of gastroenteropancreatic neuroendocrine (carcinoid) tumours. Aliment Pharmacol Ther. 2010;31(2):169–188.
  • Laskaratos FM, Armeni E, Shah H, et al. Predictors of antiproliferative effect of lanreotide autogel in advanced gastroenteropancreatic neuroendocrine neoplasms. Endocrine. 2020;67(1):233–242.
  • Ramage JK, Ahmed A, Ardill J, et al. Guidelines for the management of gastroenteropancreatic neuroendocrine (including carcinoid) tumours (NETs). Gut. 2012;61(1):6–32.
  • Kvols LK, Oberg KE, O’Dorisio TM, 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):657–666.
  • Wolin EM, Jarzab B, Eriksson B, 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 Devel Ther. 2015;9:5075–5078.
  • Pavel M, Valle JW, Eriksson B, et al. Antibes consensus conference participants. ENETS consensus guidelines for the standards of care in neuroendocrine neoplasms: systemic therapy - biotherapy and novel targeted agents. Neuroendocrinology. 2017;105(3):266–280.
  • Strosberg J, El-Haddad G, Wolin E, et al., Phase 3 trial of 1lu-dotatate for midgut neuroendocrine tumors. N Engl J Med. 2017;376(2): 125–135.
  • Diamantopoulos L, Laskaratos FM, Kalligeros M, et al. Antiproliferative effect of above-label doses of somatostatin analogs for the management of gastroenteropancreatic neuroendocrine tumors. Neuroendocrinology. 2020 Online ahead of print
  • Hofland LJ, Lamberts SW. The pathophysiological consequences of somatostatin receptor internalization and resistance. Endocr Rev. 2003;24(1):28–47.
  • Michael M, Garcia-Carbonero R, Weber MM, et al. The antiproliferative role of lanreotide in controlling growth of neuroendocrine tumors: a systematic review. Oncologist. 2017;22(3):272–285.
  • Lima-Martínez MM, López-Méndez G. Mangupli R: bradicardia sinusal inducida por octreotide en un varón con acromegalia. Endocrinol Nutr. 2013;60:e7–e9.
  • Dilger J, Rhoe E, Que F. Sprung J: octreotide-induced bradycardia and heart block during surgical resection of a carcinoid tumor. Anesth Analg. 2004;98:318–320.
  • Herrington AM, George KW. Moulds CC: octreotide-Induced Bradycardia. Pharmacotherapy. 1998;18(2):413–416.
  • Caplin M, Pavel M, Cwikla JB, et al. Antitumor effects of lanreotide for pancreatic and intestinal neuroendocrine tumors: the CLARINET open-label extension study. Endocr Relat Cancer. 2016;23(3):191–199.
  • Lamberti G, Faggiano A, Brighi N, et al. Nonconventional doses of somatostatin analogs in patients with progressing well-differentiated neuroendocrine tumor. J ClinEndocrinolMetab. 2020;105(1):dgz035.
  • Lamarca Α, McCallum L, Nuttal L, et al. Somatostatin analogue-induced pancreatic exocrine insufficiency in patients with neuroendocrine tumors: results of a prospective observational study. Expert Rev Gastroenterol Hepatol. 2018;12(7):723–727.
  • Rinzivillo M, De Felice I, Magi L, et al. Occurrence of exocrine pancreatic insufficiency in patients with advanced neuroendocrine tumors treated with somatostatin analogs. Pancreatology. 2020;20(5):875–879.
  • Plöckinger U, Dienemann D. Quabbe HJ: gastrointestinal side-effects of octreotide during long-term treatment of acromegaly. J Clin Endocrinol Metab. 1990;71(6):1658–1662.
  • Haugen B. Drugs that suppress TSH or cause central hypothyroidism. Best Pract Res Clin Endocrinol Metab. 2009;23(6):793–800.
  • Oberg K, Eriksson B. The role of interferons in the management of carcinoid tumours. Br J Haematol. 1991;79(Suppl 1):74–77.
  • Yao J, Guthrie K, Moran C, et al. SWOG S0518: phase III prospective randomized comparison of depot octreotide plus interferon alpha-2b versus depot octreotide plus bevacizumab (NSC #704865) in advanced, poor prognosis carcinoid patients (NCT00569127). J Clin Oncol. 2015;33(15_suppl):4004.
  • Mirvis E, Mandair D, Garcia-Hernandez J, et al. Role of interferon-alpha in patients with neuroendocrine tumors: a retrospective study. Anticancer Res. 2014;34(11):6601–6607.
  • Oberg K. Interferon in the management of neuroendocrine GEP-tumors: a review. Digestion. 2000;62(Suppl 1):92–97.
  • Faiss S, Pape UF, Böhmig M, et al.; International Lanreotide and Interferon Alfa Study Group. 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. J Clin Oncol. 2003;21:2689–2696.
  • Arnold R, Rinke A, Klose KJ, et al. Octreotide versus octreotide plus interferon-alpha in endocrine gastroenteropancreatic tumors: a randomized trial. Clin Gastroenterol Hepatol. 2005;3(8):761–771.
  • Pavel ME, Baum U, Hahn EG, et al. Efficacy and tolerability of pegylated IFN-alpha in patients with neuroendocrine gastroenteropancreatic carcinomas. J Interferon Cytokine Res. 2006;26(1):8–13.
  • Schaefer M, Capuron L, Friebe A, et al. Hepatitis C infection, antiviral treatment and mental health: a European expert consensus statement. J Hepatol. 2012;57(6):1379–1390.
  • Yao JC, Phan A, Hoff PM, et al. Targeting vascular endothelial growth factor in advanced carcinoid tumor: a random assignment phase II study of depot octreotide with bevacizumab and pegylated interferon alpha-2b. J Clin Oncol. 2008;26:1316–1323.
  • Kulke MH, O’ Dorisio T, Phan A, 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):705–714.
  • Kulke MH, Hörsch D, Caplin ME, et al., Telotristat ethyl, a tryptophan hydroxylase inhibitor for the treatment of carcinoid syndrome. J Clin Oncol. 2017;35(1): 14–23.
  • Pavel M, Hörsch D, Caplin M, et al. Telotristat etiprate for carcinoid syndrome: a single-arm, multicenter trial. J Clin Endocrinol Metab. 2015;100(4):1511–1519.
  • Pavel M, Gross DJ, Benavent M, et al., Telotristat ethyl in carcinoid syndrome: safety and efficacy in the TELECAST phase III trial. Endocr Relat Cancer. 2018;25(3): 309–322.
  • Strosberg J, Joish VN, Giacalone S, et al. TELEPRO: patient-reported carcinoid syndrome symptom improvement following initiation of telotristat ethyl in the real world. Oncologist. 2019 Nov;24(11):1446–1452.
  • Anthony LB, Kulke MH, Caplin ME, et al. Long-term safety experience with telotristat ethyl across five clinical studies in patients with carcinoid syndrome. Oncologist. 2019 Aug;24(8):e662–e670.
  • Raymond E, Dahan L, Raoul JL, et al., Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. N Engl J Med. 2011;364(6): 501–513.
  • Wiedmann MW, Mossner J. Safety and efficacy of sunitinib in patients with unresectable pancreatic neuroendocrine tumors. Clin Med Insights Oncol. 2012;6:381–393.
  • Pavel M, O’Toole D, Costa F, 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):172–185.
  • Yao JC, Shah MH, Ito T, et al., Everolimus for advanced pancreatic neuroendocrine tumors. N Engl J Med. 2011;364(6): 514–523.
  • Pavel ME, Hainsworth JD, Baudin E, et al., Everolimus plus octreotide long-acting repeatable for the treatment of advanced neuroendocrine tumours associated with carcinoid syndrome (RADIANT-2): a randomised, placebo-controlled, phase 3 study. Lancet. 2011;378(9808): 2005–2012.
  • Yao JC, Fazio N, Singh S, 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 3 study. Lancet. 2016;387(10022): 968–977.
  • Bainbridge HE, Larbi E, Middleton G. Symptomatic control of neuroendocrine tumours with everolimus. Horm Cancer. 2015;6(5–6):254–259.
  • Faivre S, Niccoli P, Castellano D, 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):339–343.
  • Trelinska J, Dachowska I, Kotulska K, et al. Complications of mammalian target of rapamycin inhibitor anticancer treatment among patients with tuberous sclerosis complex are common and occasionally life-threatening. Anticancer Drugs. 2015;26(4):437–442.
  • Koffas A, Dolman GE, Kennedy PT. Hepatitis B virus reactivation in patients treated with immunosuppressive drugs: a practical guide for clinicians. Clin Med (Lond). 2018;18(3):212–218.
  • Valle JW, Faivre S, Hubner RA, et al. Practical management of Sunitinib toxicities in the treatment of pancreatic neuroendocrine tumors. Cancer Treat Rev. 2014;40(10):1230–1238.
  • Funakoshi T. Shimada YJ: risk of hypothyroidism in patients with cancer treated with sunitinib: a systematic review and meta-analysis. Acta Oncol. 2013;52(4):691–702.
  • Cives M, Strosberg J. Radionuclide therapy for neuroendocrine tumors. Curr Oncol Rep. 2017;19(2):9.
  • Pool SE, Krenning E, Koning GA, et al. Preclinical and clinical studies of peptide receptor radionuclide therapy. Semin Nucl Med. 2010;40(3):209–218.
  • Kwekkeboom DJ, Kam BL, van Essen M, et al. Somatostatin-receptor-based imaging and therapy of gastroenteropancreatic neuroendocrine tumors. Endocr Relat Cancer. 2010;17(1):R53–73.
  • Severi S, Grassi I, Nicolini 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;10:551–557.
  • Bushnell DL Jr, O’ Dorisio TM, O’ Dorisio MS, et al. 90 Y-edotreotide for metastatic carcinoid refractory to octreotide. J Clin Oncol. 2010;28(10):1652–1659.
  • Strosberg J, Kunz PL, Hendifar A. Impact of liver tumour burden, alkaline phosphatase elevation, and target lesion size on treatment outcomes with 177 Lu-Dotatate: an analysis of the NETTER-1 study. Eur J Nucl Med Mol Imaging. 2020. Online ahead of print. doi:10.1007/s00259-020-04709-x
  • Severi S, Sansovini M, Ianniello A, 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):1955–1963.
  • Forrer F, Uusijärvi H, Storch D, et al. Treatment with 177Lu-DOTATOC of patients with relapse of neuroendocrine tumors after treatment with 90Y-DOTATOC. J Nucl Med. 2005;46(8):1310–1316.
  • Vaughan E, Machta J, Walker M, et al. Retreatment with peptide receptor radionuclide therapy in patients with progressing neuroendocrine tumours: efficacy and prognostic factors for response. Br J Radiol. 2018;91(1091):20180041.
  • Brabander T, Van der Zwan WA, Teunissen JJM, 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):4617–4624.
  • Bodei L, Kidd M, Paganelli G, et al. Long-term tolerability of PRRT in 807 patients with neuroendocrine tumours: the value and limitations of clinical factors. Eur J Nucl Med Mol Imaging. 2015;42(1):5–19.
  • Sabet A, Ezziddin K, Pape UF, et al. Long-term hematotoxicity after peptide receptor radionuclide therapy with 177Lu-octreotate. J Nucl Med. 2013;54(11):1857–1861.
  • Pencharz D, Walker M, Yalchin M, et al. Early efficacy of and toxicity from lutetium-177-DOTATATE treatment in patients with progressive metastatic NET. Nucl Med Commun. 2017;38(7):593–600. Jul.
  • Garcia-Carbonero R, Sorbye H, Baudin E, et al. ENETS consensus guidelines for high-grade gastroenteropancreatic neuroendocrine tumors and neuroendocrine carcinomas. Neuroendocrinology. 2016;103(2):186–194.
  • Welin S, Sorbye H, Sebjornsen S, et al. Clinical effect of temozolomide-based chemotherapy in poorly differentiated endocrine carcinoma after progression on first-line chemotherapy. Cancer. 2011;117(20):4617–4622.
  • Walenkamp AME, Sonke GS, Sleijfer DT. Clinical and therapeutic aspects of extrapulmonary small cell carcinoma. Cancer Treat Rev. 2009;35(3):228–236.
  • Tang LH, Basturk O, Sue JJ, et al. A practical approach to the classification of WHO grade 3 (G3) well-differentiated neuroendocrine tumor (WD-NET) and Poorly differentiated neuroendocrine carcinoma (PD-NEC) of the pancreas. Am J Surg Pathol. 2016;40(9):1192–1202.
  • Vélayoudom-Céphise FL, Duvillard P, Foucan L, et al. Are G3 ENETS neuroendocrine neoplasms heterogeneous? Endocr Relat Cancer. 2013;20(5):649–657.
  • Sorbye H, Welin S, Langer SW, et al. Predictive and prognostic factors for treatment and survival in 305 patients with advanced gastrointestinal neuroendocrine carcinoma (WHO G3): the NORDIC NEC study. Ann Oncol. 2013;24(1):152–160.vol. Article ID mds276.
  • Sonbol MB, Halfdanarson TR. Management of well-differentiated high-grade (g3) neuroendocrine tumors. Curr Treat Options Oncol. 2019;20(9):74.
  • Sorbye H, Grace Kong G, Simona Grozinsky-Glasberg S. PRRT in high-grade gastroenteropancreatic neuroendocrine neoplasms (WHO G3). Endocr Relat Cancer. 2020;27(3): R67–R77. Mar.
  • Turner NC, Strauss SJ, Sarker D, et al. Chemotherapy with 5-fluorouracil, cisplatin and streptozocin for neuroendocrine tumours. Br J Cancer. 2010;102(7):1106–1112.
  • JR S, RL F, Choi J, et al. First-line chemotherapy with capecitabine and temozolomide in patients with metastatic pancreatic endocrine carcinomas. Cancer. 2011;117(2):268–275.
  • Fine R, Gulati AP, Krantz BA, et al. Capecitabine and temozolomide (CAPTEM) for metastatic, well-differentiated neuroendocrine cancers: the Pancreas Center at Columbia University experience. Cancer Chemother Pharmacol. 2013;71(3):663–670.
  • Kulke MH, Stuart K, Enzinger PC, et al. Phase II study of temozolomide and thalidomide in patients with metastatic neuroendocrine tumors. J clin oncol. 2006;24(3):401–406.
  • Kulke MH, Stuart K, Earle CC, et al. A phase II study of temozolomide and bevacizumab in patients with advanced neuroendocrine tumors. J clin oncol. 2006;24(18):4044
  • Chan JA, Stuart K, Earle CC, et al. Prospective study of bevacizumab plus temozolomide in patients with advanced neuroendocrine tumors. J clin oncol. 2012;30(24):2963–2968.
  • Hadoux J, Malka D, Planchard D, et al. Post-first-line FOLFOX chemotherapy for grade 3 neuroendocrine carcinoma. Endocr Relat Cancer. 2015;22(3):289–298.
  • Hentic O, Hammel P, Couvelard A, et al. FOLFIRI regimen: an effective second-line chemotherapy after failure of etoposide-platinum combination in patients with neuroendocrine carcinomas grade 3. Endocr Relat Cancer. 2012;19(6):751–757.
  • Saif MW, Kaley K, Brennan M, et al. A retrospective study of capecitabine/temozolomide (CAPTEM) regimen in the treatment of metastatic pancreatic neuroendocrine tumors (pNETs) after failing previous therapy. J Pancreas. 2013;14(5):498–501.
  • Sahu Α JΜ, Lai-Kwon J, Thai A, et al. CAPTEM in metastatic well-differentiated intermediate to high grade neuroendocrine tumors: a single centre experience. J Oncol. 2019;2019:9032753.
  • Chatzellis E, Angelousi A, Daskalakis K, et al. Activity and safety of standard and prolonged capecitabine/temozolomide administration in patients with advanced neuroendocrine neoplasms. Neuroendocrinology. 2019;109(4):333–345.
  • Moertel CG, Kvols LK, O’Connell MJ, et al. Treatment of neuroendocrine carcinomas with combined etoposide and cisplatin. Evidence of major therapeutic activity in the anaplastic variants of these neoplasms. Cancer. 1991 Jul 15;68(2):227–232.
  • Pavel M, Baudin E, Couvelard A, et al. ENETS consensus guidelines for the management of patients with liver and other distant metastases from neuroendocrine neoplasms of foregut, midgut, hindgut, and unknown primary. Neuroendocrinology. 2012;95(2):157–176.
  • Eriksson J, Stalberg P, Nilsson A, et al. Surgery and radiofrequency ablation for treatment of liver metastases from midgut and foregut carcinoids and endocrine pancreatic tumors. World J Surg. 2008;32(5):930–938.
  • Livraghi T, Solbiati L, Meloni MF, et al. Treatment of focal liver tumors with percutaneous radio-frequency ablation: complications encountered in a multicenter study. Radiology. 2003;226:441–451.
  • Mulier S, Mulier P, Ni Y, et al. Complications of radiofrequency coagulation of liver tumours. Br J Surg. 2002;89(10):1206–1222.
  • de Baere T, Elias D, Dromain C, et al. Radiofrequency ablation of 100 hepatic metastases with a mean follow-up of more than 1 year. AJR Am J Roentgenol. 2000;175(6):1619–1625.
  • Gupta S. Intra-arterial liver-directed therapies for neuroendocrine hepatic metastases. Semin Intervent Radiol. 2013;30(1):28–38.
  • Davar J, Connolly HML, Caplin ME. Diagnosing and managing carcinoid heart disease in patients with neuroendocrine tumors: an expert statement. J Am Coll Cardiol. 2017;69(10):1288–1304.
  • Eriksson B, Oberg K. Summing up 15 years of somatostatin analog therapy in neuroendocrine tumors: future outlook. Ann Oncol. 1999;2:S31–8. 10Suppl.
  • Pavel ME, Singh S, Strosberg J, et al. Health-related quality of life for everolimus versus placebo in patients with advanced, non-functional, well-differentiated gastrointestinal or lung neuroendocrine tumours (RADIANT-4): a multicentre, randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2017;18(10):1411–1422.
  • Strosberg J, Wolin E, Chasen B, et al. Health-related quality of life in patients with progressive midgut neuroendocrine tumors treated with 177 Lu-Dotatate in the Phase III NETTER-1 Trial. J Clin Oncol. 2018 Sep 1;36(25):2578–2584.
  • Laskaratos FM, Caplin M. Treatment challenges in and outside a network setting: gastrointestinal neuroendocrine tumours. Eur J Surg Oncol. 2019 Jan;45(1):52–59.
  • Metz DC, Choi J, Jonathan Strosberg J, et al. A rationale for multidisciplinary care in treating neuroendocrine tumours. Curr Opin Endocrinol Diabetes Obes. 2012;19(4):306–313.
  • Magi L, Mazzuca F, Rinzivillo M, et al. Multidisciplinary management of neuroendocrine neoplasia: a real-world experience from a referral center. J Clin Med. 2019;8(6):910.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.