11,528
Views
36
CrossRef citations to date
0
Altmetric
Drug Evaluation

Everolimus in the treatment of neuroendocrine tumors: efficacy, side-effects, resistance, and factors affecting its place in the treatment sequence

, &
Pages 909-928 | Received 16 Mar 2018, Accepted 09 May 2018, Published online: 24 May 2018

References

  • Ito T, Hijioka S, Masui T, et al. Advances in the diagnosis and treatment of pancreatic neuroendocrine neoplasms in Japan. J Gastroenterol. 2017;52:9–18.
  • 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;3:1335–1342.
  • Yao JC, Hassan M, Phan A, et al. One hundred years after “carcinoid”: epidemiology of and prognostic factors for neuroendocrine tumors in 35,825 cases in the United States. J Clin Oncol. 2008;26:3063–3072.
  • Modlin IM, Lye KD, Kidd M. A 5-decade analysis of 13,715 carcinoid tumors. Cancer. 2003;97:934–959.
  • Laplante M, Sabatini DM. mTOR signaling in growth control and disease. Cell. 2012;149:274–293.
  • Wolin EM. PI3K/Akt/mTOR pathway inhibitors in the therapy of pancreatic neuroendocrine tumors. Cancer Lett. 2013;335:1–8.
  • Shida T, Kishimoto T, Furuya M, et al. Expression of an activated mammalian target of rapamycin (mTOR) in gastroenteropancreatic neuroendocrine tumors. Cancer Chemother Pharmacol. 2010;65:889–893.
  • Righi L, Volante M, Rapa I, et al. Mammalian target of rapamycin signaling activation patterns in neuroendocrine tumors of the lung. Endocr Relat Cancer. 2010;17:977–987.
  • Qian ZR, Ter-Minassian M, Chan JA, et al. Prognostic significance of MTOR pathway component expression in neuroendocrine tumors. J Clin Oncol. 2013;31:3418–3425.
  • Kasajima A, Pavel M, Darb-Esfahani S, et al. mTOR expression and activity patterns in gastroenteropancreatic neuroendocrine tumours. Endocr Relat Cancer. 2011;18:181–192.
  • Komori Y, Yada K, Ohta M, et al. Mammalian target of rapamycin signaling activation patterns in pancreatic neuroendocrine tumors. J Hepatobiliary Pancreat Sci. 2014;21:288–295.
  • Missiaglia E, Dalai I, Barbi S, et al. Pancreatic endocrine tumors: expression profiling evidences a role for AKT-mTOR pathway. J Clin Oncol. 2010;28:245–255.
  • Casciano R, Chulikavit M, Perrin A, et al. Cost-effectiveness of everolimus vs sunitinib in treating patients with advanced, progressive pancreatic neuroendocrine tumors in the United States. J Med Econ. 2012;15(Suppl 1):55–64.
  • Perren A, Komminoth P, Saremaslani P, et al. Mutation and expression analyses reveal differential subcellular compartmentalization of PTEN in endocrine pancreatic tumors compared to normal islet cells. Am J Pathol. 2000;157:1097–1103.
  • Jiao Y, Shi C, Edil BH, et al. DAXX/ATRX, MEN1, and mTOR pathway genes are frequently altered in pancreatic neuroendocrine tumors. Science. 2011;331:1199–1203.
  • Scarpa A, Chang DK, Nones K, et al. Whole-genome landscape of pancreatic neuroendocrine tumours. Nature. 2017;543:65–71.
  • Grozinsky-Glasberg S, Franchi G, Teng M, et al. Octreotide and the mTOR inhibitor RAD001 (everolimus) block proliferation and interact with the Akt-mTOR-p70S6K pathway in a neuro-endocrine tumour cell Line. Neuroendocrinology. 2008;87:168–181.
  • Chan J, Kulke M. Targeting the mTOR signaling pathway in neuroendocrine tumors. Curr Treat Options Oncol. 2014;15:365–379.
  • Yang H, Rudge DG, Koos JD, et al. mTOR kinase structure, mechanism and regulation. Nature. 2013;497:217–223.
  • 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;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;378:2005–2012.
  • Yao JC, Shah MH, Ito T, et al. Everolimus for advanced pancreatic neuroendocrine tumors. N Engl J Med. 2011;364:514–523.
  • 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;387:968–977.
  • Raymond E, Dahan L, Raoul JL, et al. Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. N Engl J Med. 2011;364:501–513.
  • Caplin ME, Pavel M, Cwikla J, et al. Lanreotide in metastatic enteropancreatic neuroendocrine tumors. N Engl J Med. 2014;371:224–233.
  • Strosberg J, El-Haddad G, Wolin E, et al. Phase 3 Trial of (177)Lu-Dotatate for midgut neuroendocrine tumors. N Engl J Med. 2017;376:125–135.
  • Kwekkeboom DJ, de Herder WW, Kam BL, et al. Treatment with the radiolabeled somatostatin analog [177 Lu-DOTA 0,Tyr3]octreotate: toxicity, efficacy, and survival. J Clin Oncol. 2008;26:2124–2130.
  • Fidelman N, Kerlan RK Jr., Hawkins RA, et al. Radioembolization with (90)Y glass microspheres for the treatment of unresectable metastatic liver disease from chemotherapy-refractory gastrointestinal cancers: final report of a prospective pilot study. J Gastrointest Oncol. 2016;7:860–874.
  • Benson AB III, Geschwind JF, Mulcahy MF, et al. Radioembolisation for liver metastases: results from a prospective 151 patient multi-institutional phase II study. Eur J Cancer. 2013;49:3122–3130.
  • Kulke MH, Horsch D, Caplin ME, et al. Telotristat ethyl, a tryptophan hydroxylase inhibitor for the treatment of carcinoid syndrome. J Clin Oncol. 2017;35:14–23.
  • Vinik AI, Wolin EM, Liyanage N, et al. Evaluation of lanreotide depot/autogel efficacy and safety as a carcinoid syndrome treatment(ELECT): A randomized, double-blind, placebo-controlled trial. Endocr Pract. 2016;22:1068–1080.
  • Bosman FT, Carneiro F, Hruban RH, et al. WHO World Health Organization classification of tumors and genetics of the digestive system. Lyon, France: IARC press; 2010.
  • WHO classification of tumours of the lung, pleura, thymus and heart. Lyon, France: International Agency for Research on Cancer; 2015.
  • 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;104:764–777.
  • Luo G, Javed A, Strosberg JR, et al. Modified staging classification for pancreatic neuroendocrine tumors on the basis of the American Joint Committee on Cancer and European Neuroendocrine Tumor Society Systems. J Clin Oncol. 2017;35:274–280.
  • Morin E, Cheng S, Mete O, et al. Hormone profiling, WHO 2010 grading, and AJCC/UICC staging in pancreatic neuroendocrine tumor behavior. Cancer Med. 2013;2:701–711.
  • 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):152–260.
  • 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):84–93.
  • Heetfeld M, Chougnet CN, Olsen IH, et al. Characteristics and treatment of patients with G3 gastroenteropancreatic neuroendocrine neoplasms. Endocr Relat Cancer. 2015;22:657–664.
  • Hijioka S, Hosoda W, Matsuo K, et al. Rb Loss and KRAS mutation are predictors of the response to platinum-based chemotherapy in pancreatic neuroendocrine neoplasm with Grade 3: A Japanese multicenter pancreatic NEN-G3 study. Clin Cancer Res. 2017;23:4625–4632.
  • Cho MY, Kim JM, Sohn JH, et al. Current trends of the incidence and pathological diagnosis of gastroenteropancreatic neuroendocrine tumors (GEP-NETs) in Korea 2000–2009: multicenter study. Cancer Res Treat. 2012;44:157–165.
  • Hijioka S, Hosoda W, Mizuno N, et al. Does the WHO 2010 classification of pancreatic neuroendocrine neoplasms accurately characterize pancreatic neuroendocrine carcinomas? J Gastroenterol. 2015;50:564–572.
  • 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;40:1192–1202.
  • Tang LH, Untch BR, Reidy DL, et al. Well-differentiated neuroendocrine tumors with a morphologically apparent high-grade component: a pathway distinct from poorly differentiated neuroendocrine carcinomas. Clin Cancer Res. 2016;22:1011–1017.
  • Basturk O, Yang Z, Tang LH, et al. The high-grade (WHO G3) pancreatic neuroendocrine tumor category is morphologically and biologically heterogenous and includes both well differentiated and poorly differentiated neoplasms. Am J Surg Pathol. 2015;39:683–690.
  • WHO classification of tumours of endocrine organs. 4th ed. Vol. 10, Lloyd RV, Osamura RY, Kloppel G, Rosai J, editors. Lyon, France: Agency for Research on Cancer; 2017.
  • Haug AR, Cindea-Drimus R, Auernhammer CJ, et al. The role of 68Ga-DOTATATE PET/CT in suspected neuroendocrine tumors. J Nucl Med. 2012;53:1686–1692.
  • Haug AR, Cindea-Drimus R, Auernhammer CJ, et al. Neuroendocrine tumor recurrence: diagnosis with 68Ga-DOTATATE PET/CT. Radiology. 2014;270:517–525.
  • Deppen SA, Liu E, Blume JD, et al. Safety and efficacy of 68Ga-DOTATATE PET/CT for diagnosis, staging, and treatment management of neuroendocrine tumors. J Nucl Med. 2016;57:708–714.
  • Has Simsek D, Kuyumcu S, Turkmen C, et al. Can complementary 68Ga-DOTATATE and 18F-FDG PET/CT establish the missing link between histopathology and therapeutic approach in gastroenteropancreatic neuroendocrine tumors? J Nucl Med. 2014;55:1811–1817.
  • Bahri H, Laurence L, Edeline J, et al. High prognostic value of 18F-FDG PET for metastatic gastroenteropancreatic neuroendocrine tumors: a long-term evaluation. J Nucl Med. 2014;55:1786–1790.
  • Ma XM, Blenis J. Molecular mechanisms of mTOR-mediated translational control. Nat Rev Mol Cell Biol. 2009;10:307–318.
  • Motzer RJ, Escudier B, Oudard S, et al. Efficacy of everolimus in advanced renal cell carcinoma: a double-blind, randomised, placebo-controlled phase III trial. Lancet. 2008;372:449–456.
  • Krueger DA, Care MM, Holland K, et al. Everolimus for subependymal giant-cell astrocytomas in tuberous sclerosis. N Engl J Med. 2010;363:1801–1811.
  • Dantal J. Everolimus: preventing organ rejection in adult kidney transplant recipients. Expert Opin Pharmacother. 2012;13:767–778.
  • Baselga J, Campone M, Piccart M, et al. Everolimus in postmenopausal hormone-receptor-positive advanced breast cancer. N Engl J Med. 2012;366:520–529.
  • Kirchner GI, Meier-Wiedenbach I, Manns MP. Clinical pharmacokinetics of everolimus. Clin Pharmacokinet. 2004;43:83–95.
  • O’Donnell A, Faivre S, Burris HA III, et al. Phase I pharmacokinetic and pharmacodynamic study of the oral mammalian target of rapamycin inhibitor everolimus in patients with advanced solid tumors. J Clin Oncol. 2008;26:1588–1595.
  • Pavel ME, Baudin E, Oberg KE, et al. Efficacy of everolimus plus octreotide LAR in patients with advanced neuroendocrine tumor and carcinoid syndrome: final overall survival from the randomized, placebo-controlled phase 3 RADIANT-2 study. Ann Oncol. 2017;28(7):1569–1575.
  • Fazio N, Granberg D, Grossman A, et al. Everolimus plus octreotide long-acting repeatable in patients with advanced lung neuroendocrine tumors: analysis of the phase 3, randomized, placebo-controlled RADIANT-2 study. Chest. 2013;143:955–962.
  • Castellano D, Bajetta E, Panneerselvam A, 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;18:46–53.
  • Anthony LB, Pavel ME, Hainsworth JD, 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;102:18–25.
  • Strosberg JR, Yao JC, Bajetta E, et al. Efficacy of octreotide long-acting repeatable in neuroendocrine tumors: RADIANT-2 placebo arm post hoc analysis. Endocr Relat Cancer. 2015;22:933–940.
  • Pavel ME, Becerra C, Grosch K, et al. Effect of everolimus on the pharmacokinetics of octreotide long-acting repeatable in patients with advanced neuroendocrine tumors: an analysis of the randomized phase III RADIANT-2 trial. Clin Pharmacol Ther. 2017;101:462–468.
  • Yao JC, Pavel M, Lombard-Bohas C, 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;34:3906–3913.
  • Ito T, Okusaka T, Ikeda M, et al. Everolimus for advanced pancreatic neuroendocrine tumours: a subgroup analysis evaluating Japanese patients in the RADIANT-3 trial. Jpn J Clin Oncol. 2012;42:903–911.
  • Lombard-Bohas C, Yao JC, Hobday T, 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;44:181–189.
  • Singh S, Carnaghi C, Buzzoni R, et al. Everolimus in neuroendocrine tumors of the gastrointestinal tract and unknown primary. Neuroendocrinology. 2018;106(3):211–220.
  • Fazio N, Buzzoni R, Delle Fave G, et al. Everolimus in advanced, progressive, well-differentiated, non-functional neuroendocrine tumors: RADIANT-4 lung subgroup analysis. Cancer Sci. 2018;109:174–181.
  • Buzzoni R, Carnaghi C, Strosberg J, et al. Impact of prior therapies on everolimus activity: an exploratory analysis of RADIANT-4. Onco Targets Ther. 2017;10:5013–5030.
  • 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;18:1411–1422.
  • Yao JC, Lombard-Bohas C, Baudin E, et al. Daily oral everolimus activity in patients with metastatic pancreatic neuroendocrine tumors after failure of cytotoxic chemotherapy: a phase II trial. J Clin Oncol. 2010;28:69–76.
  • Bajetta E, Catena L, Fazio N, et al. Everolimus in combination with octreotide long-acting repeatable in a first-line setting for patients with neuroendocrine tumors: an ITMO group study. Cancer. 2014;120:2457–2463.
  • Bajetta E, Catena L, Pusceddu S, et al. Everolimus in combination with Octreotide LAR in first line setting for patients with neuroendocrine tumors: a 5-years update. Neuroendocrinology. 2017. In press.
  • Kulke MH, Ruszniewski P, Van Cutsem E, et al. A randomized, open-label, phase 2 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;28:1309–1315.
  • Ferolla P, Brizzi MP, Meyer T, 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 2 trial. Lancet Oncol. 2017;18:1652–1664.
  • Chan JA, Ryan DP, Zhu AX, et al. Phase I study of pasireotide (SOM 230) and everolimus (RAD001) in advanced neuroendocrine tumors. Endocr Relat Cancer. 2012;19:615–623.
  • Dasari A, Phan A, Gupta S, et al. Phase I study of the anti-IGF1R antibody cixutumumab with everolimus and octreotide in advanced well-differentiated neuroendocrine tumors. Endocr Relat Cancer. 2015;22:431–441.
  • Kim HS, Shaib WL, Zhang C, et al. Phase 1b study of pasireotide, everolimus, and selective internal radioembolization therapy for unresectable neuroendocrine tumors with hepatic metastases. Cancer. 2018;124(9):1992–2000.
  • O’Reilly T, McSheehy PM. Biomarker development for the clinical activity of the mTOR inhibitor Everolimus (RAD001): processes, limitations, and further proposals. Transl Oncol. 2010;3:65–79.
  • von Wichert G, Jehle PM, Hoeflich A, et al. Insulin-like growth factor-I is an autocrine regulator of chromogranin A secretion and growth in human neuroendocrine tumor cells. Cancer Res. 2000;60:4573–4581.
  • Weckbecker G, Briner U, Lewis I, et al. SOM230: a new somatostatin peptidomimetic with potent inhibitory effects on the growth hormone/insulin-like growth factor-I axis in rats, primates, and dogs. Endocrinology. 2002;143:4123–4130.
  • Chan JA, Blaszkowsky L, Stuart K, et al. A prospective, phase 1/2 study of everolimus and temozolomide in patients with advanced pancreatic neuroendocrine tumor. Cancer. 2013;119(17):3212–3218.
  • Chan JA, Mayer RJ, Jackson N, et al. Phase I study of sorafenib in combination with everolimus (RAD001) in patients with advanced neuroendocrine tumors. Cancer Chemother Pharmacol. 2013;71:1241–1246.
  • Claringbold PG, Turner JH. NeuroEndocrine tumor therapy with Lutetium-177-octreotate and Everolimus (NETTLE): a phase I study. Cancer Biother Radiopharm. 2015;30:261–269.
  • Salazar R, Garcia-Carbonero R, Libutti SK, et al. Phase II study of BEZ235 versus Everolimus in patients with mammalian target of Rapamycin inhibitor-naive advanced pancreatic neuroendocrine tumors. Oncologist. 2017. In press.
  • Oh DY, Kim TW, Park YS, et al. Phase 2 study of everolimus monotherapy in patients with nonfunctioning neuroendocrine tumors or pheochromocytomas/paragangliomas. Cancer. 2012;118:6162–6170.
  • Hobday TJ, Qin R, Reidy-Lagunes D, et al. Multicenter phase II Trial of Temsirolimus and Bevacizumab in pancreatic neuroendocrine tumors. J Clin Oncol. 2015;33:1551–1556.
  • 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.
  • Chan JA, Stuart K, Earle CC, et al. Prospective study of bevacizumab plus temozolomide in patients with advanced neuroendocrine tumors. J Clin Oncol. 2012;30:2963–2968.
  • Panzuto F, Rinzivillo M, Fazio N, et al. Real-world study of everolimus in advanced progressive neuroendocrine tumors. Oncologist. 2014;19:966–974.
  • Berardi R, Torniai M, Pusceddu S, et al. Prognostic impact of the cumulative dose and dose intensity of everolimus in patients with pancreatic neuroendocrine tumors. Cancer Med. 2017;6:1493–1499.
  • Liu CT, Chen MH, Chen JS, et al. The efficacy and safety of everolimus for the treatment of progressive gastroenteropancreatic neuroendocrine tumors: a multi-institution observational study in Taiwan. Asia Pac J Clin Oncol. 2016;12:396–402.
  • Lee L, Ito T, Igarashi H, et al. Impact of everolimus on Japanese patients with advanced pancreatic neuroendocrine neoplasms. J Hepatobiliary Pancreat Sci. 2017;24:95–102.
  • Lee KJ, Cho JH, Lee SH, et al. Clinical outcomes of everolimus in patients with advanced, nonfunctioning pancreatic neuroendocrine tumors: a multicenter study in Korea. Cancer Chemother Pharmacol. 2017;80:799–805.
  • Yao JC, Phan AT, Hess K, et al. Perfusion computed tomography as functional biomarker in randomized run-in study of bevacizumab and everolimus in well-differentiated neuroendocrine tumors. Pancreas. 2015;44:190–197.
  • Kamp K, Gumz B, Feelders RA, et al. Safety and efficacy of everolimus in gastrointestinal and pancreatic neuroendocrine tumors after (177)Lu-octreotate. Endocr Relat Cancer. 2013;20:825–831.
  • Angelousi A, Kamp K, Kaltsatou M, et al. Sequential Everolimus and Sunitinib treatment in pancreatic metastatic well-differentiated neuroendocrine tumours resistant to prior treatments. Neuroendocrinology. 2017;105:394–402.
  • Yoo C, Cho H, Song MJ, et al. Efficacy and safety of everolimus and sunitinib in patients with gastroenteropancreatic neuroendocrine tumor. Cancer Chemother Pharmacol. 2017;79:139–146.
  • Capdevila J, Sevilla I, Alonso V, et al. Evaluation of the efficacy and safety of lanreotide in combination with targeted therapies in patients with neuroendocrine tumours in clinical practice: a retrospective cross-sectional analysis. BMC Cancer. 2015;15:495.
  • Wolin EM. Long-term everolimus treatment of patients with pancreatic neuroendocrine tumors. Chemotherapy. 2014;60:143–150.
  • Ter-Minassian M, Zhang S, Brooks NV, et al. Association between tumor progression endpoints and overall survival in patients with advanced neuroendocrine tumors. Oncologist. 2017;22:165–172.
  • Ito T, Lee L, Jensen RT. Treatment of symptomatic neuroendocrine tumor syndromes: recent advances and controversies. Expert Opin Pharmacother. 2016;17:2191–2205.
  • Ito T, Lee L, Jensen RT. Carcinoid-syndrome: recent advances, current status and controversies. Curr Opin Endocrinol Diabetes Obes. 2018;25:22–35.
  • 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;103:153–171.
  • Strosberg JR, Halfdanarson TR, Bellizzi AM, et al. The north american neuroendocrine tumor society consensus guidelines for surveillance and medical management of midgut neuroendocrine tumors. Pancreas. 2017;46:707–714.
  • Roy PK, Venzon DJ, Feigenbaum KM, et al. Gastric secretion in Zollinger-Ellison syndrome: correlation with clinical expression, tumor extent and role in diagnosis - a prospective NIH study of 235 patients and review of the literature in 984 cases. Medicine. 2001;80:189–222, (Baltimore)
  • Berna MJ, Hoffmann KM, Serrano J, et al. Serum gastrin in Zollinger-Ellison syndrome: I. prospective study of fasting serum gastrin in 309 patients from the National Institutes of Health and comparison with 2229 cases from the literature. Medicine. 2006;85:295–330, (Baltimore)
  • Berna MJ, Hoffmann KM, Long SH, et al. Serum gastrin in Zollinger-Ellison syndrome: II. Prospective study of gastrin provocative testing in 293 patients from the National Institutes of Health and comparison with 537 cases from the literature. evaluation of diagnostic criteria, proposal of new criteria, and correlations with clinical and tumoral features. Medicine. 2006;85:331–364, (Baltimore)
  • Ito T, Igarashi H, Jensen RT. Zollinger-Ellison syndrome: recent advances and controversies. Curr Opin Gastroenterol. 2013;29:650–661.
  • Metz DC, Jensen RT. Gastrointestinal neuroendocrine tumors: pancreatic endocrine tumors. Gastroenterology. 2008;135:1469–1492.
  • Collen MJ, Howard JM, McArthur KE, et al. Comparison of ranitidine and cimetidine in the treatment of gastric hypersecretion. Ann Intern Med. 1984;100:52–58.
  • Ito T, Igarashi H, Uehara H, et al. Pharmacotherapy of Zollinger-Ellison syndrome. Expert Pharmacother. 2013;14:307–321.
  • Ito T, Igarashi H, Jensen RT. Pancreatic neuroendocrine tumors: clinical features, diagnosis and medical treatment: advances. Best Pract Res Clin Gastroenterol. 2012;26:737–753.
  • Ito T, Igarashi H, Jensen RT. Therapy of metastatic pancreatic neuroendocrine tumors (pNETs): recent insights and advances. J Gastroenterol. 2012;47:941–960.
  • Stehouwer CD, Lems WF, Fischer HR, et al. Aggravation of hypoglycemia in insulinoma patients by the long-acting somatostatin analogue octreotide (Sandostatin). Acta Endocrinol (Copenh). 1989;121:34–40.
  • Pavel ME, Chen D, He W, et al. Everolimus effect on gastrin and glucagon in pancreatic neuroendocrine tumors. Pancreas. 2017;46:751–757.
  • Capdevila J, Diez Miranda I, Obiols G, et al. Control of carcinoid syndrome with everolimus. Ann Oncol. 2011;22:237–239.
  • Bainbridge HE, Larbi E, Middleton G. Symptomatic control of neuroendocrine tumours with everolimus. Horm Cancer. 2015;6:254–259.
  • Houde VP, Brule S, Festuccia WT, et al. Chronic rapamycin treatment causes glucose intolerance and hyperlipidemia by upregulating hepatic gluconeogenesis and impairing lipid deposition in adipose tissue. Diabetes. 2010;59:1338–1348.
  • Bernard V, Lombard-Bohas C, Taquet MC, et al. Efficacy of Everolimus in patients with metastatic insulinoma and refractory Hypoglycemia. Eur J Endocrinol. 2013;168:665–674.
  • Bozkirli E, Bakiner O, Abali H, et al. A case of inoperable malignant insulinoma with resistant hypoglycemia who experienced the most significant clinical improvement with everolimus. Case Rep Endocrinol. 2013;2013:636175.
  • Asayama M, Yamada-Murano T, Hara H, et al. Everolimus dramatically improves glycemic control in unresectable metastatic insulinoma: a case report. Jpn J Clin Oncol. 2014;44:186–190.
  • Kulke MH, Bergsland EK, Yao JC. Glycemic control in patients with insulinoma treated with everolimus. N Engl J Med. 2009;360:195–197.
  • Fiebrich HB, Siemerink EJ, Brouwers AH, et al. Everolimus induces rapid plasma glucose normalization in insulinoma patients by effects on tumor as well as normal tissues. Oncologist. 2011;16:783–787.
  • Thomas NJ, Brooke AM, Besser GM. Long-term maintenance of normoglycaemia using everolimus in a patient with disseminated insulinoma and severe hypoglycaemia. Clin Endocrinol (Oxf). 2013;78:799–800.
  • Scharf M, Mueller D, Koenig U, et al. Management of a metastasized high grade insulinoma (G3) with refractory hypoglycemia: case report and review of the literature. Pancreatology. 2014;14:542–545.
  • Garcia-Carbonero R, Sorbye H, Baudin E, et al. ENETS consensus guidelines for high-grade gastroenteropancreatic neuroendocrine tumors and neuroendocrine carcinomas. Neuroendocrinology. 2016;103:186–194.
  • Panzuto F, Rinzivillo M, Spada F, et al. Everolimus in pancreatic neuroendocrine carcinomas G3. Pancreas. 2017;46:302–305.
  • Genc CG, Klumpen HJ, Denecke T, et al. Successful treatment of high-grade pancreatic neuroendocrine neoplasms with everolimus. Acta Oncol. 2018;57(5):686–688.
  • Fonseca PJ, Uriol E, Galvan JA, et al. Prolonged clinical benefit of everolimus therapy in the management of high-grade pancreatic neuroendocrine carcinoma. Case Rep Oncol. 2013;6:441–449.
  • Tanaka H, Matsusaki S, Baba Y, et al. Neuroendocrine tumor G3: a pancreatic well-differentiated neuroendocrine tumor with a high proliferative rate. Clin J Gastroenterol. 2015;8:414–420.
  • Chung DC, Brown SB, Graeme-Cook F, et al. Localization of putative tumor suppressor loci by genome-wide allelotyping in human pancreatic endocrine tumors. Cancer Res. 1998;58:3706–3711.
  • Bollard J, Couderc C, Blanc M, et al. Antitumor effect of everolimus in preclinical models of high-grade gastroenteropancreatic neuroendocrine carcinomas. Neuroendocrinology. 2013;97:331–340.
  • Gilabert M, Rho YS, Kavan P. Targeted therapies provide treatment options for poorly differentiated pancreatic neuroendocrine carcinomas. Oncology. 2017;92:170–172.
  • Tarhini A, Kotsakis A, Gooding W, et al. Phase II study of everolimus (RAD001) in previously treated small cell lung cancer. Clin Cancer Res. 2010;16:5900–5907.
  • Sun JM, Kim JR, Do IG, et al. A phase-1b study of everolimus plus paclitaxel in patients with small-cell lung cancer. Br J Cancer. 2013;109:1482–1487.
  • Christopoulos P, Engel-Riedel W, Grohe C, et al. Everolimus with paclitaxel and carboplatin as first-line treatment for metastatic large-cell neuroendocrine lung carcinoma: a multicenter phase II trial. Ann Oncol. 2017;28:1898–1902.
  • Dasari A, Mehta K, Byers LA, et al. Comparative study of lung and extrapulmonary poorly differentiated neuroendocrine carcinomas: A SEER database analysis of 162,983 cases. Cancer. 2018;124:807–815.
  • Martins D, Spada F, Lambrescu I, et al. Predictive markers of response to everolimus and sunitinib in neuroendocrine tumors. Target Oncol. 2017;12:611–622.
  • Yao JC, Pavel M, Phan AT, et al. Chromogranin A and neuron-specific enolase as prognostic markers in patients with advanced pNET treated with everolimus. J Clin Endocrinol Metab. 2011;96:3741–3749.
  • Serra S, Zheng L, Hassan M, et al. The FGFR4-G388R single-nucleotide polymorphism alters pancreatic neuroendocrine tumor progression and response to mTOR inhibition therapy. Cancer Res. 2012;72:5683–5691.
  • Cros J, Moati E, Raffenne J, et al. Gly388Arg FGFR4 polymorphism is not predictive of everolimus efficacy in well-differentiated digestive neuroendocrine tumors. Neuroendocrinology. 2016;103:495–499.
  • Meric-Bernstam F, Akcakanat A, Chen H, et al. PIK3CA/PTEN mutations and Akt activation as markers of sensitivity to allosteric mTOR inhibitors. Clin Cancer Res. 2012;18:1777–1789.
  • Gagliano T, Bellio M, Gentilin E, et al. mTOR, p70S6K, AKT, and ERK1/2 levels predict sensitivity to mTOR and PI3K/mTOR inhibitors in human bronchial carcinoids. Endocr Relat Cancer. 2013;20:463–475.
  • Benslama N, Bollard J, Vercherat C, et al. Prediction of response to everolimus in neuroendocrine tumors: evaluation of clinical, biological and histological factors. Invest New Drugs. 2016;34:654–662.
  • Boulay A, Zumstein-Mecker S, Stephan C, et al. Antitumor efficacy of intermittent treatment schedules with the rapamycin derivative RAD001 correlates with prolonged inactivation of ribosomal protein S6 kinase 1 in peripheral blood mononuclear cells. Cancer Res. 2004;64:252–261.
  • Tanaka C, O’Reilly T, Kovarik JM, et al. Identifying optimal biologic doses of everolimus (RAD001) in patients with cancer based on the modeling of preclinical and clinical pharmacokinetic and pharmacodynamic data. J Clin Oncol. 2008;26:1596–1602.
  • van Asselt SJ, Oosting SF, Brouwers AH, et al. Everolimus reduces (89)Zr-Bevacizumab tumor uptake in patients with neuroendocrine tumors. J Nucl Med. 2014;55:1087–1092.
  • Rugo HS, Hortobagyi GN, Yao J, et al. Meta-analysis of stomatitis in clinical studies of everolimus: incidence and relationship with efficacy. Ann Oncol. 2016;27:519–525.
  • Abdel-Rahman O, Fouad M. Risk of mucocutaneous toxicities in patients with solid tumors treated with everolimus; a systematic review and meta-analysis. Expert Rev Anticancer Ther. 2014;14:1529–1536.
  • Ravaud A, Urva SR, Grosch K, et al. Relationship between everolimus exposure and safety and efficacy: meta-analysis of clinical trials in oncology. Eur J Cancer. 2014;50:486–495.
  • Shameem R, Lacouture M, Wu S. Incidence and risk of rash to mTOR inhibitors in cancer patients–a meta-analysis of randomized controlled trials. Acta Oncol. 2015;54:124–132.
  • Ramirez-Fort MK, Case EC, Rosen AC, et al. Rash to the mTOR inhibitor everolimus: systematic review and meta-analysis. Am J Clin Oncol. 2014;37:266–271.
  • Abdel-Rahman O, Fouad M. Risk of fatigue and hepatic and metabolic toxicities in patients with solid tumors treated with everolimus: a meta-analysis. Future Oncol. 2015;11:79–90.
  • Garcia CA, Wu S. Attributable risk of infection to mTOR inhibitors Everolimus and Temsirolimus in the treatment of cancer. Cancer Invest. 2016;34:521–530.
  • Iacovelli R, Palazzo A, Mezi S, et al. Incidence and risk of pulmonary toxicity in patients treated with mTOR inhibitors for malignancy. A meta-analysis of published trials. Acta Oncol. 2012;51:873–879.
  • Noguchi S, Shinohara N, Ito T, et al. Relationship between pulmonary adverse events and Everolimus exposure in Japanese and non-japanese patients: a meta-analysis of oncology trials. Oncology. 2017;92:243–254.
  • Xu KY, Shameem R, Wu S. Risk of hyperglycemia attributable to everolimus in cancer patients: A meta-analysis. Acta Oncol. 2016;55:1196–1203.
  • Xu J, Tian D. Hematologic toxicities associated with mTOR inhibitors temsirolimus and everolimus in cancer patients: a systematic review and meta-analysis. Curr Med Res Opin. 2014;30:67–74.
  • Shameem R, Hamid MS, Wu S. Risk of anemia attributable to everolimus in patients with cancer: a meta-analysis of randomized controlled trials. Anticancer Res. 2015;35:2333–2340.
  • Qi WX, Huang YJ, Yao Y, et al. Incidence and risk of treatment-related mortality with mTOR inhibitors everolimus and temsirolimus in cancer patients: a meta-analysis. PLoS One. 2013;8:e65166.
  • Rogers SC, Garcia CA, Wu S. Discontinuation of Everolimus due to related and unrelated adverse events in cancer patients: a meta-analysis. Cancer Invest. 2017;35:552–561.
  • Choueiri TK, Je Y, Sonpavde G, et al. Incidence and risk of treatment-related mortality in cancer patients treated with the mammalian target of rapamycin inhibitors. Ann Oncol. 2013;24:2092–2097.
  • Plante A, Baudin E, Do Cao C, et al. Patient-reported tolerance in treatments approved in neuroendocrine tumors: a national survey from the French Group of Endocrine Tumors. Clin Res Hepatol Gastroenterol. 2018;42(2):153–159.
  • Pavel M, Unger N, Borbath I, et al. Safety and QOL in patients with advanced NET in a Phase 3b expanded access study of Everolimus. Target Oncol. 2016;11:667–675.
  • Deppenweiler M, Falkowski S, Saint-Marcoux F, et al. Towards therapeutic drug monitoring of everolimus in cancer? Results of an exploratory study of exposure-effect relationship. Pharmacol Res. 2017;121:138–144.
  • Aapro M, Andre F, Blackwell K, et al. Adverse event management in patients with advanced cancer receiving oral everolimus: focus on breast cancer. Ann Oncol. 2014;25:763–773.
  • Hsieh AC, Costa M, Zollo O, et al. Genetic dissection of the oncogenic mTOR pathway reveals druggable addiction to translational control via 4EBP-eIF4E. Cancer Cell. 2010;17:249–261.
  • Wullschleger S, Loewith R, Hall MN. TOR signaling in growth and metabolism. Cell. 2006;124:471–484.
  • Houghton PJ. Everolimus. Clin Cancer Res. 2010;16:1368–1372.
  • Tamburini J, Chapuis N, Bardet V, et al. Mammalian target of rapamycin (mTOR) inhibition activates phosphatidylinositol 3-kinase/Akt by up-regulating insulin-like growth factor-1 receptor signaling in acute myeloid leukemia: rationale for therapeutic inhibition of both pathways. Blood. 2008;111:379–382.
  • Kuo SH, Hsu CH, Chen LT, et al. Lack of compensatory pAKT activation and eIF4E phosphorylation of lymphoma cells towards mTOR inhibitor, RAD001. Eur J Cancer. 2011;47:1244–1257.
  • Svejda B, Kidd M, Kazberouk A, et al. Limitations in small intestinal neuroendocrine tumor therapy by mTor kinase inhibition reflect growth factor-mediated PI3K feedback loop activation via ERK1/2 and AKT. Cancer. 2011;117:4141–4154.
  • Wang X, Yue P, Kim YA, et al. Enhancing mammalian target of rapamycin (mTOR)-targeted cancer therapy by preventing mTOR/raptor inhibition-initiated, mTOR/rictor-independent Akt activation. Cancer Res. 2008;68:7409–7418.
  • Choo AY, Yoon SO, Kim SG, et al. Rapamycin differentially inhibits S6Ks and 4E-BP1 to mediate cell-type-specific repression of mRNA translation. Proc Natl Acad Sci U S A. 2008;105:17414–17419.
  • Kang SA, Pacold ME, Cervantes CL, et al. mTORC1 phosphorylation sites encode their sensitivity to starvation and rapamycin. Science. 2013;341:1236566.
  • Gupta M, Ansell SM, Novak AJ, et al. Inhibition of histone deacetylase overcomes rapamycin-mediated resistance in diffuse large B-cell lymphoma by inhibiting Akt signaling through mTORC2. Blood. 2009;114:2926–2935.
  • Sarbassov DD, Guertin DA, Ali SM, et al. Phosphorylation and regulation of Akt/PKB by the rictor-mTOR complex. Science. 2005;307:1098–1101.
  • Stan R, McLaughlin MM, Cafferkey R, et al. Interaction between FKBP12-rapamycin and TOR involves a conserved serine residue. J Biol Chem. 1994;269:32027–32030.
  • Carracedo A, Ma L, Teruya-Feldstein J, et al. Inhibition of mTORC1 leads to MAPK pathway activation through a PI3K-dependent feedback loop in human cancer. J Clin Invest. 2008;118:3065–3074.
  • Di Nicolantonio F, Arena S, Tabernero J, et al. Deregulation of the PI3K and KRAS signaling pathways in human cancer cells determines their response to everolimus. J Clin Invest. 2010;120:2858–2866.
  • Vandamme T, Beyens M, de Beeck KO, et al. Long-term acquired everolimus resistance in pancreatic neuroendocrine tumours can be overcome with novel PI3K-AKT-mTOR inhibitors. Br J Cancer. 2016;114:650–658.
  • Antonuzzo L, Del RM, Barucca V, et al. Critical focus on mechanisms of resistance and toxicity of m-TOR inhibitors in pancreatic neuroendocrine tumors. Cancer Treat Rev. 2017;57:28–35.
  • Freitag H, Christen F, Lewens F, et al. Inhibition of mTOR’s catalytic site by PKI-587 is a promising therapeutic option for gastroenteropancreatic neuroendocrine tumor disease. Neuroendocrinology. 2017;105:90–104.
  • Passacantilli I, Capurso G, Archibugi L, et al. Combined therapy with RAD001 e BEZ235 overcomes resistance of PET immortalized cell lines to mTOR inhibition. Oncotarget. 2014;5:5381–5391.
  • Zitzmann K, Ruden J, Brand S, et al. Compensatory activation of Akt in response to mTOR and Raf inhibitors - a rationale for dual-targeted therapy approaches in neuroendocrine tumor disease. Cancer Lett. 2010;295:100–109.
  • Bendell JC, Kurkjian C, Infante JR, et al. A phase 1 study of the sachet formulation of the oral dual PI3K/mTOR inhibitor BEZ235 given twice daily (BID) in patients with advanced solid tumors. Invest New Drugs. 2015;33:463–471.
  • Fazio N, Buzzoni R, Baudin E, et al. A phase II study of BEZ235 in patients with everolimus-resistant, advanced pancreatic neuroendocrine tumours. Anticancer Res. 2016;36:713–719.
  • Rinke A, Muller 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.
  • 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.
  • Dilz LM, Denecke T, Steffen IG, et al. Streptozocin/5-fluorouracil chemotherapy is associated with durable response in patients with advanced pancreatic neuroendocrine tumours. Eur J Cancer. 2015;51:1253–1262.
  • Clewemar Antonodimitrakis P, Sundin A, Wassberg C, et al. Streptozocin and 5-Fluorouracil for the treatment of pancreatic neuroendocrine tumors: efficacy, prognostic factors and toxicity. Neuroendocrinology. 2016;103:345–353.
  • 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;23:759–767.
  • von Schrenck T, Howard JM, Doppman JL, et al. Prospective study of chemotherapy in patients with metastatic gastrinoma. Gastroenterology. 1988;94:1326–1334.
  • Crona J, Fanola I, Lindholm DP, et al. Effect of temozolomide in patients with metastatic bronchial carcinoids. Neuroendocrinology. 2013;98:151–155.
  • Crona J, Bjorklund P, Welin S, et al. Treatment, prognostic markers and survival in thymic neuroendocrine tumours. Lung Cancer. 2013;79:289–293.
  • 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;24:401–406.
  • Krug S, Gress TM, Michl P, et al. The role of cytotoxic chemotherapy in advanced pancreatic neuroendocrine tumors. Digestion. 2017;96:67–75.
  • Partelli S, Bartsch DK, Capdevila J, et al. ENETS consensus guidelines for standard of care in neuroendocrine tumours: surgery for small intestinal and pancreatic neuroendocrine tumours. Neuroendocrinology. 2017;105:255–265.
  • 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. Pancreas. 2017;46:715–731.
  • Singh S, Dey C, Kennecke H, et al. Consensus recommendations for the diagnosis and management of pancreatic neuroendocrine tumors: guidelines from a Canadian national expert group. Ann Surg Oncol. 2015;22:2685–2699.
  • Oberg K, Knigge U, Kwekkeboom D, et al. Neuroendocrine gastro-entero-pancreatic tumors: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2012;23(Suppl 7):vii124–vii130.
  • Caplin ME, Baudin E, Ferolla P, et al. Pulmonary neuroendocrine (carcinoid) tumors: European Neuroendocrine Tumor Society expert consensus and recommendations for best practice for typical and atypical pulmonary carcinoids. Ann Oncol. 2015;26:1604–1620.
  • 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;11:815–824.
  • Marinova M, Mucke M, Mahlberg L, et al. Improving quality of life in patients with pancreatic neuroendocrine tumor following peptide receptor radionuclide therapy assessed by EORTC QLQ-C30. Eur J Nucl Med Mol Imaging. 2018;45:38–46.
  • 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;103:172–185.
  • Kulke MH, Shah MH, Benson AB, et al. Neuroendocrine tumors: version 3.2017: NCCN clinical practice guidelines in oncology. NCCN Clin Pract Guidel Oncol. 2017;1–116.
  • Janson ET, Sorbye H, Welin S, et al. Nordic guidelines 2014 for diagnosis and treatment of gastroenteropancreatic neuroendocrine neoplasms. Acta Oncol. 2014;53:1284–1297.
  • Kulke MH, Anthony LB, Bushnell DL, et al. NANETS treatment guidelines: well-differentiated neuroendocrine tumors of the stomach and pancreas. Pancreas. 2010;39:735–752.
  • Strosberg JR, Coppola D, Klimstra DS, et al. The NANETS consensus guidelines for the diagnosis and management of poorly differentiated (high-grade) extrapulmonary neuroendocrine carcinomas. Pancreas. 2010;39:799–800.
  • Hicks RJ, Kwekkeboom DJ, Krenning E, et al. ENETS consensus guidelines for the standards of care in neuroendocrine neoplasia: peptide receptor radionuclide therapy with radiolabeled somatostatin analogues. Neuroendocrinology. 2017;105:295–309.
  • 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;42:5–19.
  • Shipkova M, Hesselink DA, Holt DW, et al. Therapeutic drug monitoring of everolimus: a consensus report. Ther Drug Monit. 2016;38:143–169.

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.