343
Views
5
CrossRef citations to date
0
Altmetric
Review

New and emerging systemic therapy options for well-differentiated gastroenteropancreatic neuroendocrine tumors

, & ORCID Icon
Pages 183-191 | Received 16 Apr 2019, Accepted 13 Nov 2019, Published online: 24 Nov 2019

References

  • 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 Oct;3(10):1335–1342.
  • Lee MR, Harris C, Baeg KJ, et al. Incidence trends of gastroenteropancreatic neuroendocrine tumors in the United States. Clin Gastroenterol Hepatol. 2019 Oct;17(11):2212–2217.e1. DOI:10.1016/j.cgh.2018.12.017. Epub 2018 Dec 20.
  • Lloyd RV, Osamura RY, Klèoppel G, et al. WHO classification of tumours of endocrine organs. 4th ed. Lyon, France: World Health Organization Classification of Tumours; 2017. p. 355.
  • Capurso G, Archibugi L, Delle Fave G. Molecular pathogenesis and targeted therapy of sporadic pancreatic neuroendocrine tumors. J Hepatobiliary Pancreat Sci. 2015 Aug;22(8):594–601.
  • Akerström G, Hellman P. Surgery on neuroendocrine tumours. Best Pract Res Clin Endocrinol Metab. 2007 Mar;21(1):87–109.
  • Frilling A, Clift AK. Therapeutic strategies for neuroendocrine liver metastases. Cancer. 2015 Apr;121(8):1172–1186.
  • Saxena A, Chua TC, Perera M, et al. Surgical resection of hepatic metastases from neuroendocrine neoplasms: a systematic review. Surg Oncol. 2012 Sep;21(3):e131–e41.
  • Zhang XF, Beal EW, Chakedis J, et al. Early recurrence of neuroendocrine liver metastasis after curative hepatectomy: risk factors, prognosis, and treatment. J Gastrointest Surg. 2017 Nov;21(11):1821–1830.
  • Sadot E, Reidy-Lagunes DL, Tang LH, et al. Observation versus resection for small asymptomatic pancreatic neuroendocrine tumors: a matched case-control study. Ann Surg Oncol. 2016 Apr;23(4):1361–1370.
  • Delle Fave G, O’Toole D, Sundin A, et al. ENETS consensus guidelines update for gastroduodenal neuroendocrine neoplasms. Neuroendocrinology. 2016;103(2):119–124.
  • Ramage JK, De Herder WW, Delle Fave G, et al. ENETS consensus guidelines update for colorectal neuroendocrine neoplasms. Neuroendocrinology. 2016;103(2):139–143.
  • Nishikawa H, Kita R, Kimura T, et al. Transcatheter arterial embolic therapies for hepatocellular carcinoma: a literature review. Anticancer Res. 2014 Dec;34(12):6877–6886.
  • Mohan H, Nicholson P, Winter DC, et al. Radiofrequency ablation for neuroendocrine liver metastases: a systematic review. J Vasc Interv Radiol. 2015 Jul;26(7):935–42.e1.
  • Elf AK, Andersson M, Henrikson O, et al. Radioembolization versus bland embolization for hepatic metastases from small intestinal neuroendocrine tumors: short-term results of a randomized clinical trial. World J Surg. 2018 2;42(2):506–513.
  • Bloomston M, Al-Saif O, Klemanski D, et al. Hepatic artery chemoembolization in 122 patients with metastatic carcinoid tumor: lessons learned. J Gastrointest Surg. 2007 Mar;11(3):264–271.
  • Kennedy AS, Dezarn WA, McNeillie P, Coldwell D, Nutting C, Carter D, et al. Radioembolization for unresectable neuroendocrine hepatic metastases using resin 90Y-microspheres: early results in 148 patients. Am J Clin Oncol. 2008 Jun;31(3):271–279.
  • Minh D D, Chapiro J, Gorodetski B, et al. Intra-arterial therapy of neuroendocrine tumour liver metastases: comparing conventional TACE, drug-eluting beads TACE and yttrium-90 radioembolisation as treatment options using a propensity score analysis model. Eur Radiol. 2017 Dec;27(12):4995–5005.
  • Reubi JC. Somatostatin and other Peptide receptors as tools for tumor diagnosis and treatment. Neuroendocrinology. 2004;80(Suppl 1):51–56.
  • Møller LN, Stidsen CE, Hartmann B, et al. Somatostatin receptors. Biochim Biophys Acta. 2003 Sep;1616(1):1–84.
  • Narayanan S, Kunz PL. Role of somatostatin analogues in the treatment of neuroendocrine tumors. J Natl Compr Canc Netw. 2015 Jan;13(1):109–117. quiz 17.
  • Merola E, Panzuto F, Delle Fave G. Antiproliferative effect of somatostatin analogs in advanced gastro-entero-pancreatic neuroendocrine tumors: a systematic review and meta-analysis. Oncotarget. 2017 Jul;8(28):46624–46634.
  • 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 Oct;27(28):4656–4663.
  • Caplin ME, Pavel M, Ćwikła JB, et al. Lanreotide in metastatic enteropancreatic neuroendocrine tumors. N Engl J Med. 2014 Jul;371(3):224–233.
  • Alonso-Gordoa T, Capdevila J, Grande E. GEP-NETs update: biotherapy for neuroendocrine tumours. Eur J Endocrinol. 2015 Jan;172(1):R31–R46.
  • Boerlin V, van der Hoek J, Beglinger C, et al. New insights on SOM230, a universal somatostatin receptor ligand. J Endocrinol Invest. 2003;26(8 Suppl):14–16.
  • Gadelha MR, Bronstein MD, Brue T, et al. Pasireotide versus continued treatment with octreotide or lanreotide in patients with inadequately controlled acromegaly (PAOLA): a randomised, phase 3 trial. Lancet Diabetes Endocrinol. 2014 Nov;2(11):875–884.
  • Hofland LJ, Lamberts SW. The pathophysiological consequences of somatostatin receptor internalization and resistance. Endocr Rev. 2003 Feb;24(1):28–47.
  • 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 Oct;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;9:5075–5086.
  • Vitale G, Dicitore A, Sciammarella C, et al. Pasireotide in the treatment of neuroendocrine tumors: a review of the literature. Endocr Relat Cancer. 2018 6;25(6):R351–R64.
  • Kulke MH, Hörsch D, Caplin ME, et al. Telotristat ethyl, a tryptophan hydroxylase inhibitor for the treatment of carcinoid syndrome. J Clin Oncol. 2017 Jan;35(1):14–23.
  • Pavel M, Gross DJ, Benavent M, et al. Telotristat ethyl in carcinoid syndrome: safety and efficacy in the TELECAST phase 3 trial. Endocr Relat Cancer. 2018 3;25(3):309–322.
  • Lamarca A, Barriuso J, McNamara MG, et al. Telotristat ethyl: a new option for the management of carcinoid syndrome. Expert Opin Pharmacother. 2016 Dec;17(18):2487–2498.
  • Mohamed A, Strosberg JR. Medical management of gastroenteropancreatic neuroendocrine tumors: current strategies and future advances. J Nucl Med. 2019 Feb;60:721–727.
  • Broder LE, Carter SK. Pancreatic islet cell carcinoma. II. Results of therapy with streptozotocin in 52 patients. Ann Intern Med. 1973 Jul;79(1):108–118.
  • Moertel CG, Hanley JA, Johnson LA. Streptozocin alone compared with streptozocin plus fluorouracil in the treatment of advanced islet-cell carcinoma. N Engl J Med. 1980 Nov;303(21):1189–1194.
  • Cives M, Ghayouri M, Morse B, et al. Analysis of potential response predictors to capecitabine/temozolomide in metastatic pancreatic neuroendocrine tumors. Endocr Relat Cancer. 2016 9;23(9):759–767.
  • Kunz PL, Catalano PJ, Nimeiri H, et al. A randomized study of temozolomide or temozolomide and capecitabine in patients with advanced pancreatic neuroendocrine tumors: A trial of the ECOG-ACRIN Cancer Research Group (E2211). J Clin Oncol. 2018 May 20;36(15_suppl):4004.
  • Spada F, Antonuzzo L, Marconcini R, et al. Oxaliplatin-based chemotherapy in advanced neuroendocrine tumors: clinical outcomes and preliminary correlation with biological factors. Neuroendocrinology. 2016;103(6):806–814.
  • 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.
  • Milione M, Maisonneuve P, Spada F, et al. The clinicopathologic heterogeneity of Grade 3 gastroenteropancreatic neuroendocrine neoplasms: morphological differentiation and proliferation identify different prognostic categories. Neuroendocrinology. 2017;104(1):85–93.
  • 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 Jan;24(1):152–160.
  • Coriat R, Walter T, Terris B, et al. Gastroenteropancreatic well-differentiated Grade 3 neuroendocrine tumors: review and position statement. Oncologist. 2016 10;21(10):1191–1199.
  • Fazio N, Milione M. Heterogeneity of grade 3 gastroenteropancreatic neuroendocrine carcinomas: new insights and treatment implications. Cancer Treat Rev. 2016 Nov;50:61–67.
  • Yao JC, Phan AT, Chang DZ, et al. Efficacy of RAD001 (everolimus) and octreotide LAR in advanced low- to intermediate-grade neuroendocrine tumors: results of a phase II study. J Clin Oncol. 2008 Sep;26(26):4311–4318.
  • 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 Dec;378(9808):2005–2012.
  • Yao JC, Shah MH, Ito T, et al. Everolimus for advanced pancreatic neuroendocrine tumors. N Engl J Med. 2011 Feb;364(6):514–523.
  • JC Y, 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 Mar;387(10022):968–977.
  • Bainbridge HE, Larbi E, Middleton G. Symptomatic control of neuroendocrine tumours with everolimus. Horm Cancer. 2015 Dec;6(5–6):254–259.
  • Kulke MH, Bergsland EK, Yao JC. Glycemic control in patients with insulinoma treated with everolimus. N Engl J Med. 2009 Jan;360(2):195–197.
  • Pavel ME, Singh S, Strosberg JR, 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 10;18(10):1411–1422.
  • Grillo F, Florio T, Ferraù F, et al. Emerging multitarget tyrosine kinase inhibitors in the treatment of neuroendocrine neoplasms. Endocr Relat Cancer. 2018 9;25(9):R453–R66.
  • Le Tourneau C, Raymond E, Faivre S. Sunitinib: a novel tyrosine kinase inhibitor. A brief review of its therapeutic potential in the treatment of renal carcinoma and gastrointestinal stromal tumors (GIST). Ther Clin Risk Manag. 2007 Jun;3(2):341–348.
  • Raymond E, Dahan L, Raoul JL, et al. Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. N Engl J Med. 2011 Feb;364(6):501–513.
  • Vinik A, Bottomley A, Korytowsky B, et al. Patient-reported outcomes and quality of life with sunitinib versus placebo for pancreatic neuroendocrine tumors: results from an international Phase III trial. Target Oncol. 2016 12;11(6):815–824.
  • Kulke MH, Lenz HJ, Meropol NJ, et al. Activity of sunitinib in patients with advanced neuroendocrine tumors. J Clin Oncol. 2008 Jul;26(20):3403–3410.
  • Yao JC, Guthrie KA, Moran C, 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 May;35(15):1695–1703.
  • Mitry E, Walter T, Baudin E, et al. Bevacizumab plus capecitabine in patients with progressive advanced well-differentiated neuroendocrine tumors of the gastro-intestinal (GI-NETs) tract (BETTER trial)–a phase II non-randomised trial. Eur J Cancer. 2014 Dec;50(18):3107–3115.
  • Berruti A, Fazio N, Ferrero A, et al. Bevacizumab plus octreotide and metronomic capecitabine in patients with metastatic well-to-moderately differentiated neuroendocrine tumors: the XELBEVOCT study. BMC Cancer. 2014;14:184.
  • Ducreux M, Dahan L, Smith D, et al. Bevacizumab combined with 5-FU/streptozocin in patients with progressive metastatic well-differentiated pancreatic endocrine tumours (BETTER trial)–a phase II non-randomised trial. Eur J Cancer. 2014 Dec;50(18):3098–3106.
  • Abdel-Rahman O, Fouad M. Bevacizumab-based combination therapy for advanced gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs): a systematic review of the literature. J Cancer Res Clin Oncol. 2015 Feb;141(2):295–305.
  • Chan JA, Stuart K, Earle CC, et al. Prospective study of bevacizumab plus temozolomide in patients with advanced neuroendocrine tumors. J Clin Oncol. 2012 Aug;30(24):2963–2968.
  • Crabtree JS. Clinical and preclinical advances in gastroenteropancreatic neuroendocrine tumor therapy. Front Endocrinol (Lausanne). 2017;8:341.
  • Ranpura V, Hapani S, Wu S. Treatment-related mortality with bevacizumab in cancer patients: a meta-analysis. JAMA. 2011 Feb;305(5):487–494.
  • Ahn HK, Choi JY, Kim KM, et al. Phase II study of pazopanib monotherapy in metastatic gastroenteropancreatic neuroendocrine tumours. Br J Cancer. 2013 Sep;109(6):1414–1419.
  • Phan AT, Halperin DM, Chan JA, et al. Pazopanib and depot octreotide in advanced, well-differentiated neuroendocrine tumours: a multicentre, single-group, phase 2 study. Lancet Oncol. 2015 Jun;16(6):695–703.
  • Grande E, Capdevila J, Castellano D, et al. Pazopanib in pretreated advanced neuroendocrine tumors: a phase II, open-label trial of the Spanish task force group for neuroendocrine tumors (GETNE). Ann Oncol. 2015 Sep;26(9):1987–1993.
  • Chan JA, Faris JE, Murphy JE, et al. Phase II trial of cabozantinib in patients with carcinoid and pancreatic neuroendocrine tumors (pNET). J Clin Oncol. 2017 Feb 1;35(4_suppl):228.
  • Modlin IM, Pavel M, Kidd M, et al. Review article: somatostatin analogues in the treatment of gastroenteropancreatic neuroendocrine (carcinoid) tumours. Aliment Pharmacol Ther. 2010 Jan;31(2):169–188.
  • Brabander T, van der Zwan WA, Teunissen JJM, et al. Long-term efficacy, survival, and safety of [177Lu-DOTA°,Tyr3]octreotate in patients with gastroenteropancreatic and bronchial neuroendocrine tumors. Clin Cancer Res. 2017 Aug;23(16):4617–4624.
  • Kim SJ, Pak K, Koo PJ, 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 Dec;42(13):1964–1970.
  • Strosberg J, El-Haddad G, Wolin E, et al. Phase 3 trial of 177Lu-dotatate for midgut neuroendocrine tumors. N Engl J Med. 2017 1;376(2):125–135.
  • Strosberg J, Wolin E, Chasen B, et al. Health-related quality of life in patients with progressive midgut neuroendocrine tumors treated with 177Lu-Dotatate in the phase III NETTER-1 trial. J Clin Oncol. 2018 Sep;36(25):2578–2584.
  • Cives M, Strosberg J. Radionuclide therapy for neuroendocrine tumors. Curr Oncol Rep. 2017 Feb;19(2):9.
  • Bodei L, Kwekkeboom DJ, Kidd M, et al. Radiolabeled somatostatin analogue therapy of gastroenteropancreatic cancer. Semin Nucl Med. 2016 May;46(3):225–238.
  • Kratochwil C, Giesel FL, Bruchertseifer F, et al. 213Bi-DOTATOC receptor-targeted alpha-radionuclide therapy induces remission in neuroendocrine tumours refractory to beta radiation: a first-in-human experience. Eur J Nucl Med Mol Imaging. 2014 Nov;41(11):2106–2119.
  • Falconi M, Eriksson B, Kaltsas G, 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):153–171.
  • Ballal S, Yadav MP, Damle NA, et al. Concomitant 177Lu-DOTATATE and capecitabine therapy in patients with advanced neuroendocrine tumors: a long-term-outcome, toxicity, survival, and quality-of-life study. Clin Nucl Med. 2017 Nov;42(11):e457–e66.
  • Yordanova A, Wicharz MM, Mayer K, et al. The role of adding somatostatin analogues to peptide receptor radionuclide therapy as a combination and maintenance therapy. Clin Cancer Res. 2018 Oct;24(19):4672–4679.
  • Nonnekens J, van Kranenburg M, Beerens CE, et al. Potentiation of peptide receptor radionuclide therapy by the PARP inhibitor Olaparib. Theranostics. 2016;6(11):1821–1832.
  • Kölby L, Persson G, Franzén S, et al. Randomized clinical trial of the effect of interferon alpha on survival in patients with disseminated midgut carcinoid tumours. Br J Surg. 2003 Jun;90(6):687–693.
  • Cunha LL, Marcello MA, Rocha-Santos V, et al. Immunotherapy against endocrine malignancies: immune checkpoint inhibitors lead the way. Endocr Relat Cancer. 2017 12;24(12):T261–T81.
  • Kim ST, Ha SY, Lee S, et al. The impact of PD-L1 expression in patients with metastatic GEP-NETs. J Cancer. 2016;7(5):484–489.
  • Ott PA, Bang YJ, Piha-Paul SA, et al. T-cell-inflamed gene-expression profile, programmed death ligand 1 expression, and tumor mutational burden predict efficacy in patients treated with pembrolizumab across 20 cancers: KEYNOTE-028. J Clin Oncol. 2019 Feb;37(4):318–327.
  • Weber MM, Fottner C. Immune checkpoint inhibitors in the treatment of patients with neuroendocrine neoplasia. Oncol Res Treat. 2018;41(5):306–312.
  • Jiao Y, Shi C, Edil BH, et al. DAXX/ATRX, MEN1, and mTOR pathway genes are frequently altered in pancreatic neuroendocrine tumors. Science. 2011 Mar;331(6021):1199–1203.
  • Scarpa A, Chang DK, Nones K, et al. Whole-genome landscape of pancreatic neuroendocrine tumours. Nature. 2017 3;543(7643):65–71.
  • Marinoni I, Kurrer AS, Vassella E, et al. Loss of DAXX and ATRX are associated with chromosome instability and reduced survival of patients with pancreatic neuroendocrine tumors. Gastroenterology. 2014 Feb;146(2):453–60.e5.
  • Singhi AD, Liu TC, Roncaioli JL, et al. Alternative lengthening of telomeres and loss of DAXX/ATRX expression predicts metastatic disease and poor survival in patients with pancreatic neuroendocrine tumors. Clin Cancer Res. 2017 Jan;23(2):600–609.
  • Mafficini A, Scarpa A. Genetics and epigenetics of gastroenteropancreatic neuroendocrine neoplasms. Endocr Rev. 2019 4;40(2):506–536.
  • Heaphy CM, de Wilde RF, Jiao Y, et al. Altered telomeres in tumors with ATRX and DAXX mutations. Science. 2011 Jul;333(6041):425.
  • Francis JM, Kiezun A, Ramos AH, et al. Somatic mutation of CDKN1B in small intestine neuroendocrine tumors. Nat Genet. 2013 Dec;45(12):1483–1486.
  • Karpathakis A, Dibra H, Pipinikas C, et al. Prognostic impact of novel molecular subtypes of small intestinal neuroendocrine tumor. Clin Cancer Res. 2016 Jan;22(1):250–258.
  • Walter D, Harter PN, Battke F, et al. Genetic heterogeneity of primary lesion and metastasis in small intestine neuroendocrine tumors. Sci Rep. 2018 2;8(1):3811.
  • Vernieri C, Pusceddu S, de Braud F. Impact of metformin on systemic metabolism and survival of patients with advanced pancreatic neuroendocrine tumors. Front Oncol. 2019 September 20;9(902).
  • Cives M, Strosberg J. Treatment strategies for metastatic neuroendocrine tumors of the gastrointestinal tract. Curr Treat Options Oncol. 2017 3;18(3):14.
  • Trendle MC, Moertel CG, Kvols LK. Incidence and morbidity of cholelithiasis in patients receiving chronic octreotide for metastatic carcinoid and malignant islet cell tumors. Cancer. 1997 Feb;79(4):830–834.
  • Cuyle PJ, Prenen H. Practical management of toxicities associated with targeted therapies for advanced gastroenteropancreatic neuroendocrine tumors. Ann Gastroenterol. 2018;31(2):140–150. Mar-Apr.

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.