Guidelines for the management of neuroendocrine tumours by the Brazilian gastrointestinal tumour group

Rachel P Riechelmann, Rui F Weschenfelder, Frederico P Costa, Aline Chaves Andrade, Alessandro Bersch Osvaldt, Ana Rosa P Quidute, Allan Dos Santos, Ana Amélia O Hoff, Brenda Gumz, Carlos Buchpiguel, Bruno S Vilhena Pereira, Delmar Muniz Lourenço Junior, Duilio Reis da Rocha Filho, Eduardo Antunes Fonseca, Eduardo Linhares Riello Mello, Fabio Ferrari Makdissi, Fabio Luiz Waechter, Francisco Cesar Carnevale, George B Coura-Filho, Gustavo Andrade de Paulo, Gustavo Colagiovanni Girotto, João Evangelista Bezerra Neto, João Glasberg, Jose Claudio Casali-da-Rocha, Juliana Florinda M Rego, Luciana Rodrigues de Meirelles, Ludhmila Hajjar, Marcos Menezes, Marcello D Bronstein, Marcelo Tatit Sapienza, Maria Candida Barisson Villares Fragoso, Maria Adelaide Albergaria Pereira, Milton Barros, Nora Manoukian Forones, Paulo Cezar Galvão do Amaral, Raphael Salles Scortegagna de Medeiros, Raphael L C Araujo, Regis Otaviano França Bezerra, Renata D'Alpino Peixoto, Samuel Aguiar Jr, Ulysses Ribeiro Jr, Tulio Pfiffer, Paulo M Hoff, Anelisa K Coutinho, Rachel P Riechelmann, Rui F Weschenfelder, Frederico P Costa, Aline Chaves Andrade, Alessandro Bersch Osvaldt, Ana Rosa P Quidute, Allan Dos Santos, Ana Amélia O Hoff, Brenda Gumz, Carlos Buchpiguel, Bruno S Vilhena Pereira, Delmar Muniz Lourenço Junior, Duilio Reis da Rocha Filho, Eduardo Antunes Fonseca, Eduardo Linhares Riello Mello, Fabio Ferrari Makdissi, Fabio Luiz Waechter, Francisco Cesar Carnevale, George B Coura-Filho, Gustavo Andrade de Paulo, Gustavo Colagiovanni Girotto, João Evangelista Bezerra Neto, João Glasberg, Jose Claudio Casali-da-Rocha, Juliana Florinda M Rego, Luciana Rodrigues de Meirelles, Ludhmila Hajjar, Marcos Menezes, Marcello D Bronstein, Marcelo Tatit Sapienza, Maria Candida Barisson Villares Fragoso, Maria Adelaide Albergaria Pereira, Milton Barros, Nora Manoukian Forones, Paulo Cezar Galvão do Amaral, Raphael Salles Scortegagna de Medeiros, Raphael L C Araujo, Regis Otaviano França Bezerra, Renata D'Alpino Peixoto, Samuel Aguiar Jr, Ulysses Ribeiro Jr, Tulio Pfiffer, Paulo M Hoff, Anelisa K Coutinho

Abstract

Neuroendocrine tumours are a heterogeneous group of diseases with a significant variety of diagnostic tests and treatment modalities. Guidelines were developed by North American and European groups to recommend their best management. However, local particularities and relativisms found worldwide led us to create Brazilian guidelines. Our consensus considered the best feasible strategies in an environment involving more limited resources. We believe that our recommendations may be extended to other countries with similar economic standards.

Keywords: cancer; chemotherapy; guideline; neuroendcrine tumours; radionuclide peptide therapy; targeted therapy.

References

    1. Bosman FT. WHO Classification of Tumours of the Digestive System. Lyon, France: IARC Press; 2010.
    1. Klimstra DS. Pathology reporting of neuroendocrine tumours: essential elements for accurate diagnosis, classification, and staging. Semin Oncol. 2013;40:23–36. doi: 10.1053/j.seminoncol.2012.11.001.
    1. Klimstra DS, et al. Pathology reporting of neuroendocrine tumours: application of the Delphic consensus process to the development of a minimum pathology data set. Am J Surg Pathol. 2010;34:300–313. doi: 10.1097/PAS.0b013e3181ce1447.
    1. Luo G, et al. Modified staging classification for pancreatic neuroendocrine tumours on the basis of the American Joint Committee on Cancer and European Neuroendocrine Tumour Society systems. J Clin Oncol. 2016;34 [ahead of print]
    1. Klimstra DS, et al. The pathologic classification of neuroendocrine tumours: a review of nomenclature, grading, and staging systems. Pancreas. 2010;39:707–712. doi: 10.1097/MPA.0b013e3181ec124e.
    1. Lawrence B, et al. The epidemiology of GEP neuroendocrine tumours. Endocrinol Metab Clin North Am. 2011;40:1–18. doi: 10.1016/j.ecl.2010.12.005.
    1. Massironi S, et al. Chromogranin A in diagnosing and monitoring patients with GEP neuroendocrine neoplasms: a large series from a single institution. Neuroendocrinology. 2014;100:240–249. doi: 10.1159/000369818.
    1. Rossi RE, et al. Chromogranin A as a predictor of radiological disease progression in neuroendocrine tumours. Ann Transl Med. 2015;9:118.
    1. Turner GB, et al. Circulating markers of prognosis and response to treatment in patients with midgut carcinoid tumours. Gut. 2006;55:1586–1591. doi: 10.1136/gut.2006.092320.
    1. Okabayashi T, et al. Diagnosis and management of insulinoma. World J Gastroenterol. 2013;19:829. doi: 10.3748/wjg.v19.i6.829.
    1. Cryer PE, et al. Evaluation and management of adult hypoglycemic disorders: an Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab. 2009;3:709–728. doi: 10.1210/jc.2008-1410.
    1. Guettier J-M, et al. The role of proinsulin and insulin in the diagnosis of insulinoma: a critical evaluation of the endocrine society clinical practice guideline. J Clin Endocrinol Metab. 2013;12:4752–4758. doi: 10.1210/jc.2013-2182.
    1. Hirshberg B, et al. Forty-eight-hour fast: the diagnostic test for insulinoma. J Clin Endocrinol Metab. 2000;9:3222–3226. doi: 10.1210/jcem.85.9.6807.
    1. Kulke MH, et al. NANETS treatment guidelines: well-differentiated neuroendocrine tumours of the stomach and pancreas. Pancreas. 2010;39:735–752. doi: 10.1097/MPA.0b013e3181ebb168.
    1. Berna MJ, 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 tumoural features. Medicine (Baltimore) 2006;85:331–364. doi: 10.1097/MD.0b013e31802b518c.
    1. Sundin A, et al. ENETS consensus guidelines for the standards of care in neuroendocrine tumours: radiological examinations. Neuroendocrinology. 2009;90:167–183. doi: 10.1159/000184855.
    1. Anderson MA, et al. Endoscopic ultrasound is highly accurate and directs management in patients with neuroendocrine tumours of the pancreas. Am J Gastroenterol. 2000;95:2271–2277. doi: 10.1111/j.1572-0241.2000.02480.x.
    1. Gouya H, et al. CT, endoscopic sonography, and a combined protocol for preoperative evaluation of pancreatic insulinomas. AJR Am J Roentgenol. 2003;181:987–989. doi: 10.2214/ajr.181.4.1810987.
    1. Khashab MA, et al. EUS is still superior to multidetector computerized tomography for detection of pancreatic neuroendocrine tumours. Gastrointest Endosc. 2011;73:691–696. doi: 10.1016/j.gie.2010.08.030.
    1. Horton KM, et al. Multi-detector row CT of pancreatic islet cell tumours. Radiographics. 2006;2:453–464. doi: 10.1148/rg.262055056.
    1. Versari A, et al. Ga-68 DOTATOC PET, endoscopic ultrasonography, and multidetector CT in the diagnosis of duodenopancreatic neuroendocrine tumours: a single-centre retrospective study. Clin Nucl Med. 2010;35:321–328. doi: 10.1097/RLU.0b013e3181d6677c.
    1. Caramella C, et al. Endocrine pancreatic tumours: which are the most useful MRI sequences? Eur Radiol. 2010;11:2618–2627. doi: 10.1007/s00330-010-1840-5.
    1. Baumann T, et al. Gastroenteropancreatic neuroendocrine tumours (GEP-NET) - Imaging and staging. Best Pract Res Clin Endocrinol Metab. 2016;30:45–57. doi: 10.1016/j.beem.2016.01.003.
    1. Fidler JL, et al. Small bowel imaging: computed tomography enterography, magnetic resonance enterography, angiography, and nuclear medicine. Gastrointest Endosc Clin N Am. 2017;27:133–152. doi: 10.1016/j.giec.2016.08.008.
    1. Puli SR, et al. Diagnostic accuracy of endoscopic ultrasound in pancreatic neuroendocrine tumours: a systematic review and meta-analysis. World J Gastroenterol. 2013;23:3678–3684. doi: 10.3748/wjg.v19.i23.3678.
    1. Sugimoto M, et al. Conventional versus contrast-enhanced harmonic endoscopic ultrasonography-guided fine-needle aspiration for diagnosis of solid pancreatic lesions: a prospective randomized trial. Pancreatology. 2015;15:538–541. doi: 10.1016/j.pan.2015.06.005.
    1. Reidy-Lagunes D, et al. Assition of octreotide functional imaging to cress-sectional computed tomography or magnetic resonance imaging for the detection of neuroendocrine tumours: added value or an anachronism? J Clin Oncol. 2011;20:e74–e75. doi: 10.1200/JCO.2010.32.8559.
    1. Geijer H, Breimer LH. Somatostatin receptor PET/CT in neuroendocrine tumours: update on systematic review and meta-analysis. Eur J Nucl Med Mol Imaging. 2013;11:1770–1780. doi: 10.1007/s00259-013-2482-z.
    1. Etchebehere EC, et al. 68Ga-DOTATATE PET/CT, 99mTc-HYNIC-octreotide SPECT/CT, and whole-body MR imaging in detection of neuroendocrine tumours: a prospective trial. J Nucl Med. 2014;10:1598–1604. doi: 10.2967/jnumed.114.144543.
    1. Panagiotidis E, et al. Comparison of the impact of 68Ga – DOTATATE and 18F – DG PET/CT on clinical management in patients with neuroendocrine tumours. J Nucl Med. 2016.
    1. Panagiotidis E, Bomanji J. Role of 18F-fluorodeoxyglucose PET in the study of neuroendocrine tumours. PET Clin. 2016;9:43–55. doi: 10.1016/j.cpet.2013.08.008.
    1. Naswa N, et al. Ga-DOTANOC PET/CT in patients with carcinoma of unknown primary of neuroendocrine origin. Clin Nucl Med. 2012;37:245–251. doi: 10.1097/RLU.0b013e31823ea730.
    1. Nakamoto Y, et al. Additional information gained by positron emission tomography with (68)Ga-DOTATOC for suspected unknown primary or recurrent neuroendocrine tumours. Ann Nucl Med. 2015;29:512–518. doi: 10.1007/s12149-015-0973-7.
    1. Moryoussef F, et al. Impact on management of liver and whole-body diffusion weighted magnetic resonance imaging sequences for neuroendocrine tumours: a pilot study. Neuroendocrinology. 2016;28 [Epub ahead of print]
    1. Boudreaux JP, et al. Surgical treatment of advanced-stage carcinoid tumours: lessons learned. Ann Surg. 2005;241:839–845. doi: 10.1097/01.sla.0000164073.08093.5d.
    1. Hellman P, et al. Effect of surgery on the outcome of midgut carcinoid disease with lymph node and liver metastases. World J Surg. 2002;8:991–997. doi: 10.1007/s00268-002-6630-z.
    1. Givi B, et al. Operative resection of primary carcinoid neoplasms in patients with liver metastases yields significantly better survival. Surgery. 2006;140:891–897. doi: 10.1016/j.surg.2006.07.033.
    1. Bartlett EK, et al. Surgery for metastatic neuroendocrine tumours with occult primaries. J Surg Res. 2013;184:221–227. doi: 10.1016/j.jss.2013.04.008.
    1. Bellutti M. Detection of neuroendocrine tumours of the small bowel by double balloon enteroscopy. Dig Dis Sci. 2009;54:1050–1058. doi: 10.1007/s10620-008-0456-y.
    1. Naswa N, et al. 68Ga-DOTANOC PET/CT in patients with carcinoma of unknown primary of neuroendocrine origin. Clin Nucl Med. 2012;37:245–251. doi: 10.1097/RLU.0b013e31823ea730.
    1. Prasad V, et al. Detection of unknown primary neuroendocrine tumours (CUP-NET) using 68 Ga-DOTA-NOC receptor PET/CT. Eur J Nucl Med Mol Imaging. 2010;37:67–77. doi: 10.1007/s00259-009-1205-y.
    1. Kunikowska J, et al. A frequency and semiquantitative analysis of pathological 68Ga DOTATATE PET/CT uptake by primary site-dependent neuroendocrine tumour metastasis. Clin Nucl Med. 2014;39:855–861. doi: 10.1097/RLU.0000000000000533.
    1. Tan, et al. Diagnostic value of (68)Ga-DOTATATE PET/CT in liver metastases of neuroendocrine tumours of unknown origin. Nucl Med Mol Imaging. 2014;48:212–215. doi: 10.1007/s13139-013-0258-9.
    1. Savelli G, et al. Feasibility of somatostatin receptor scintigraphy in the detection of occult primary gastro-entero-pancreatic (GEP) neuroendocrine tumours. Nucl Med Commun. 2004;25:445–449. doi: 10.1097/00006231-200405000-00004.
    1. Chang S, et al. Neuroendocrine neoplasms of the gastrointestinal tract: classification, pathologic basis and imaging features. Radiographics. 2007;27:1667–1679. doi: 10.1148/rg.276075001.
    1. Van Tuyl, et al. Detection of small-bowel neuroendocrine tumours by video capsule endoscopy. Gastrointest Endosc. 2006;64:66–72. doi: 10.1016/j.gie.2006.01.054.
    1. Frilling A, et al. Capsule endoscopy to detect primary tumour site in metastatic neuroendocrine tumours. Dig Liver Dis. 2014;46:1038–1042. doi: 10.1016/j.dld.2014.07.004.
    1. Karagiannis S, et al. Capsule retention: a feared complication of wireless capsule endoscopy. Scand J Gastroenterol. 2009;44:1158–1165. doi: 10.1080/00365520903132039.
    1. Oberg K, Castellano D. Current knowledge on diagnosis and staging of neuroendrocrine tumours. Cancer Metastasis Rev. 2011;30(Suppl 1):3–7. doi: 10.1007/s10555-011-9292-1.
    1. Syngal S, et al. Surveillance of patients at high risk for colorectal cancer. Med Clin North Am. 2005;89:61–84. doi: 10.1016/j.mcna.2004.08.013.
    1. Herrera MF, et al. AACE/ACE disease state clinical review: pancreatic neuroendocrine incidentalomas. Endocr Pract. 2015;21:546–553. doi: 10.4158/EP14465.DSC.
    1. Jung JG, et al. Behaviour of small, asymptomatic, nonfunctioning pancreatic neuroendocrine tumours (NF-PNETs) Medicine (Baltimore) 2015;94:e983. doi: 10.1097/MD.0000000000000983.
    1. Partelli S, et al. Systematic review of active surveillance versus surgical management of asymptomatic small non-functioning pancreatic neuroendocrine neoplasm. Br J Surg. 2016.
    1. Capurso G, et al. Systematic review of resection of primary midgut carcinoid tumour in patients with unresectable liver metastases. Br J Surg. 2012;99:1480–1486. doi: 10.1002/bjs.8842.
    1. Keutgen XM, et al. Non-functioning pancreatic endocrine tumour. Surgery. 2016;159:311–318. doi: 10.1016/j.surg.2015.05.042.
    1. Bettini R, et al. Primary tumour resection in metastatic nonfunctioning pancreatic endocrine carcinomas. Dig Liver Dis. 2009;41:49–55. doi: 10.1016/j.dld.2008.03.015.
    1. Charnsangavej C, et al. Selection of patients for resection of hepatic colorectal metastases: expert consensus statement. Ann Surg Oncol. 2006;10:1261–1268. doi: 10.1245/s10434-006-9023-y.
    1. Abdalla EK, et al. Locoregional surgical and interventional therapies for advanced colorectal cancer liver metastases: expert consensus statements. HPB (Oxford) 2013;15:119–130. doi: 10.1111/j.1477-2574.2012.00597.x.
    1. Kianmanesh R, et al. Surgical treatment of gastric, enteric, and pancreatic endocrine tumours Part 1. Treatment of primary endocrine tumours. J Chir. 2015;142:132–149. doi: 10.1016/S0021-7697(05)80881-6.
    1. Sarmiento JM, et al. Surgical treatment of neuroendocrine metastases to the liver: a plea for resection to increase survival. J Am Coll Surg. 2003;197:29–37. doi: 10.1016/S1072-7515(03)00230-8.
    1. Chen J, et al. Simultaneous vs. staged resection for synchronous colorectal liver metastases: a metaanalysis. Int J Colorectal Dis. 2011;2:191–199. doi: 10.1007/s00384-010-1018-2.
    1. Graff-Baker AN, et al. Expanded criteria for carcinoid liver debulking: maintaining survival and increasing the number of eligible patients. Surgery. 2014;156:1369–1376. doi: 10.1016/j.surg.2014.08.009.
    1. Hodul P, et al. The role of cytoreductive hepatic surgery as an adjunct to the management of metastatic neuroendocrine carcinomas. Cancer Control. 2006;1:61–71.
    1. D'Haese JG, et al. Update on surgical treatment of pancreatic neuroendocrine neoplasm. World J Gastroenterol. 2014;38:13893–13898. doi: 10.3748/wjg.v20.i38.13893.
    1. Kennedy A, et al. Role of hepatic intra-arterial therapies in metastatic neuroendocrine tumours (NET): guidelines from the NET-Liver-Metastases Consensus Conference. HPB (Oxford) 2015;17:29–37. doi: 10.1111/hpb.12326.
    1. Frilling A, Clift AK. Therapeutic strategies of neuroendocrine liver metastases. Cancer. 2015;121:1172–1186. doi: 10.1002/cncr.28760.
    1. Lewis M, Hobday T. Treatment of neuroendocrine tumour liver metastases. Int J Hepatol. 2012. pp. 1–12.
    1. Coldwell . Treatment of NET with SIRT. In: Bilbao JI, Reiser MF, et al., editors. Liver Radioembolization with Y90 Microspheres. Berlin: Springer; 2013. p. 153.
    1. Device Z, et al. The efficacy of hepatic 90Y resin radioembolization for metastatic neuroendocrine tumours: a meta-analysis. J Nucl Med. 2014;55:1404–1410. doi: 10.2967/jnumed.113.135855.
    1. Yang TX, et al. Radioembolization and chemoembolization for unresectable neuroendocrine liver metastases - a systematic review. Surg Oncol. 2012;21:299–308. doi: 10.1016/j.suronc.2012.07.001.
    1. Mazzaferro V, et al. Neuroendocrine tumours metastatic to the liver: how to select patients for liver transplantation? J Hepatol. 2007;47:460–466. doi: 10.1016/j.jhep.2007.07.004.
    1. Mazzaferro V, et al. Liver transplantation for the treatment of small hepatocellular carcinomas in patients with cirrhosis. N Engl J Med. 1996;334:693–699. doi: 10.1056/NEJM199603143341104.
    1. Rossi RE, et al. Liver transplantation for unresectable neuroendocrine tumour liver metastases. Ann Surg Oncol. 2014;7:2398–2405. doi: 10.1245/s10434-014-3523-y.
    1. Pape UF, et al. 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. doi: 10.1159/000335629.
    1. Caplin ME, et al. Lanreotide in metastatic enteropancreatic neuroendocrine tumours. N Engl J Med. 2014;371:224–233. doi: 10.1056/NEJMoa1316158.
    1. Rinke A, et al. Placebo-controlled, double-blind, prospective, randomized study on the effect of octreotide LAR in the control of tumour growth in patients with metastatic neuroendocrine midgut tumours: a report from the PROMID Study Group. J Clin Oncol. 2009;27:4656–4663. doi: 10.1200/JCO.2009.22.8510.
    1. Janson ET, et al. [111in-DTPA-D-Phe1] octreotide scintigraphy in patients with carcinoid tumours: the predictive value for somatostatin analogue treatment. Eur J Endocrinol. 1994;131:577–581. doi: 10.1530/eje.0.1310577.
    1. Riechelmann R, et al. Refractory carcinoid syndrome: a review of treatment options. Ther Adv Med Oncol. 2016. pp. 1–11.
    1. Strosberg J, et al. The Role of chemotherapy in well-differentiated GEP neuroendocrine tumours. Front Horm Res. 2015;44:239–247. doi: 10.1159/000403785.
    1. Ferrarotto R, et al. Combination of capecitabine and oxaliplatin is an effective treatment option for advanced neuroendocrine tumours. Rare Tumours. 2013;24:e35. doi: 10.4081/rt.2013.e35.
    1. Dussol AS, et al. Gemcitabine and oxaliplatin or alkylating agents for neuroendocrine tumours: comparison of efficacy and search for predictive factors guiding treatment choice. Cancer. 2015;121:3428–3434. doi: 10.1002/cncr.29517.
    1. 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 3 study. Lancet. 2011;378:2005–2012. doi: 10.1016/S0140-6736(11)61742-X.
    1. 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 3 study. Lancet. 2016;387:968–977. doi: 10.1016/S0140-6736(15)00817-X.
    1. Kulke MH, et al. Glycemic control in patients with insulinoma treated with everolimus. N Engl J Med. 2009;360:195–197. doi: 10.1056/NEJMc0806740.
    1. Kulke MH, et al. Activity of sunitinib in patients with advanced neuroendocrine tumours. J Clin Oncol. 2008;26:3403–3410. doi: 10.1200/JCO.2007.15.9020.
    1. Yao JC, et al. Everolimus for advanced pancreatic neuroendocrine tumours. N Engl J Med. 2011;364:514–523. doi: 10.1056/NEJMoa1009290.
    1. Raymond E, et al. Sunitinib malate for the treatment of pancreatic neuroendocrine tumours. N Engl J Med. 2011;364:501–513. doi: 10.1056/NEJMoa1003825.
    1. Iacovelli R, 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;7:873–879. doi: 10.3109/0284186X.2012.705019.
    1. Bodei L, et al. The joint IAEA, EANM, and SNMMI practical guidance on peptide receptor radionuclide therapy (PRRNT) in neuroendocrine tumours. Eur J Nucl Med Mol Imaging. 2013;40:800–816. doi: 10.1007/s00259-012-2330-6.
    1. Kwekkeboom DJ, et al. Treatment with the radiolabeled somatostatin analogue [177 Lu-DOTA 0,Tyr3] octreotate: toxicity, efficacy, and survival. J Clin Oncol. 2008;26:2124–2130. doi: 10.1200/JCO.2007.15.2553.
    1. Strosberg JR, et al. Netter 1 phase III: progression free-survival, radiographic response, and preliminary overall survival results in patients with midgut neuroendocrine tumours treated with 177 lu dotate. N Engl J Med. 2017;376(2):125.
    1. Leboulleux S, et al. Prediction and diagnosis of metastases in well-differentiated gastro-entero-pancreatic endocrine cancer: a prospective comparison of whole body magnetic resonance imaging and somatostatin receptor scintigraphy. J Clin Endocrinol Metab. 2008;93:3021–3028. doi: 10.1210/jc.2008-0459.
    1. Lebtahi R, et al. Detection of bone metastases in patients with endocrine GEP tumours: bone scintigraphy compared with somatostatin receptor scintigraphy. J Nucl Med. 1999;40:1602–1608.
    1. Janson ET, et al. Carcinoid tumours: analysis of prognostic factors and survival in 301 patients from a referral center. Ann Oncol. 1997;8:685–690. doi: 10.1023/A:1008215730767.
    1. Van Loon K, et al. Bone metastases and skeletal-related events from neuroendocrine tumours. Endocr Connect. 2015;4:9–17. doi: 10.1530/EC-14-0119.
    1. Van Binnebeek S, et al. Comparison of diagnostic accuracy of (111)In-pentetreotide SPECT and (68)Ga-DOTATOC PET/CT: a lesion-by-lesion analysis in patients with metastatic neuroendocrine tumours. Eur Radiol. 2016;26:900–909. doi: 10.1007/s00330-015-3882-1.
    1. Buchmann I, et al. Comparison of 68Ga-DOTATOC PET and 111In-DTPAOC (Octreoscan) SPECT in patients with neuroendocrine tumours. Eur J Nucl Med Mol Imaging. 2007;34:1617–1626. doi: 10.1007/s00259-007-0450-1.
    1. Mota JM, et al. Complications from carcinoid syndrome: review of the current evidence. Ecancermedicalscience. 2016;8:662.
    1. Kulke MH, et al. Telotristat ethyl, a tryptophan hydroxylase inhibitor for the treatment of carcinoid syndrome. J Clin Oncol. 2016;28
    1. Moerman VM, et al. Carcinoid heart disease: typical findings on echocardiography and cardiac magnetic resonance. Acta Cardiol. 2012;67:245–248.
    1. Connolly HM, et al. Early and late outcomes of surgical treatment in carcinoid heart disease. J Am Coll Cardiol. 2015;66:2189–2196. doi: 10.1016/j.jacc.2015.09.014.
    1. Jensen RT, et al. Gastrinoma (duodenal and pancreatic) Neuroendocrinology. 2006;84:173–182. doi: 10.1159/000098009.
    1. Stephen AE, Hodin RA. Neuroendocrine tumours of the pancreas, excluding gastrinoma. Surg Oncol Clin N Am. 2006;15:497–510. doi: 10.1016/j.soc.2006.05.012.
    1. Munhoz RR, et al. Combination of irinotecan and a platinum agent for poorly differentiated neuroendocrine carcinomas. Rare Tumours. 2013;5:e39. doi: 10.4081/rt.2013.e39.
    1. Bezerra EB, et al. Poorly differentiated neuroendocrine tumours. Neuroendocrine Tumour. 2011. pp. 157–176.
    1. Sorbye H, et al. GEP high-grade neuroendocrine carcinoma. Cancer. 2014;120:2814–2823. doi: 10.1002/cncr.28721.
    1. Tang LH, et al. A practical approach to the classification of WHO grade 3 (G3) well-differentiated neuroendocrine tumour (WD-NET) and poorly differentiated neuroendocrine carcinoma (PD-NEC) of the pancreas. Am J Surg Pathol. 2016;40:1192–1202. doi: 10.1097/PAS.0000000000000662.
    1. Walter DT, et al. Characteristics, prognosis and treatments of 294 patients with poorly differentiated neuroendocrine carcinoma: the FFCD-GTE national cohort. J Clin Oncol. 2015;33(May 20 Supplement):4095.
    1. Welin S, et al. Clinical effect of temozolomide-based chemotherapy in poorly differentiated endocrine carcinoma after progression on first line chemotherapy. Cancer. 2011;117:4617–4622. doi: 10.1002/cncr.26124.
    1. Marx SJ. Molecular genetics of multiple endocrine neoplasia types 1 and 2. Nat Rev Cancer. 2005;5:367–375. doi: 10.1038/nrc1610.
    1. Brandi ML, et al. Guidelines for diagnosis and therapy of MEN type 1 and type 2. J Clin Endocrinol Metab. 2001;86:5658–5671. doi: 10.1210/jcem.86.12.8070.
    1. Lourenço DM, Jr, et al. The impact of clinical and genetic screenings on the management of the multiple endocrine neoplasia type 1. Clinics. 2007;62:465–476. doi: 10.1590/S1807-59322007000400014.
    1. Thakker RV, et al. Clinical practice guidelines for multiple endocrine neoplasia type 1 (MEN1) J Clin Endocrinol Metab. 2012;97:2990–3011. doi: 10.1210/jc.2012-1230.
    1. Norton JA, et al. Surgery increases survival in patients with gastrinoma. Ann Surg. 2006;244:410–419.
    1. Bartsch DK, et al. Outcome of duodenopancreatic resections in patients with multiple endocrine neoplasia type 1. Ann Surg. 2005;242:757–764. doi: 10.1097/01.sla.0000189549.51913.d8.
    1. Jensen RT, et al. ENETS Consensus Guidelines for the management of patients with digestive neuroendocrine neoplasms: functional pancreatic endocrine tumour syndromes. Neuroendocrinology. 2012;95:98–119. doi: 10.1159/000335591.
    1. Triponez F, et al. Epidemiology data on 108 MEN 1 patients from the GTE with isolated nonfunctioning tumours of the pancreas. Ann Surg. 2006;243:265–272. doi: 10.1097/01.sla.0000197715.96762.68.
    1. Gonçalves TD, et al. Penetrance of functioning and nonfunctioning pancreatic neuroendocrine tumours in multiple endocrine neoplasia type 1 in the second decade of life. J Clin Endocrinol Metab. 2014;99:e89–96. doi: 10.1210/jc.2013-1768.
    1. Maia MC, et al. Efficacy and long-term safety of everolimus in pancreatic neuroendocrine tumour associated with multiple endocrine neoplasia type I: case report. Oncol Res Treat. 2016;39:643–645. doi: 10.1159/000448699.
    1. Webster J, et al. A comparison of cabergoline and bromocriptine in the treatment of hyperprolactinemic amenorrhea. Cabergoline Comparative Study Group. N Engl J Med. 1994;331:904–909. doi: 10.1056/NEJM199410063311403.
    1. Glezer A, Bronstein MD. Prolactinomas. Endocrinol Metab Clin North Am. 2015;44:71–78. doi: 10.1016/j.ecl.2014.11.003.
    1. Giustina A, et al. Expert consensus document: a consensus on the medical treatment of acromegaly. Nat Rev Endocrinol. 2014;10:243–248. doi: 10.1038/nrendo.2014.21.
    1. Trainer PJ, et al. A randomized, controlled, multicentre trial comparing pegvisomant alone with combination therapy of pegvisomant and long-acting octreotide in patients with acromegaly. Clin Endocrinol (Oxf) 2009;71:549–557. doi: 10.1111/j.1365-2265.2009.03620.x.
    1. Kasuki L, et al. Experience with pegvisomant treatment in acromegaly in a single Brazilian tertiary reference center: efficacy, safety and predictors of response. Arch Endocrinol Metab. 2016;60:479–485. doi: 10.1590/2359-3997000000210.
    1. McCormack AI, et al. Aggressive pituitary tumours: the role of temozolomide and the assessment of MGMT status. Eur J Clin Invest. 2011;41:1133–1148. doi: 10.1111/j.1365-2362.2011.02520.x.
    1. Libutti SK, et al. Clinical and genetic analysis of patients with pancreatic neuroendocrine tumours associated with von Hippel–Lindau disease. Surgery. 2000;128:1022–1027. doi: 10.1067/msy.2000.110239.
    1. Riechelmann R, et al. Guideline for the management of bile duct cancers by the Brazilian gastrointestinal tumour group. Arq Gastroenterol. 2016;53:5–9. doi: 10.1590/S0004-28032016000100003.
    1. Libutti SK, et al. Pancreatic neuroendocrine tumours associated with von Hippel–Lindau disease: diagnostic and management recommendations. Surgery. 1998;124:1153–1159. doi: 10.1067/msy.1998.91823.

Source: PubMed

3
Abonneren