11
Views
0
CrossRef citations to date
0
Altmetric
Review

Neoplastic lesions of endocrine cells in the gastrointestinal tract: ten evolving principles as a basis for clinical understanding

, , , , &
Pages 1-17 | Published online: 28 Dec 2012

References

  • Modlin IM, Oberg K, Chung DC, et al. Gastroenteropancreatic neuroendocrine tumours. Lancet Oncol. 2008;9(1):61–72.
  • Rindi G, Klimstra DS, Arnold R, et al. Nomenclature and classification of neuroendocrine neoplasms of the digestive system. In: Bosman FT, Carneiro F, Hruban RH, Theise ND, editors. WHO Classification of the Digestive System. 4th ed. Lyon, France: International Agency for Research on Cancer; 2010.
  • Gustafsson BI, Bakke I, Tommeras K, Waldum HL. A new method for visualization of gut mucosal cells, describing the enterochro- maffin cell in the rat gastrointestinal tract. Scand J Gastroenterol. 2006;41(4):390–395.
  • Solcia E. Histological Typing of Endocrine Tumors. World Health Organization International Histological Classification of Tumors. 2nd ed. Berlin, Germany: Springer; 2000.
  • 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(28):4656–4663.
  • 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.
  • Yao JC, Shah MH, Ito T, et al. Everolimus for advanced pancreatic neuroendocrine tumors. N Engl J Med. 2011;364(6):514–523.
  • Kwekkeboom DJ, Teunissen JJ, Bakker WH, et al. Radiolabeled somatostatin analog [177Lu-D0TA0,Tyr3]octreotate in patients with endocrine gastroenteropancreatic tumors. J Clin Oncol. 2005;23(12):2754–2762.
  • Schonhoff SE, Giel-Moloney M, Leiter AB. Minireview: development and differentiation of gut endocrine cells. Endocrinology. 2004;145(6):2639–2644.
  • Power DG, Shia J, Allen PJ, Jarnagin WR, O’Reilly EM. Synchronous epithelial and neuroendocrine cancers of the pancreas: case series of a rare occurrence. Clin Colorectal Cancer. 2011;10(2):146–150.
  • Oberndorfer S. Karzinoide Tumoren des Dünndarms. [Carcinoid tumors of the small intestine]. FrankfZ Pathol. 1907;1:426–432. Germany.
  • Williams ED, Sandler M. The classification of carcinoid tumours. Lancet. 1963;1(7275):238–239.
  • Solcia E. Histological typing of endocrine tumors. World Health Organization International Histological Classification of Tumors. 2nd ed. Berlin, Germany: Springer; 2000.
  • Rindi G, Bordi C, Rappel S, La Rosa S, Stolte M, Solcia E. Gastric carcinoids and neuroendocrine carcinomas: pathogenesis, pathology, and behavior. World JSurg. 1996;20(2):168–172.
  • Plockinger U, Rindi G, Arnold R, et al. Guidelines for the diagnosis and treatment of neuroendocrine gastrointestinal tumours. A consensus statement on behalf of the European Neuroendocrine Tumour Society (ENETS). Neuroendocrinology. 2004;80(6):394–424.
  • 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.
  • Klimstra DS, Modlin IR, Coppola D, Lloyd RV, Suster S. The pathologic classification of neuroendocrine tumors: a review of nomenclature, grading, and staging systems. Pancreas. 2010;39(6):707–712.
  • 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.
  • Jann H, Roll S, Couvelard A, et al. Neuroendocrine tumors of midgut and hindgut origin: tumor-node-metastasis classification determines clinical outcome. Cancer. 2011;117(15):3332–3341.
  • Strosberg JR, Cheema A, Weber J, Han G, Coppola D, Kvols LK. Prognostic validity of a novel American Joint Committee on Cancer Staging Classification for pancreatic neuroendocrine tumors. J Clin Oncol. 2011;29(22):3044–3049.
  • Panzuto F, Nasoni S, Falconi M, et al. Prognostic factors and survival in endocrine tumor patients: comparison between gastrointestinal and pancreatic localization. Endocr Relat Cancer. 2005;12(4):1083–1092.
  • Rindi G, Solcia E. Endocrine hyperplasia and dysplasia in the pathogenesis of gastrointestinal and pancreatic endocrine tumors. Gastroenterol Clin North Am. 2007;36(4):851–865, vi.
  • Rindi G, Inzani F, Solcia E. Pathology of gastrointestinal disorders. Endocrinol Metab Clin North Am. 2010;39(4):713–727.
  • Bordi C, Yu JY, Baggi MT, et al. Gastric carcinoids and their precursor lesions. A histologic and immunohistochemical study of 23 cases. Cancer. 1991;67(3):663–672.
  • Lawrence B, Kidd M, Svejda B, Modlin I. A clinical perspective on gastric neuroendocrine neoplasia. Curr Gastroenterol Rep. 2011;13(1):101–109.
  • Berge T, Linell F. Carcinoid tumours. Frequency in a defined population during a 12-year period. Acta Pathol Microbiol Scand A. 1976;84(4):322–330.
  • Lam KY, Lo CY. Pancreatic endocrine tumour: a 22-year clinico- pathological experience with morphological, immunohistochemi- cal observation and a review of the literature. Eur J Surg Oncol. 1997;23(1):36–42.
  • Lawrence B, Gustafsson BI, Chan A, Svejda B, Kidd M, Modlin IM. The epidemiology of gastroenteropancreatic neuroendocrine tumors. Endocrinol Metab Clin North Am. 2011;40(1):1–18, vii.
  • 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(18):3063–3072.
  • Lindor NM, McMaster ML, Lindor CJ, Greene MH. Concise handbook of familial cancer susceptibility syndromes – second edition. J Natl Cancer Inst Monogr. 2008;38:1–93.
  • Larsson C, Skogseid B, Oberg K, Nakamura Y, Nordenskjold M. Multiple endocrine neoplasia type 1 gene maps to chromosome 11 and is lost in insulinoma. Nature. 1988;332(6159):85–87.
  • Marx SJ, Agarwal SK, Kester MB, et al. Germline and somatic mutation of the gene for multiple endocrine neoplasia type 1 (MEN1). J Intern Med. 1998;243(6):447–453.
  • Fujii T, Kawai T, Saito K, et al. MEN1 gene mutations in sporadic neuroendocrine tumors of foregut derivation. Pathol Int. 1999;49(11):968–973.
  • Latif F, Tory K, Gnarra J, et al. Identification of the von Hippel-Lindau disease tumor suppressor gene. Science. 1993;260(5112):1317–1320.
  • Maxwell PH, Wiesener MS, Chang GW, et al. The tumour suppressor protein VHL targets hypoxia-inducible factors for oxygen-dependent proteolysis. Nature. 1999;399(6733):271–275.
  • 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(6021):1199–1203.
  • 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(2):245–255.
  • Zhou CF, Ji J, Yuan F, et al. mTOR activation in well differentiated pancreatic neuroendocrine tumors: a retrospective study on 34 cases. Hepatogastroenterology. 2011;58(112):2140–2143.
  • Heaphy CM, de Wilde RF, Jiao Y, et al. Altered telomeres in tumors with ATRX and DAXX mutations. Science. 2011;333(6041):425.
  • Yachida S, Vakiani E, White CM, et al. Small cell and large cell neuroendocrine carcinomas of the pancreas are genetically similar and distinct from well-differentiated pancreatic neuroendocrine tumors. Am J Surg Pathol. 2012;36(2):173–184.
  • Hu W, Feng Z, Modica I, et al. Gene amplifications in well-differentiated pancreatic neuroendocrine tumors inactivate the p53 pathway. Genes Cancer. 2010;1(4):360–368.
  • Hauso O, Gustafsson BI, Kidd M, et al. Neuroendocrine tumor epidemiology: contrasting Norway and North America. Cancer. 2008;113(10):2655–2664.
  • Qubaiah O, Devesa SS, Platz CE, Huycke MM, Dores GM. Small intestinal cancer: a population-based study of incidence and survival patterns in the United States, 1992 to 2006. Cancer Epidemiol Biomarkers Prev. 2010;19(8):1908–1918.
  • Garcia-Carbonero R, Capdevila J, Crespo-Herrero G, et al. Incidence, patterns of care and prognostic factors for outcome of gastroen- teropancreatic neuroendocrine tumors (GEP-NETs): results from the National Cancer Registry of Spain (RGETNE). Ann Oncol. 2010;21(9):1794–1803.
  • Modlin IM, Lye KD, Kidd M. A 5-decade analysis of 13,715 carcinoid tumors. Cancer. 2003;97(4):934–959.
  • Niederle MB, Hackl M, Kaserer K, Niederle B. Gastroenteropancreatic neuroendocrine tumours: the current incidence and staging based on the WHO and European Neuroendocrine Tumour Society classification: an analysis based on prospectively collected parameters. Endocr Relat Cancer. 2010;17(4):909–918.
  • Pape UF, Berndt U, Muller-Nordhorn J, et al. Prognostic factors of long-term outcome in gastroenteropancreatic neuroendocrine tumours. Endocr Relat Cancer. 2008;15(4):1083–1097.
  • Hanahan D, Weinberg RA. The hallmarks of cancer. Cell. 2000;100(1):57–70.
  • Kurzrock R, Gutterman JU, Talpaz M. The molecular genetics of Philadelphia chromosome-positive leukemias. N Engl J Med. 1988;319(15):990–998.
  • Miettinen M, Lasota J. Gastrointestinal stromal tumors: review on morphology, molecular pathology, prognosis, and differential diagnosis. Arch Pathol Lab Med. 2006;130(10):1466–1478.
  • Jamieson S, Fuller PJ. Molecular pathogenesis of granulosa cell tumors of the ovary. Endocr Rev. 2012;33(1):109–144.
  • Kirshbom PM, Kherani AR, Onaitis MW, Feldman JM, Tyler DS. Carcinoids of unknown origin: comparative analysis with foregut, midgut, and hindgut carcinoids. Surgery. 1998;124(6):1063–1070.
  • Saclarides TJ, Szeluga D, Staren ED. Neuroendocrine cancers of the colon and rectum. Results of a ten-year experience. Dis Colon Rectum. 1994;37(7):635–642.
  • Bernick PE, Klimstra DS, Shia J, et al. Neuroendocrine carcinomas of the colon and rectum. Dis Colon Rectum. 2004;47(2):163–169.
  • Ahlman H, Nilsson O, McNicol AM, et al. Poorly-differentiated endocrine carcinomas of midgut and hindgut origin. Neuroendocrinology. 2008;87(1):40–46.
  • Nilsson O, Van Cutsem E, Delle Fave G, et al. Poorly differentiated carcinomas of the foregut (gastric, duodenal and pancreatic). Neuroendocrinology. 2006;84(3):212–215.
  • Olsen IH, Langer SW, Jepsen I, et al. First-line treatment of patients with disseminated poorly differentiated neuroendocrine carcinomas with carboplatin, etoposide, and vincristine: a single institution experience. Acta Oncol. 2012;51(1):97–100.
  • 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(6):799–800.
  • Lindholm DP, Eriksson B, Granberg D. Response to temozolomide and bevacizumab in a patient with poorly differentiated neuroendocrine carcinoma. Med Oncol. 2012;29(1):301–303.
  • Catena L, Bajetta E, Milione M, et al. Mammalian target of rapamycin expression in poorly differentiated endocrine carcinoma: clinical and therapeutic future challenges. Target Oncol. 2011;6(2):65–68.
  • Delle Fave G, Kwekkeboom DJ, Van Cutsem E, et al. ENETS consensus guidelines for the management of patients with gastroduodenal neoplasms. Neuroendocrinology. 2012;95(2):74–87.
  • Ozao-Choy J, Buch K, Strauchen JA, Warner RR, Divino CM. Laparoscopic antrectomy for the treatment of type I gastric carcinoid tumors. J Surg Res. 2010;162(1):22–25.
  • Ruszniewski P, Delle Fave G, Cadiot G, et al. Well-differentiated gastric tumors/carcinomas. Neuroendocrinology. 2006;84(3):158–164.
  • Modlin IM, Kumar R, Nangia A, Soroka CJ, Pasikhov D, Goldenring JR. Gastrin-dependent inhibitory effects of octreotide on the genesis of gastric ECLomas. Surgery. 1992;112(6):1048–1056.
  • Tomassetti P, Migliori M, Caletti GC, Fusaroli P, Corinaldesi R, Gullo L. Treatment of type II gastric carcinoid tumors with somatostatin analogues. N Engl J Med. 2000;343(8):551–554.
  • Jianu CS, Fossmark R, Syversen U, Hauso O, Fykse V, Waldum HL. Five-year follow-up of patients treated for 1 year with octreotide long-acting release for enterochromaffin-like cell carcinoids. Scand J Gastroenterol. 2011;46(4):456–463.
  • Falconi M, Bartsch DK, Eriksson B, et al. ENETS consensus guidelines for the management of patients with digestive neuroendocrine neoplasms of the digestive system: well-differentiated pancreatic non-functioning tumors. Neuroendocrinology. 2012;95(2):120–134.
  • 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.
  • Falconi M, Plockinger U, Kwekkeboom DJ, et al. Well-differentiated pancreatic nonfunctioning tumors/carcinoma. Neuroendocrinology. 2006;84(3):196–211.
  • Panzuto F, Di Fonzo M, Iannicelli E, et al. Long-term clinical outcome of somatostatin analogues for treatment of progressive, metastatic, well-differentiated entero-pancreatic endocrine carcinoma. Ann Oncol. 2006;17(3):461–466.
  • Aparicio T, Ducreux M, Baudin E, et al. Antitumour activity of somatostatin analogues in progressive metastatic neuroendocrine tumours. Eur J Cancer. 2001;37(8):1014–1019.
  • Faiss S, Pape UF, Bohmig M, et al. Prospective, randomized, multicenter trial on the antiproliferative effect of lanreotide, interferon alfa, and their combination for therapy of metastatic neuroendocrine gastroenteropan- creatic tumors – the International Lanreotide and Interferon Alfa Study Group. J Clin Oncol. 2003;21(14):2689–2696.
  • Moertel CG, Lefkopoulo M, Lipsitz S, Hahn RG, Klaassen D. Streptozocin-doxorubicin, streptozocin-fluorouracil or chlorozotocin in the treatment of advanced islet-cell carcinoma. N Engl J Med. 1992;326(8):519–523.
  • Kouvaraki MA, Ajani JA, Hoff P, et al. Fluorouracil, doxorubicin, and streptozocin in the treatment of patients with locally advanced and metastatic pancreatic endocrine carcinomas. J Clin Oncol. 2004;22(23):4762–4771.
  • Bajetta E, Catena L, Procopio G, et al. Are capecitabine and oxali- platin (XELOX) suitable treatments for progressing low-grade and high-grade neuroendocrine tumours? Cancer Chemother Pharmacol. 2007;59(5):637–642.
  • Ducreux MP, Boige V, Leboulleux S, et al. A phase II study of irinotecan with 5-fluorouracil and leucovorin in patients with pretreated gastro- enteropancreatic well-differentiated endocrine carcinomas. Oncology. 2006;70(2):134–140.
  • Strosberg JR, Fine RL, Choi J, et al. First-line chemotherapy with capecitabine and temozolomide in patients with metastatic pancreatic endocrine carcinomas. Cancer. 2011;117(2):268–275.
  • Hobday TJ, Qin R, Reidy DL, et al. Multicenter Phase II trial of temsi- rolimus and bevacizumab in pancreatic neuroendocrine tumors: results of a planned interim efficacy analysis. Paper presented at the American Society of Clinical Oncology Gastrointestinal Symposium, January 19-21, 2012, San Francisco, CA.
  • Broglio KR, Berry DA. Detecting an overall survival benefit that is derived from progression-free survival. J Natl Cancer Inst. 2009;101(23):1642–1649.
  • Grande E, Jose Diez J, Pachon V, et al. Response by Choi criteria to sunitinib plus octreotide LAR in a functional heavily pretreated advanced pancreatic neuroendocrine tumor. Anticancer Drugs. 2011;22(5):477–479.
  • Faivre S, Ronot M, Dreyer C, et al. Imaging response in neuroendocrine tumors treated with targeted therapies: the experience of sunitinib. Target Oncol. 2012;7(2):127–133.
  • Kwekkeboom DJ, Mueller-Brand J, Paganelli G, et al. Overview of results of peptide receptor radionuclide therapy with 3 radiolabeled somatostatin analogs. J Nucl Med. 2005;46 Suppl 1:62S—66S.
  • van Essen M, Krenning EP, Kam BL, de Herder WW, van Aken MO, Kwekkeboom DJ. Report on short-term side effects of treatments with 177Lu-octreotate in combination with capecitabine in seven patients with gastroenteropancreatic neuroendocrine tumours. Eur J Nucl Med Mol Imaging. 2008;35(4):743–748.
  • Kong G, Johnston V, Ramdave S, Lau E, Rischin D, Hicks RJ. High-administered activity In-111 octreotide therapy with concomitant radiosensitizing 5FU chemotherapy for treatment of neuroendocrine tumors: preliminary experience. Cancer Biother Radiopharm. 2009;24(5):527–533.
  • De Jong M, Valkema R, Jamar F, et al. Somatostatin receptor-targeted radionuclide therapy of tumors: preclinical and clinical findings. Semin Nucl Med. 2002;32(2):133–140.
  • Villard L, Romer A, Marincek N, et al. Cohort study of somatostatin- based radiopeptide therapy with [90Y-D0TA]-T0C versus [90Y- D0TA]-T0C plus [177Lu-D0TA]-T0C in neuroendocrine cancers. J Clin Oncol. 2012;30(10):1100–1106.
  • Forrer F, Krenning EP, Kooij PP, et al. Bone marrow dosimetry in peptide receptor radionuclide therapy with [177Lu-D0TA(0),Tyr(3)] octreotate. Eur J Nucl Med Mol Imaging. 2009;36(7):1138–1146.
  • Sun W, Lipsitz S, Catalano P, Mailliard JA, Haller DG; Eastern Cooperative 0ncology G. Phase II/III study of doxorubicin with fluorouracil compared with streptozocin with fluorouracil or dacar- bazine in the treatment of advanced carcinoid tumors: Eastern Cooperative 0ncology Group Study E1281. J Clin Oncol. 2005;23(22):4897–4904.
  • Eriksson B, Kloppel G, Krenning E, et al. Consensus guidelines for the management of patients with digestive neuroendocrine tumors – well-differentiated jejunal-ileal tumor/carcinoma. Neuroendocrinology. 2008;87(1):8–19.
  • Ahlman H, Wangberg B, Jansson S, et al. Interventional treatment of gastrointestinal neuroendocrine tumours. Digestion. 2000;62 Suppl 1:59–68.
  • Kam BL, Teunissen JJ, Krenning EP, et al. Lutetium-labelled peptides for therapy of neuroendocrine tumours. Eur J Nucl Med Mol Imaging. 2012;39 Suppl 1:S103–S112.
  • Florio T. Molecular mechanisms of the antiproliferative activity of somatostatin receptors (SSTRs) in neuroendocrine tumors. Front Biosci. 2008;13:822–840.
  • Chua YJ, Michael M, Zalcberg JR, et al. Antitumor effect of somatostatin analogs in neuroendocrine tumors. J Clin Oncol. 2010;28(3): e41–e42.
  • Schmid HA. Pasireotide (S0M230): development, mechanism of action and potential applications. Mol Cell Endocrinol. 2008;286(1–2):69–74.
  • Strosberg J, Kvols L. Antiproliferative effect of somatostatin analogs in gastroenteropancreatic neuroendocrine tumors. World J Gastroenterol. 2010;16(24):2963–2970.
  • Colao A, Petersenn S, Newell-Price J, et al. A 12-month phase 3 study of pasireotide in Cushing’s disease. N Engl J Med. 2012;366(10):914–924.
  • Yao JC. Neuroendocrine tumors. Molecular targeted therapy for carcinoid and islet-cell carcinoma. Best Pract Res Clin Endocrinol Metab. 2007;21(1):163–172.
  • Kulke MH, Lenz HJ, Meropol NJ, et al. Activity of sunitinib in patients with advanced neuroendocrine tumors. J Clin Oncol. 2008;26(20):3403–3410.
  • Hobday TJ, Rubin J, Holen K. MC044h, a Phase II trial of sorafenib in patients (pts) with metastatic neuroendocrine tumors (NET): a Phase II consortium (P2C) study. J Clin Oncol. 2007;25(18S):4504.
  • Pavel ME, Bartel C, Heuck F. Open-label, non-randomized, multicenter phase II study evaluating the angiogenesis inhibitor PTK787/ ZK222584 (PTK/ZK) in patients with advanced neuroendocrine carcinomas (NEC). J Clin Oncol. 2008;26 Suppl:14684.
  • Hobday TJ, Holen K, Donehower R. A Phase II trial of gefitinib in patients (pts) with progressive metastatic neuroendocrine tumors (NET): a Phase II consortium (P2C) study. J Clin Oncol. 2006;24(18S):189s.
  • Duran I, Kortmansky J, Singh D, et al. A Phase II clinical and pharmacodynamic study of temsirolimus in advanced neuroendocrine carcinomas. Br J Cancer. 2006;95(9):1148–1154.
  • 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(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 andomized, placebo-controlled, phase 3 study. Lancet. 2011;378(9808):2005–2012.
  • Pape UF, Perren A, Niederle B, 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(2):135–156.
  • Plockinger U, Couvelard A, Falconi M, et al. Consensus guidelines for the management of patients with digestive neuroendocrine tumours: well-differentiated tumour/carcinoma of the appendix and goblet cell carcinoma. Neuroendocrinology. 2008;87(1):20–30.
  • Caplin M, Sundin A, Nillson O, et al. ENETS consensus guidelines for the management of patients with digestive neuroendocrine neoplasms: colorectal neuroendocrine neoplasms. Neuroendocrinology. 2012;95(2):88–97.
  • Kaltsas GA, Besser GM, Grossman AB. The diagnosis and medical management of advanced neuroendocrine tumors. Endocr Rev. 2004;25(3):458–511.
  • Basu B, Sirohi B, Corrie P. Systemic therapy for neuroendocrine tumours of gastroenteropancreatic origin. Endocr Relat Cancer. 2010;17(1):R75–R90.
  • Modlin IM, Kidd M, Drozdov I, Siddique ZL, Gustafsson BI. Pharmacotherapy of neuroendocrine cancers. Expert Opin Pharmacother. 2008;9(15):2617–2626.
  • 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(1):69–76.
  • Taupenot L, Harper KL, O’Connor DT. The chromogranin- secretogranin family. N Engl J Med. 2003;348(12):1134–1149.
  • Bajetta E, Ferrari L, Martinetti A, et al. Chromogranin A, neuron specific enolase, carcinoembryonic antigen, and hydroxyindole acetic acid evaluation in patients with neuroendocrine tumors. Cancer. 1999;86(5):858–865.
  • Seregni E, Ferrari L, Bajetta E, Martinetti A, Bombardieri E. Clinical significance of blood chromogranin A measurement in neuroendocrine tumours. Ann Oncol. 2001;12 Suppl 2:S69–S72.
  • Lawrence B, Gustafsson BI, Kidd M, Pavel M, Svejda B, Modlin IM. The clinical relevance of chromogranin A as a biomarker for gas- troenteropancreatic neuroendocrine tumors. Endocrinol Metab Clin North Am. 2011;40(1):111–134.
  • Oberg K, Stridsberg M. Chromogranins as diagnostic and prognostic markers in neuroendocrine tumours. Adv Exp Med Biol. 2000;482:329–337.
  • Boudreaux JP, Klimstra DS, Hassan MM, et al. The NANETS consensus guideline for the diagnosis and management of neuroendocrine tumors: well-differentiated neuroendocrine tumors of the jejunum, ileum, appendix, and cecum. Pancreas. 2010;39(6):753–766.
  • Janson ET, Holmberg L, Stridsberg M, et al. Carcinoid tumors: analysis of prognostic factors and survival in 301 patients from a referral center. Ann Oncol. 1997;8(7):685–690.
  • Kulke MH, Siu LL, Tepper JE, et al. Future directions in the treatment of neuroendocrine tumors: consensus report of the National Cancer Institute Neuroendocrine Tumor clinical trials planning meeting. J Clin Oncol. 2011;29(7):934–943.
  • Williams ED. Histological Typing of Tumors of the Enodcrine System. Berlin, Germany: Springer-Verlag; 1980.
  • Ehehalt F, Saeger HD, Schmidt CM, Grutzmann R. Neuroendocrine tumors of the pancreas. Oncologist. 2009;14(5):456–467.
  • Jensen RT. Pancreatic endocrine tumors: recent advances. Ann Oncol. 1999;10 Suppl 4:170–176.
  • Jensen R. Endocrine tumors of the gastrointestinal tract and pancreas. In: Kasper DL, Fauci AS, Braunwald E, Longo DL, JL Jameson JL, editors. Harrison’s Principles of Internal Medicine. 17th ed. New York, NY: McGraw-Hill; 2008:2.
  • Schimmack S, Svejda B, Lawrence B, Kidd M, Modlin IM. The diversity and commonalities of gastroenteropancreatic neuroendocrine tumors. Langenbecks Arch Surg. 2011;396(3):273–298.