Yamada’s Textbook of Gastroenterology

Sixth Edition

Edited by Daniel K. Podolsky,
Michael Camilleri, J. Gregory Fitz,
Anthony N. Kalloo, Fergus Shanahan, Timothy C. Wang

References

Neuroendocrine tumors of the pancreas

1. ModlinI, ShapiroM, KiddM. Siegfried Oberndorfer: origins and perspectives of carcinoid tumors. Hum Pathol2004;35:1440. CrossRef

2. LawrenceB, GustafssonB, ChanA, et al.The epidemiology of gastroenteropancreatic neuroendocrine tumors. Endocrinol Metab Clin North Am2011;40:1. CrossRef

3. CarriagaM, HensonD. Liver, gallbladder, extrahepatic bile ducts, and pancreas. Cancer1995;75(1 Suppl):171. CrossRef

4. BuchananK, JohnstonC, O'HareM, et al.Neuroendocrine tumors. A European view. Am J Med1986;81:14. CrossRef

5. ErikssonB, ObergK, SkogseidB. Neuroendocrine pancreatic tumors. Clinical findings in a prospective study of 84 patients. Acta Oncol (Stockholm, Sweden)1989;28:373. CrossRef

6. WatsonR, JohnstonC, O'HareM, et al.The frequency of gastrointestinal endocrine tumours in a well‐defined population–Northern Ireland 1970–1985. Q J Med1989;72:647.

7. LamK, LoC. Pancreatic endocrine tumour: a 22‐year clinico‐pathological experience with morphological, immunohistochemical observation and a review of the literature. Eur J Surg Oncol1997;23:36. CrossRef

8. MoldowR, ConnellyR. Epidemiology of pancreatic cancer in Connecticut. Gastroenterology1968;55:677.

9. LepageC, BouvierA, PhelipJ, et al.Incidence and management of malignant digestive endocrine tumours in a well defined French population. Gut2004;53:549. CrossRef

10. GrimeliusL, HultquistG, StenkvistB. Cytological differentiation of asymptomatic pancreatic islet cell tumours in autopsy material. Virchows Arch A Pathol Anat Histol1975;365:275. CrossRef

11. KimuraW, KurodaA, MoriokaY. Clinical pathology of endocrine tumors of the pancreas. Analysis of autopsy cases. Dig Dis Sci1991;36:933. CrossRef

12. RajaA, WrightC, SodicksonA, et al.Negative appendectomy rate in the era of CT: an 18‐year perspective. Radiology2010;256:460. CrossRef

13. RekhtmanN. Neuroendocrine tumors of the lung: an update. Arch Pathol Lab Med2010;134:1628.

14. ModlinIM, ObergK, ChungDC, et al.Gastroenteropancreatic neuroendocrine tumours. Lancet Oncol2008;9:61. CrossRef

15. HalfdanarsonT, RabeK, RubinJ, et al.Pancreatic neuroendocrine tumors (PNETs): incidence, prognosis and recent trend toward improved survival. Ann Oncol2008;19:1727. CrossRef

16. HausoO, GustafssonB, KiddM, et al.Neuroendocrine tumor epidemiology: contrasting Norway and North America. Cancer2008;113:2655. CrossRef

17. ItoT, IgarashiH, NakamuraK, et al.Epidemiological trends of pancreatic and gastrointestinal neuroendocrine tumors in Japan: a nationwide survey analysis. J Gastroenterol2014; epub ahead of print.

18. BoudreauxJ, KlimstraD, HassanM, 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. Pancreas2010;39:753. CrossRef

19. HaselkornT, WhittemoreA, LilienfeldD. Incidence of small bowel cancer in the United States and worldwide: geographic, temporal, and racial differences. Cancer Causes Control2005;16:781. CrossRef

20. YaoJ, HassanM, PhanA, 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 Oncol2008;26:3063. CrossRef

21. GuG, BrownJ, MeltonD. Direct lineage tracing reveals the ontogeny of pancreatic cell fates during mouse embryogenesis. Mech Dev2003;120:35. CrossRef

22. BarkerN, van EsJ, KuipersJ, et al.Identification of stem cells in small intestine and colon by marker gene Lgr5. Nature2007;449:1003. CrossRef

23. DorY, BrownJ, MartinezO, et al.Adult pancreatic beta‐cells are formed by self‐duplication rather than stem‐cell differentiation. Nature2004;429:41. CrossRef

24. XuX, D'HokerJ, StangéG, et al.Beta cells can be generated from endogenous progenitors in injured adult mouse pancreas. Cell2008;132:197. CrossRef

25. VortmeyerA, HuangS, LubenskyI, et al.Non‐islet origin of pancreatic islet cell tumors. J Clin Endocrinol Metab2004;89:1934. CrossRef

26. PerrenA, AnlaufM, HenoppT, et al.Multiple endocrine neoplasia type 1 (MEN1): loss of one MEN1 allele in tumors and monohormonal endocrine cell clusters but not in islet hyperplasia of the pancreas. J Clin Endocrinol Metab2007;92:1118. CrossRef

27. CrabtreeJ, ScacheriP, WardJ, et al.Of mice and MEN1: insulinomas in a conditional mouse knockout. Mol Cell Biol2003;23:6075. CrossRef

28. BertolinoP, TongW‐M, HerreraP, et al.Pancreatic beta‐cell‐specific ablation of the multiple endocrine neoplasia type 1 (MEN1) gene causes full penetrance of insulinoma development in mice. Cancer Res2003;63:4836.

29. BiondiC, GartsideM, WaringP, et al.Conditional inactivation of the MEN1 gene leads to pancreatic and pituitary tumorigenesis but does not affect normal development of these tissues. Mol Cell Biol2004;24:3125. CrossRef

30. ThakkerR. Multiple endocrine neoplasia type 1 (MEN1). Best Pract Res Clin Endocrinol Metab2010;24:355. CrossRef

31. LemosM, ThakkerR. Multiple endocrine neoplasia type 1 (MEN1): analysis of 1336 mutations reported in the first decade following identification of the gene. Hum Mutat2008;29:22. CrossRef

32. PellegataN, Quintanilla‐MartinezL, SiggelkowH, et al.Germ‐line mutations in p27Kip1 cause a multiple endocrine neoplasia syndrome in rats and humans. Proc Natl Acad Sci U S A2006;103:15558. CrossRef

33. ChenYX, YanJ, KeeshanK, et al.The tumor suppressor menin regulates hematopoiesis and myeloid transformation by influencing Hox gene expression. Proc Natl Acad Sci U S A2006;103:1018. CrossRef

34. MilneT, HughesC, LloydR, et al.Menin and MLL cooperatively regulate expression of cyclin‐dependent kinase inhibitors. Proc Natl Acad Sci U S A2005;102:749. CrossRef

35. KarnikS, HughesC, GuX, et al.Menin regulates pancreatic islet growth by promoting histone methylation and expression of genes encoding p27Kip1 and p18INK4c. Proc Natl Acad Sci U S A2005;102:14659. CrossRef

36. RoymansD, VissenbergK, De JongheC, et al.Identification of the tumor metastasis suppressor Nm23‐H1/Nm23‐R1 as a constituent of the centrosome. Exp Cell Res2001;262:145. CrossRef

37. YaguchiH, OhkuraN, TsukadaT, et al.Menin, the multiple endocrine neoplasia type 1 gene product, exhibits GTP‐hydrolyzing activity in the presence of the tumor metastasis suppressor nm23. J Biol Chem2002;277:38197. CrossRef

38. SchneppR, HouZ, WangH, et al.Functional interaction between tumor suppressor menin and activator of S‐phase kinase. Cancer Res2004;64:6791. CrossRef

39. SukhodoletsK, HickmanA, AgarwalS, et al.The 32‐kilodalton subunit of replication protein A interacts with menin, the product of the MEN1 tumor suppressor gene. Mol Cell Biol2003;23:493. CrossRef

40. JinS, MaoH, SchneppR, et al.Menin associates with FANCD2, a protein involved in repair of DNA damage. Cancer Res2003;63:4204.

41. MaherE, IseliusL, YatesJ, et al.Von Hippel‐Lindau disease: a genetic study. J Med Genet1991;28:443. CrossRef

42. RasmussenS, FriedmanJ. NF1 gene and neurofibromatosis 1. Am J Epidemiol2000;151:33. CrossRef

43. WilliamsV, LucasJ, BabcockM, et al.Neurofibromatosis type 1 revisited. Pediatrics2009;123:124. CrossRef

44. WallaceM, MarchukD, AndersenL, et al.Type 1 neurofibromatosis gene: identification of a large transcript disrupted in three NF1 patients. Science (New York, NY)1990;249:181. CrossRef

45. CawthonR, WeissR, XuG, et al.A major segment of the neurofibromatosis type 1 gene: cDNA sequence, genomic structure, and point mutations. Cell1990;62:193. CrossRef

46. BasuT, GutmannD, FletcherJ, et al.Aberrant regulation of ras proteins in malignant tumour cells from type 1 neurofibromatosis patients. Nature1992;356:713. CrossRef

47. CrinoP, NathansonK, HenskeE. The tuberous sclerosis complex. N Engl J Med2006;355:1345. CrossRef

48. OsborneJ, FryerA, WebbD. Epidemiology of tuberous sclerosis. Ann N Y Acad Sci1991;615:125. CrossRef

49. SaucedoL, GaoX, ChiarelliD, et al.Rheb promotes cell growth as a component of the insulin/TOR signalling network. Nat Cell Biol2003;5:566. CrossRef

50. ZhangY, GaoX, SaucedoL, et al.Rheb is a direct target of the tuberous sclerosis tumour suppressor proteins. Nat Cell Biol2003;5:578. CrossRef

51. HessmanO, LindbergD, EinarssonA, et al.Genetic alterations on 3p, 11q13, and 18q in nonfamilial and MEN 1‐associated pancreatic endocrine tumors. Genes Chromosomes Cancer1999;26:258. CrossRef

52. LöllgenR, HessmanO, SzaboE, et al.Chromosome 18 deletions are common events in classical midgut carcinoid tumors. Int J Cancer2001;92:812. CrossRef

53. WangG, YaoJ, WorahS, et al.Comparison of genetic alterations in neuroendocrine tumors: frequent loss of chromosome 18 in ileal carcinoid tumors. Mod Pathol2005;18:1079. CrossRef

54. AnderssonE, SwärdC, StenmanG, et al.High‐resolution genomic profiling reveals gain of chromosome 14 as a predictor of poor outcome in ileal carcinoids. Endocr Relat Cancer2009;16:953. CrossRef

55. CunninghamJ, Díaz de StåhlT, SjöblomT, et al.Common pathogenetic mechanism involving human chromosome 18 in familial and sporadic ileal carcinoid tumors. Genes Chromosomes Cancer2011;50:82. CrossRef

56. KytöläS, HöögA, NordB, et al.Comparative genomic hybridization identifies loss of 18q22‐qter as an early and specific event in tumorigenesis of midgut carcinoids. Am J Pathol2001;158:1803. CrossRef

57. KulkeM, FreedE, ChiangD, et al.High‐resolution analysis of genetic alterations in small bowel carcinoid tumors reveals areas of recurrent amplification and loss. Genes Chromosomes Cancer2008;47:591. CrossRef

58. CapursoG, FestaS, ValenteR, et al.Molecular pathology and genetics of pancreatic endocrine tumours. J Mol Endocrinol2012;49:50. CrossRef

59. WangE, EbrahimiS, WuA, et al.Mutation of the MENIN gene in sporadic pancreatic endocrine tumors. Cancer Res1998;58:4417.

60. ZhuangZ, VortmeyerA, PackS, et al.Somatic mutations of the MEN1 tumor suppressor gene in sporadic gastrinomas and insulinomas. Cancer Res1997;57:4682.

61. HessmanO, LindbergD, SkogseidB, et al.Mutation of the multiple endocrine neoplasia type 1 gene in nonfamilial, malignant tumors of the endocrine pancreas. Cancer Res1998;58:377.

62. MooreP, MissiagliaE, AntonelloD, et al.Role of disease‐causing genes in sporadic pancreatic endocrine tumors: MEN1 and VHL. Genes Chromosomes Cancer2001;32:177. CrossRef

63. JiaoY, ShiC, EdilB, et al.DAXX/ATRX, MEN1, and mTOR pathway genes are frequently altered in pancreatic neuroendocrine tumors. Science (New York, NY)2011;331:1199. CrossRef

64. ArnoldC, SosnowskiA, Schmitt‐GräffA, et al.Analysis of molecular pathways in sporadic neuroendocrine tumors of the gastro‐entero‐pancreatic system. Int J Cancer2007;120:2157. CrossRef

65. ChungD, SmithA, LouisD, et al.A novel pancreatic endocrine tumor suppressor gene locus on chromosome 3p with clinical prognostic implications. J Clin Invest1997;100:404. CrossRef

66. SchmittA, SchmidS, RudolphT, et al.VHL inactivation is an important pathway for the development of malignant sporadic pancreatic endocrine tumors. Endocr Relat Cancer2009;16:1219. CrossRef

67. MissiagliaE, DalaiI, BarbiS, et al.Pancreatic endocrine tumors: expression profiling evidences a role for AKT‐mTOR pathway. J Clin Oncol2010;28:245. CrossRef

68. CorboV, BeghelliS, BersaniS, et al.Pancreatic endocrine tumours: mutational and immunohistochemical survey of protein kinases reveals alterations in targetable kinases in cancer cell lines and rare primaries. Ann Oncol2012;23:127. CrossRef

69. KasajimaA, PavelM, Darb‐EsfahaniS, et al.mTOR expression and activity patterns in gastroenteropancreatic neuroendocrine tumours. Endocr Relat Cancer2011;18:181. CrossRef

70. EversB, RadyP, SandovalK, et al.Gastrinomas demonstrate amplification of the HER‐2/neu proto‐oncogene. Ann Surg1994;219:596. CrossRef

71. LohmannD, FunkA, NiedermeyerH, et al.Identification of p53 gene mutations in gastrointestinal and pancreatic carcinoids by nonradioisotopic SSCA. Virchows Arch B Cell Pathol Incl Mol Pathol1993;64:293. CrossRef

72. WeckstromP, HedrumA, MakridisC, et al.Midgut carcinoids and solid Carcinomas of the intestine: differences in endocrine markers and p53 mutations. Endocr Pathol1996;7:273. CrossRef

73. MooreP, OrlandiniS, ZamboniG, et al.Pancreatic tumours: molecular pathways implicated in ductal cancer are involved in ampullary but not in exocrine nonductal or endocrine tumorigenesis. Br J Cancer2001;84:253. CrossRef

74. HuW, FengZ, ModicaI, et al.Gene amplifications in well‐differentiated pancreatic neuroendocrine tumors inactivate the p53 pathway. Genes Cancer2010;1:360. CrossRef

75. YachidaS, VakianiE, WhiteC, 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 Pathol2012;36:173. CrossRef

76. TannapfelA, VomschlossS, KarhoffD, et al.BRAF gene mutations are rare events in gastroenteropancreatic neuroendocrine tumors. Am J Clin Pathol2005;123:256. CrossRef

77. DammannR, SchagdarsurenginU, LiuL, et al.Frequent RASSF1A promoter hypermethylation and K‐ras mutations in pancreatic carcinoma. Oncogene2003;22:3806. CrossRef

78. HervieuV, LepinasseF, GouysseG, et al.Expression of beta‐catenin in gastroenteropancreatic endocrine tumours: a study of 229 cases. J Clin Pathol2006;59:1300. CrossRef

79. PerrenA, SaremaslaniP, SchmidS, et al.DPC4/Smad4: no mutations, rare allelic imbalances, and retained protein expression in pancreatic endocrine tumors. Diagn Mol Pathol2003;12:181. CrossRef

80. BartschD, HahnS, DanichevskiK, et al.Mutations of the DPC4/Smad4 gene in neuroendocrine pancreatic tumors. Oncogene1999;18:2367. CrossRef

81. TangLH, ContractorT, ClausenR, et al.Attenuation of the retinoblastoma pathway in pancreatic neuroendocrine tumors due to increased cdk4/cdk6. Clin Cancer Res2012;18:4612. CrossRef

82. GuoS, WuX, ShimoideA, et al.Frequent overexpression of cyclin D1 in sporadic pancreatic endocrine tumours. J Endocrinol2003;179:73. CrossRef

83. ChungD, BrownS, Graeme‐CookF, et al.Overexpression of cyclin D1 occurs frequently in human pancreatic endocrine tumors. J Clin Endocrinol Metab2000;85:4373.

84. SerranoJ, GoebelS, PeghiniP, et al.Alterations in the p16INK4a/CDKN2A tumor suppressor gene in gastrinomas. J Clin Endocrinol Metab2000;85:4146. CrossRef

85. WangD, JohnstonC, BuchananK. Oncogene expression in gastroenteropancreatic neuroendocrine tumors: implications for pathogenesis. Cancer1997;80:668. CrossRef

86. CapursoG, LattimoreS, Crnogorac‐JurcevicT, et al.Gene expression profiles of progressive pancreatic endocrine tumours and their liver metastases reveal potential novel markers and therapeutic targets. Endocr Relat Cancer2006;13:541. CrossRef

87. PavelicK, HrascanR, KapitanovicS, et al.Molecular genetics of malignant insulinoma. Anticancer Res1996;16:1707.

88. BollmannF. Targeting ALT: the role of alternative lengthening of telomeres in pathogenesis and prevention of cancer. Cancer Treat Rev2007;33:704. CrossRef

89. HeaphyC, de WildeR, JiaoY, et al.Altered telomeres in tumors with ATRX and DAXX mutations. Science (New York, NY)2011;333:425. CrossRef

90. MarinoniI, KurrerA, VassellaE, et al.Loss of DAXX and ATRX Are Associated with chromosome instability and reduced survival of patients with pancreatic neuroendocrine tumors. Gastroenterology2013;146:453. CrossRef

91. SrivastavaA, AlexanderJ, LomakinI, et al.Immunohistochemical expression of transforming growth factor alpha and epidermal growth factor receptor in pancreatic endocrine tumors. Hum Pathol2001;32:1184. CrossRef

92. PeghiniP, IwamotoM, RaffeldM, et al.Overexpression of epidermal growth factor and hepatocyte growth factor receptors in a proportion of gastrinomas correlates with aggressive growth and lower curability. Clin Cancer Res2002;8:2273.

93. FjällskogM‐LH, LejonklouM, ObergK, et al.Expression of molecular targets for tyrosine kinase receptor antagonists in malignant endocrine pancreatic tumors. Clin Cancer Res2003;9:1469.

94. GoebelS, IwamotoM, RaffeldM, et al.Her‐2/neu expression and gene amplification in gastrinomas: correlations with tumor biology, growth, and aggressiveness. Cancer Res2002;62:3702.

95. FurukawaM, RaffeldM, MateoC, et al.Increased expression of insulin‐like growth factor I and/or its receptor in gastrinomas is associated with low curability, increased growth, and development of metastases. Clin Cancer Res2005;11:3233. CrossRef

96. ZhangL, SmyrkT, OliveiraA, et al.KIT is an independent prognostic marker for pancreatic endocrine tumors: a finding derived from analysis of islet cell differentiation markers. Am J Surg Pathol2009;33:1562. CrossRef

97. Rubbia‐BrandtL, TerrisB, GiostraE, et al.Lymphatic vessel density and vascular endothelial growth factor‐C expression correlate with malignant behavior in human pancreatic endocrine tumors. Clin Cancer Res2004;10:6919. CrossRef

98. ZhangJ, JiaZ, LiQ, et al.Elevated expression of vascular endothelial growth factor correlates with increased angiogenesis and decreased progression‐free survival among patients with low‐grade neuroendocrine tumors. Cancer2007;109:1478. CrossRef

99. CouvelardA, O'TooleD, TurleyH, et al.Microvascular density and hypoxia‐inducible factor pathway in pancreatic endocrine tumours: negative correlation of microvascular density and VEGF expression with tumour progression. Br J Cancer2005;92:94. CrossRef

100. MaitraA, HanselD, ArganiP, et al.Global expression analysis of well‐differentiated pancreatic endocrine neoplasms using oligonucleotide microarrays. Clin Cancer Res2003;9:5988.

101. BloomstonM, DurkinA, YangI, et al.Identification of molecular markers specific for pancreatic neuroendocrine tumors by genetic profiling of core biopsies. Ann Surg Oncol2004;11:413. CrossRef

102. HanselD, RahmanA, HouseM, et al.Met proto‐oncogene and insulin‐like growth factor binding protein 3 overexpression correlates with metastatic ability in well‐differentiated pancreatic endocrine neoplasms. Clin Cancer Res2004;10:6152. CrossRef

103. CouvelardA, HuJ, SteersG, et al.Identification of potential therapeutic targets by gene‐expression profiling in pancreatic endocrine tumors. Gastroenterology2006;131:1597. CrossRef

104. DuerrE‐M, MizukamiY, NgA, et al.Defining molecular classifications and targets in gastroenteropancreatic neuroendocrine tumors through DNA microarray analysis. Endocr Relat Cancer2008;15:243. CrossRef

105. EdfeldtK, BjörklundP, ÅkerströmG, et al.Different gene expression profiles in metastasizing midgut carcinoid tumors. Endocr Relat Cancer2011;18:479. CrossRef

106. LejaJ, EssaghirA, EssandM, et al.Novel markers for enterochromaffin cells and gastrointestinal neuroendocrine carcinomas. Mod Pathol2009;22:261. CrossRef

107. RoldoC, MissiagliaE, HaganJ, et al.MicroRNA expression abnormalities in pancreatic endocrine and acinar tumors are associated with distinctive pathologic features and clinical behavior. J Clin Oncol2006;24:4677. CrossRef

108. RuebelK, LeontovichA, StillingG, et al.MicroRNA expression in ileal carcinoid tumors: downregulation of microRNA‐133a with tumor progression. Mod Pathol2010;23:367. CrossRef

109. HamiltonSR, AaltonenLA. Pathology and Genetics of Tumours of the Digestive System. Lyon: IARC Press, 2000.

110. FerroneCR, TangLH, TomlinsonJ, et al.Determining prognosis in patients with pancreatic endocrine neoplasms: can the WHO classification system be simplified?J Clin Oncol2007;25:5609. CrossRef

111. KlimstraDS, ModlinIR, CoppolaD, et al.The pathologic classification of neuroendocrine tumors: a review of nomenclature, grading, and staging systems. Pancreas2010;39:707. CrossRef

112. RindiG, KlöppelG, AlhmanH, et al.TNM staging of foregut (neuro)endocrine tumors: a consensus proposal including a grading system. Virchows Arch2006;449:395. CrossRef

113. RindiG, KlöppelG, CouvelardA, et al.TNM staging of midgut and hindgut (neuro) endocrine tumors: a consensus proposal including a grading system. Virchows Arch2007;451:757. CrossRef

114. KlöppelG, RindiG, PerrenA, et al.The ENETS and AJCC/UICC TNM classifications of the neuroendocrine tumors of the gastrointestinal tract and the pancreas: a statement. Virchows Arch2010;456:595. CrossRef

115. StrosbergJR, CoppolaD, KlimstraDS, et al.The NANETS consensus guidelines for the diagnosis and management of poorly differentiated (high‐grade) extrapulmonary neuroendocrine carcinomas. Pancreas2010;39:799. CrossRef

116. VinikAI, WolteringEA, WarnerRR, et al.NANETS consensus guidelines for the diagnosis of neuroendocrine tumor. Pancreas2010;39:713. CrossRef

117. AnthonyLB, StrosbergJR, KlimstraDS, et al.The NANETS consensus guidelines for the diagnosis and management of gastrointestinal neuroendocrine tumors (nets): well‐differentiated nets of the distal colon and rectum. Pancreas2010;39:767. CrossRef

118. KulkeMH, AnthonyLB, BushnellDL, et al.NANETS treatment guidelines: well‐differentiated neuroendocrine tumors of the stomach and pancreas. Pancreas2010;39:735. CrossRef

119. KvolsLK, BrendtroKL. North American Neuroendocrine Tumor S. The North American Neuroendocrine Tumor Society (NANETS) guidelines: mission, goals, and process. Pancreas2010;39:705. CrossRef

120. SalazarR, WiedenmannB, RindiG, et al.ENETS 2011 consensus guidelines for the management of patients with digestive neuroendocrine tumors: an update. Neuroendocrinology2011;95:71. CrossRef

121. BosmanFT, CarneiroF, HrubanRH, et al.WHO Classification of Tumours of the Digestive System, 4th edn. Lyon: World Health Organization, 2010.

122. KlimstraD, ModlinI, CoppolaD, et al.The pathologic classification of neuroendocrine tumors: a review of nomenclature, grading, and staging systems. Pancreas2010;39:707. CrossRef

123. EdgeS, ByrdDR, ComptonCC, et al.AJCC Cancer Staging Manual. New York: Springer, 2010.

124. CapelliP, FassanM, ScarpaA. Pathology – grading and staging of GEP‐NETs. Best Pract Res Clin Gastroenterol2012;26:705. CrossRef

125. CouvelardA, DeschampsL, RavaudP, et al.Heterogeneity of tumor prognostic markers: a reproducibility study applied to liver metastases of pancreatic endocrine tumors. Mod Pathol2009;22:273. CrossRef

126. LindholmEB, LyonsJ, AnthonyCT, et al.Do primary neuroendocrine tumors and metastasis have the same characteristics?J Surg Res2012;174:200. CrossRef

127. ScarpaA, MantovaniW, CapelliP, et al.Pancreatic endocrine tumors: improved TNM staging and histopathological grading permit a clinically efficient prognostic stratification of patients. Mod Pathol2010;23:824. CrossRef

128. ButturiniG, BettiniR, MissiagliaE, et al.Predictive factors of efficacy of the somatostatin analogue octreotide as first line therapy for advanced pancreatic endocrine carcinoma. Endocr Relat Cancer2006;13:1213. CrossRef

129. PanzutoF, BoninsegnaL, FazioN, et al.Metastatic and locally advanced pancreatic endocrine carcinomas: analysis of factors associated with disease progression. J Clin Oncol2011;29:2372. CrossRef

130. BoninsegnaL, PanzutoF, PartelliS, et al.Malignant pancreatic neuroendocrine tumour: lymph node ratio and Ki67 are predictors of recurrence after curative resections. Eur J Cancer2012;48:1608. CrossRef

131. RindiG, FalconiM, KlersyC, et al.TNM staging of neoplasms of the endocrine pancreas: results from a large international cohort study. J Natl Cancer Inst2012;104:764. CrossRef

132. QianZR, Ter‐MinassianM, ChanJA, et al.Prognostic significance of MTOR pathway component expression in neuroendocrine tumors. J Clin Oncol2013;31:3418. CrossRef

133. La RosaS, KlersyC, UccellaS, et al.Improved histologic and clinicopathologic criteria for prognostic evaluation of pancreatic endocrine tumors. Hum Pathol2008;40:30. CrossRef

134. PapeU‐F, JannH, Müller‐NordhornJ, et al.Prognostic relevance of a novel TNM classification system for upper gastroenteropancreatic neuroendocrine tumors. Cancer2008;113:256. CrossRef

135. EkebladS, SkogseidB, DunderK, et al.Prognostic factors and survival in 324 patients with pancreatic endocrine tumor treated at a single institution. Clin Cancer Res2008;14:7798. CrossRef

136. ErikssonB, ArnbergH, LindgrenP, et al.Neuroendocrine pancreatic tumours: clinical presentation, biochemical and histopathological findings in 84 patients. J Intern Med1990;228:103. CrossRef

137. Toth‐FejelS, PommierR. Relationships among delay of diagnosis, extent of disease, and survival in patients with abdominal carcinoid tumors. Am J Surg2004;187:575. CrossRef

138. MaglinteD, O'ConnorK, BessetteJ, et al.The role of the physician in the late diagnosis of primary malignant tumors of the small intestine. Am J Gastroenterol1991;86:304.

139. MatonP, GardnerJ, JensenR. Cushing's syndrome in patients with the Zollinger‐Ellison syndrome. N Engl J Med1986;315:1. CrossRef

140. WynickD, WilliamsS, BloomS. Symptomatic secondary hormone syndromes in patients with established malignant pancreatic endocrine tumors. N Engl J Med1988;319:605. CrossRef

141. YuF, VenzonD, SerranoJ, et al.Prospective study of the clinical course, prognostic factors, causes of death, and survival in patients with long‐standing Zollinger‐Ellison syndrome. J Clin Oncol1999;17:615.

142. CorletoV, PanzutoF, FalconiM, et al.Digestive neuroendocrine tumours: diagnosis and treatment in Italy. A survey by the oncology study section of the Italian society of gastroenterology (SIGE). Dig Liver Dis2001;33:217. CrossRef

143. GuoK‐J, LiaoH‐H, TianY‐L, et al.Surgical treatment of nonfunctioning islet cell tumor: report of 41 cases. Hepatobiliary Pancreat Dis Int2004;3:469.

144. ChuQ, HillH, DouglassH, et al.Predictive factors associated with long‐term survival in patients with neuroendocrine tumors of the pancreas. Ann Surg Oncol2002;9:855. CrossRef

145. ItoT, TanakaM, SasanoH, et al.Preliminary results of a Japanese nationwide survey of neuroendocrine gastrointestinal tumors. J Gastroenterol2007;42:497. CrossRef

146. PanzutoF, NasoniS, FalconiM, et al.Prognostic factors and survival in endocrine tumor patients: comparison between gastrointestinal and pancreatic localization. Endocr Relat Cancer2005;12:1083. CrossRef

147. ChiangH, O'DorisioT, HuangS, et al.Multiple hormone elevations in Zollinger‐Ellison syndrome. Prospective study of clinical significance and of the development of a second symptomatic pancreatic endocrine tumor syndrome. Gastroenterology1990;99:1565.

148. EkebladS, LejonklouM, GrimfjärdP, et al.Co‐expression of ghrelin and its receptor in pancreatic endocrine tumours. Clin Endocrinol (Oxf)2007;66:115.

149. BordiC, AzzoniC, D'AddaT, et al.Pancreatic polypeptide‐related tumors. Peptides2002;23:339. CrossRef

150. FleuryA, FléjouJ, SauvanetA, et al.Calcitonin‐secreting tumors of the pancreas: about six cases. Pancreas1998;16:545. CrossRef

151. SchneiderR, WaldmannJ, SwaidZ, et al.Calcitonin‐secreting pancreatic endocrine tumors: systematic analysis of a rare tumor entity. Pancreas2011;40:213. CrossRef

152. Bani‐SacchiT, BartoliniG, BiliottiG. A multihormonal tumor of the pancreas producing neurotensin associated with the WDHA syndrome. Histology, histochemistry and origin. Histol Histopathol1986;1:187.

153. GutniakM, RosenqvistU, GrimeliusL, et al.Report on a patient with watery diarrhoea syndrome caused by a pancreatic tumour containing neurotensin, enkephalin and calcitonin. Acta Med Scand1980;208:95. CrossRef

154. AmrillevaV, SlaterE, WaldmannJ, et al.A pancreatic polypeptide‐producing pancreatic tumor causing WDHA syndrome. Case Rep Gastroenterol2008;2:238. CrossRef

155. LundqvistG, KrauseU, LarssonL, et al.A pancreatic‐polypeptide‐producing tumour associated with the WDHA syndrome. Scand J Gastroenterol1978;13:715. CrossRef

156. WangH, OhD, OhningG, et al.Elevated serum ghrelin exerts an orexigenic effect that may maintain body mass index in patients with metastatic neuroendocrine tumors. J Mol Neurosci2007;33:225. CrossRef

157. WalterT, ChardonL, HervieuV, et al.Major hyperghrelinemia in advanced well‐differentiated neuroendocrine carcinomas: report of three cases. Eur J Endocrinol2009;161:639. CrossRef

158. GulloL, MiglioriM, FalconiM, et al.Nonfunctioning pancreatic endocrine tumors: a multicenter clinical study. Am J Gastroenterol2003;98:2435. CrossRef

159. FalconiM, PlockingerU, KwekkeboomD, et al.Well‐differentiated pancreatic nonfunctioning tumors/carcinoma. Neuroendocrinology2006;84:196. CrossRef

160. Cheslyn‐CurtisS, SitaramV, WilliamsonR. Management of non‐functioning neuroendocrine tumours of the pancreas. Br J Surg1993;80:625. CrossRef

161. Thomas‐MarquesL, MuratA, DelemerB, et al.Prospective endoscopic ultrasonographic evaluation of the frequency of nonfunctioning pancreaticoduodenal endocrine tumors in patients with multiple endocrine neoplasia type 1. Am J Gastroenterol2006;101:266. CrossRef

162. WamstekerE‐J, GaugerP, ThompsonN, et al.EUS detection of pancreatic endocrine tumors in asymptomatic patients with type 1 multiple endocrine neoplasia. Gastrointest Endosc2003;58:531. CrossRef

163. de WildeRF, EdilBH, HrubanRH, et al.Well‐differentiated pancreatic neuroendocrine tumors: from genetics to therapy. Nat Rev Gastroenterol Hepatol2012;9:199. CrossRef

164. GrantC. Insulinoma. Best Pract Res Clin Gastroenterol2005;19:783. CrossRef

165. StefaniniP, CarboniM, PatrassiN, et al.Beta‐islet cell tumors of the pancreas: results of a study on 1,067 cases. Surgery1974;75:597.

166. ServiceF, DaleA, ElvebackL, et al.Insulinoma: clinical and diagnostic features of 60 consecutive cases. Mayo Clin Proc1976;51:417.

167. DohertyG, DoppmanJ, ShawkerT, et al.Results of a prospective strategy to diagnose, localize, and resect insulinomas. Surgery1991;110:989.

168. NikfarjamM, WarshawA, AxelrodL, et al.Improved contemporary surgical management of insulinomas: a 25‐year experience at the Massachusetts General Hospital. Ann Surg2008;247:165. CrossRef

169. ChristiansenL, NielsenO, StadilF, et al.Delay in the diagnosis of insulinomas. Scand J Gastroenterol Suppl1979;53:43.

170. JensenR, BernaM, BinghamD, et al.Inherited pancreatic endocrine tumor syndromes: advances in molecular pathogenesis, diagnosis, management, and controversies. Cancer2008;113(7 Suppl):1807. CrossRef

171. VernerJ, MorrisonA. Islet cell tumor and a syndrome of refractory watery diarrhea and hypokalemia. Am J Med1958;25:374. CrossRef

172. CapellaC, PolakJ, BuffaR, et al.Morphologic patterns and diagnostic criteria of VIP‐producing endocrine tumors. A histologic, histochemical, ultrastructural, and biochemical study of 32 cases. Cancer1983;52:1860. CrossRef

173. SogaJ, YakuwaY. Vipoma/diarrheogenic syndrome: a statistical evaluation of 241 reported cases. J Exp Clin Cancer Res1998;17:389.

174. LongR, BryantM, MitchellS, et al.Clinicopathological study of pancreatic and ganglioneuroblastoma tumours secreting vasoactive intestinal polypeptide (vipomas). Br Med J (Clin Res Ed)1981;282:1767. CrossRef

175. NikouG, ToubanakisC, NikolaouP, et al.VIPomas: an update in diagnosis and management in a series of 11 patients. Hepatogastroenterology2005;52:1259.

176. GhaferiA, ChojnackiK, LongW, et al.Pancreatic VIPomas: subject review and one institutional experience. J Gastrointest Surg2008;12:382. CrossRef

177. SmithS, BrantonS, AvinoA, et al.Vasoactive intestinal polypeptide secreting islet cell tumors: a 15‐year experience and review of the literature. Surgery1998;124:1050. CrossRef

178. PengS, LiJ, LiuY, et al.Diagnosis and treatment of VIPoma in China: (case report and 31 cases review) diagnosis and treatment of VIPoma. Pancreas2004;28:93. CrossRef

179. PorterJ, ReidI, SaidS, et al.Stimulation of renin secretion by vasoactive intestinal peptide. Am J Physiol1982;243:10.

180. FelíuJ, MojenaM, SilvestreR, et al.Stimulatory effect of vasoactive intestinal peptide on glycogenolysis and gluconeogenesis in isolated rat hepatocytes: antagonism by insulin. Endocrinology1983;112:2120. CrossRef

181. KerinsC, SaidS. Hyperglycemic and glycogenolytic effects of vasoactive intestinal polypeptide. Proc Soc Exp Biol Med1973;142:1014. CrossRef

182. MakhloufG, ZfassA, SaidS, et al.Effects of synthetic vasoactive intestinal peptide (VIP), secretin and their partial sequences on gastric secretion. Proc Soc Exp Biol Med1978;157:565. CrossRef

183. RoostermanD, GoergeT, SchneiderS, et al.Neuronal control of skin function: the skin as a neuroimmunoendocrine organ. Physiol Rev2006;86:1309. CrossRef

184. KindmarkH, SundinA, GranbergD, et al.Endocrine pancreatic tumors with glucagon hypersecretion: a retrospective study of 23 cases during 20 years. Med Oncol2007;24:330. CrossRef

185. SogaJ, YakuwaY. Glucagonomas/diabetico‐dermatogenic syndrome (DDS): a statistical evaluation of 407 reported cases. J Hepatobiliary Pancreat Surg1998;5:312. CrossRef

186. LeichterS. Clinical and metabolic aspects of glucagonoma. Medicine1980;59:100. CrossRef

187. WermersR, FatourechiV, WynneA, et al.The glucagonoma syndrome. Clinical and pathologic features in 21 patients. Medicine1996;75:53. CrossRef

188. DruckerD. Biologic actions and therapeutic potential of the proglucagon‐derived peptides. Nat Clin Pract Endocrinol Metab2005;1:22. CrossRef

189. ChastainM. The glucagonoma syndrome: a review of its features and discussion of new perspectives. Am J Med Sci2001;321:306. CrossRef

190. van BeekA, de HaasE, van VlotenW, et al.The glucagonoma syndrome and necrolytic migratory erythema: a clinical review. Eur J Endocrinol2004;151:531. CrossRef

191. PujolR, WangC‐YE, el‐AzharyR, et al.Necrolytic migratory erythema: clinicopathologic study of 13 cases. Int J Dermatol2004;43:12. CrossRef

192. WilkinsonD. Necrolytic migratory erythema with carcinoma of the pancreas. Trans St Johns Hosp Dermatol Soc1973;59:244.

193. EldorR, GlaserB, FraenkelM, et al.Glucagonoma and the glucagonoma syndrome – cumulative experience with an elusive endocrine tumour. Clin Endocrinol (Oxf)2011;74:593. CrossRef

194. GarbrechtN, AnlaufM, SchmittA, et al.Somatostatin‐producing neuroendocrine tumors of the duodenum and pancreas: incidence, types, biological behavior, association with inherited syndromes, and functional activity. Endocr Relat Cancer2008;15:229. CrossRef

195. TanakaS, YamasakiS, MatsushitaH, et al.Duodenal somatostatinoma: a case report and review of 31 cases with special reference to the relationship between tumor size and metastasis. Pathol Int2000;50:146. CrossRef

196. SogaJ, YakuwaY. Somatostatinoma/inhibitory syndrome: a statistical evaluation of 173 reported cases as compared to other pancreatic endocrinomas. J Exp Clin Cancer Res1999;18:13.

197. JensenR, RindiG, ArnoldR, et al.Well‐differentiated duodenal tumor/carcinoma (excluding gastrinomas). Neuroendocrinology2006;84:165. CrossRef

198. LuceyM, YamadaT. Biochemistry and physiology of gastrointestinal somatostatin. Dig Dis Sci1989;34(3 Suppl):S5. CrossRef

199. MoayedoddinB, BooyaF, WermersR, et al.Spectrum of malignant somatostatin‐producing neuroendocrine tumors. Endocr Pract2006;12:394. CrossRef

200. KrejsG, OrciL, ConlonJ, et al.Somatostatinoma syndrome. Biochemical, morphologic and clinical features. N Engl J Med1979;301:285. CrossRef

201. NesiG, MarcucciT, RubioC, et al.Somatostatinoma: clinico‐pathological features of three cases and literature reviewed. J Gastroenterol Hepatol2008;23:521. CrossRef

202. HoffmannK, FurukawaM, JensenR. Duodenal neuroendocrine tumors: classification, functional syndromes, diagnosis and medical treatment. Best Pract Res Clin Gastroenterol2005;19:675. CrossRef

203. O'TooleD, Delle FaveG, JensenR. Gastric and duodenal neuroendocrine tumours. Best Pract Res Clin Gastroenterol2012;26:719. CrossRef

204. SogaJ. Carcinoids of the pancreas: an analysis of 156 cases. Cancer2005;104:1180. CrossRef

205. ModlinI, LyeK, KiddM. A 5‐decade analysis of 13,715 carcinoid tumors. Cancer2003;97:934. CrossRef

206. ModlinI, KiddM, LatichI, et al.Current status of gastrointestinal carcinoids. Gastroenterology2005;128:1717. CrossRef

207. ModlinI, ChampaneriaM, ChanA, et al.A three‐decade analysis of 3,911 small intestinal neuroendocrine tumors: the rapid pace of no progress. Am J Gastroenterol2007;102:1464. CrossRef

208. MorganJ, MarksC, HearnD. Carcinoid tumors of the gastrointestinal tract. Ann Surg1974;180:720. CrossRef

209. HellmanP, LundströmT, OhrvallU, et al.Effect of surgery on the outcome of midgut carcinoid disease with lymph node and liver metastases. World J Surg2002;26:991. CrossRef

210. BergeT, LinellF. Carcinoid tumours. Frequency in a defined population during a 12‐year period. Acta Pathol Microbiol Scand A1976;84:322.

211. MarshallJ, BodnarchukG. Carcinoid tumors of the gut. Our experience over three decades and review of the literature. J Clin Gastroenterol1993;16:123. CrossRef

212. OlneyJ, UrdanetaL, Al‐JurfA, et al.Carcinoid tumors of the gastrointestinal tract. Am Surg1985;51:37.

213. SahaS, HodaS, GodfreyR, et al.Carcinoid tumors of the gastrointestinal tract: a 44‐year experience. South Med J1989;82:1501. CrossRef

214. KemaI, de VriesE, SlooffM, et al.Serotonin, catecholamines, histamine, and their metabolites in urine, platelets, and tumor tissue of patients with carcinoid tumors. Clin Chem1994;40:86.

215. DavisZ, MoertelC, McIlrathD. The malignant carcinoid syndrome. Surg Gynecol Obstet1973;137:637.

216. DonowitzM, BinderH. Jejunal fluid and electrolyte secretion in carcinoid syndrome. Am J Dig Dis1975;20:1115. CrossRef

217. von der OheM, CamilleriM, KvolsL, et al.Motor dysfunction of the small bowel and colon in patients with the carcinoid syndrome and diarrhea. N Engl J Med1993;329:1073. CrossRef

218. ShahG, ShahR, VeilletteH, et al.Biochemical assessment of niacin deficiency among carcinoid cancer patients. Am J Gastroenterol2005;100:2307. CrossRef

219. BellH, PostonG, VoraJ, et al.Cutaneous manifestations of the malignant carcinoid syndrome. Br J Dermatol2005;152:71. CrossRef

220. LundinL, NorheimI, LandeliusJ, et al.Carcinoid heart disease: relationship of circulating vasoactive substances to ultrasound‐detectable cardiac abnormalities. Circulation1988;77:264. CrossRef

221. MøllerJ, PellikkaP, BernheimA, et al.Prognosis of carcinoid heart disease: analysis of 200 cases over two decades. Circulation2005;112:3320. CrossRef

222. SchweizerW, GloorF, Von BertrabR, et al.Carcinoid heart disease with left‐sided lesions. Circulation1964;29:253. CrossRef

223. RoyA, BrandN, YacoubM. Expression of 5‐hydroxytryptamine receptor subtype messenger RNA in interstitial cells from human heart valves. J Heart Valve Dis2000;9:256.

224. WaltenbergerJ, LundinL, ObergK, et al.Involvement of transforming growth factor‐beta in the formation of fibrotic lesions in carcinoid heart disease. Am J Pathol1993;142:71.

225. JianB, XuJ, ConnollyJ, et al.Serotonin mechanisms in heart valve disease I: serotonin‐induced up‐regulation of transforming growth factor‐beta1 via G‐protein signal transduction in aortic valve interstitial cells. Am J Pathol2002;161:2111. CrossRef

226. ConnollyH, CraryJ, McGoonM, et al.Valvular heart disease associated with fenfluramine‐phentermine. N Engl J Med1997;337:581. CrossRef

227. DruceM, RockallA, GrossmanA. Fibrosis and carcinoid syndrome: from causation to future therapy. Nat Rev Endocrinol2009;5:276. CrossRef

228. ModlinI, ShapiroM, KiddM. Carcinoid tumors and fibrosis: an association with no explanation. Am J Gastroenterol2004;99:2466. CrossRef

229. KahilM, BrownH, FredH. The carcinoid crisis. Arch Intern Med1964;114:26. CrossRef

230. YantissR, OdzeR, FarrayeF, et al.Solitary versus multiple carcinoid tumors of the ileum: a clinical and pathologic review of 68 cases. Am J Surg Pathol2003;27:811. CrossRef

231. GarbyL, CaronP, ClaustratF, et al.Clinical characteristics and outcome of acromegaly induced by ectopic secretion of growth hormone‐releasing hormone (GHRH): a French nationwide series of 21 cases. J Clin Endocrinol Metab2012;97:2093. CrossRef

232. MiehleK, TannapfelA, LameschP, et al.Pancreatic neuroendocrine tumor with ectopic adrenocorticotropin production upon second recurrence. J Clin Endocrinol Metab2004;89:3731. CrossRef

233. SalvatoriR, FintiniD, WestraW, et al.Cushing's Syndrome attributable to ectopic secretion of corticotropin in a patient with two neuroendocrine tumors. Endocr Pract2006;12:656. CrossRef

234. DoiM, SugiyamaT, IzumiyamaH, et al.Clinical features and management of ectopic ACTH syndrome at a single institute in Japan. Endocr J2010;57:1061. CrossRef

235. IliasI, TorpyD, PacakK, et al.Cushing's syndrome due to ectopic corticotropin secretion: twenty years' experience at the National Institutes of Health. J Clin Endocrinol Metab2005;90:4955. CrossRef

236. MüssigK, PetersennS, WehrmannM, et al.Somatostatin receptor expression in a parathyroid hormone‐related peptide‐secreting pancreatic neuroendocrine tumour causing severe hypercalcaemia. Eur J Gastroenterol Hepatol2007;19:719. CrossRef

237. SrirajaskanthanR, McStayM, ToumpanakisC, et al.Parathyroid hormone‐related peptide‐secreting pancreatic neuroendocrine tumours: case series and literature review. Neuroendocrinology2009;89:48. CrossRef

238. LangerP, BartschD, GerdesB, et al.Renin producing neuroendocrine pancreatic carcinoma–a case report and review of the literature. Exp Clin Endocrinol Diabetes2002;110:43. CrossRef

239. RuddyM, AtlasS, SalernoF. Hypertension associated with a renin‐secreting adenocarcinoma of the pancreas. N Engl J Med1982;307:993. CrossRef

240. SamynI, FontaineC, Van TussenbroekF, et al.Paraneoplastic syndromes in cancer: case 1. Polycythemia as a result of ectopic erythropoietin production in metastatic pancreatic carcinoid tumor. J Clin Oncol2004;22:2240. CrossRef

241. PiaditisG, AngellouA, KontogeorgosG, et al.Ectopic bioactive luteinizing hormone secretion by a pancreatic endocrine tumor, manifested as luteinized granulosa‐thecal cell tumor of the ovaries. J Clin Endocrinol Metab2005;90:2097. CrossRef

242. BrignardelloE, MantiR, PapottiM, et al.Ectopic secretion of LH by an endocrine pancreatic tumor. J Endocrinol Invest2004;27:361. CrossRef

243. RobertsR, ZhaoM, WhitelawB, et al.GLP‐1 and glucagon secretion from a pancreatic neuroendocrine tumor causing diabetes and hyperinsulinemic hypoglycemia. J Clin Endocrinol Metab2012;97:3039. CrossRef

244. ChungJ, HongS, ChoD, et al.Hypoglycemia associated with the production of insulin‐like growth factor II in a pancreatic islet cell tumor: a case report. Endocr J2008;55:607. CrossRef

245. BasturkO, TangL, HrubanR, et al.Poorly differentiated neuroendocrine carcinomas of the pancreas: a clinicopathologic analysis of 44 cases. Am J Surg Pathol2014;38:437. CrossRef

246. BrennerB, ShahM, GonenM, et al.Small‐cell carcinoma of the gastrointestinal tract: a retrospective study of 64 cases. Br J Cancer2004;90:1720.

247. BassettJ, ForbesS, PannettA, et al.Characterization of mutations in patients with multiple endocrine neoplasia type 1. Am J Hum Genet1998;62:232. CrossRef

248. GibrilF, SchumannM, PaceA, et al.Multiple endocrine neoplasia type 1 and Zollinger‐Ellison syndrome: a prospective study of 107 cases and comparison with 1009 cases from the literature. Medicine2004;83:43. CrossRef

249. ThakkerR, NeweyP, WallsG, et al.Clinical practice guidelines for multiple endocrine neoplasia type 1 (MEN1). J Clin Endocrinol Metab2012;97:2990. CrossRef

250. NortonJ, CorneliusM, DoppmanJ, et al.Effect of parathyroidectomy in patients with hyperparathyroidism, Zollinger‐Ellison syndrome, and multiple endocrine neoplasia type I: a prospective study. Surgery1987;102:958.

251. MetzD, JensenR. Gastrointestinal neuroendocrine tumors: pancreatic endocrine tumors. Gastroenterology2008;135:1469. CrossRef

252. AnlaufM, GarbrechtN, HenoppT, et al.Sporadic versus hereditary gastrinomas of the duodenum and pancreas: distinct clinico‐pathological and epidemiological features. World J Gastroenterol2006;12:5440.

253. ThompsonN, LloydR, NishiyamaR, et al.MEN I pancreas: a histological and immunohistochemical study. World J Surg1984;8:561. CrossRef

254. AsgharianB, TurnerM, GibrilF, et al.Cutaneous tumors in patients with multiple endocrine neoplasm type 1 (MEN1) and gastrinomas: prospective study of frequency and development of criteria with high sensitivity and specificity for MEN1. J Clin Endocrinol Metab2004;89:5328. CrossRef

255. AsgharianB, ChenY‐J, PatronasN, et al.Meningiomas may be a component tumor of multiple endocrine neoplasia type 1. Clin Cancer Res2004;10:869. CrossRef

256. McKeebyJ, LiX, ZhuangZ, et al.Multiple leiomyomas of the esophagus, lung, and uterus in multiple endocrine neoplasia type 1. Am J Pathol2001;159:1121. CrossRef

257. GibrilF, ChenY‐J, SchrumpD, et al.Prospective study of thymic carcinoids in patients with multiple endocrine neoplasia type 1. J Clin Endocrinol Metab2003;88:1066. CrossRef

258. MukhopadhyayB, SahdevA, MonsonJ, et al.Pancreatic lesions in von Hippel‐Lindau disease. Clin Endocrinol (Oxf)2002;57:603. CrossRef

259. HoughD, StephensD, JohnsonC, et al.Pancreatic lesions in von Hippel‐Lindau disease: prevalence, clinical significance, and CT findings. AJR Am J Roentgenol1994;162:1091. CrossRef

260. HammelP, VilgrainV, TerrisB, et al.Pancreatic involvement in von Hippel‐Lindau disease. The Groupe Francophone d'Etude de la Maladie de von Hippel‐Lindau. Gastroenterology2000;119:1087. CrossRef

261. LibuttiS, ChoykeP, AlexanderH, et al.Clinical and genetic analysis of patients with pancreatic neuroendocrine tumors associated with von Hippel‐Lindau disease. Surgery2000;128:1022. CrossRef

262. LodishM, StratakisC. Endocrine tumours in neurofibromatosis type 1, tuberous sclerosis and related syndromes. Best Pract Res Clin Endocrinol Metab2010;24:439. CrossRef

263. DayalY, TallbergK, NunnemacherG, et al.Duodenal carcinoids in patients with and without neurofibromatosis. A comparative study. Am J Surg Pathol1986;10:348. CrossRef

264. HoughD, ChanA, DavidsonH. Von Recklinghausen's disease associated with gastrointestinal carcinoid tumors. Cancer1983;51:2206. CrossRef

265. MaoC, ShahA, HansonD, et al.Von Recklinghausen's disease associated with duodenal somatostatinoma: contrast of duodenal versus pancreatic somatostatinomas. J Surg Oncol1995;59:67. CrossRef

266. SakorafasG, GiannopoulosG, ParasiA, et al.Large somatostatin‐producing endocrine carcinoma of the ampulla of vater in association with GIST in a patient with von Recklinghausen's disease. Case report and review of the literature. JOP2008;9:633.

267. CappelliC, AgostiB, BragaM, et al.Von Recklinghausen's neurofibromatosis associated with duodenal somatostatinoma. A case report and review of the literature. Minerva Endocrinol2004;29:19.

268. UsuiM, MatsudaS, SuzukiH, et al.Somatostatinoma of the papilla of Vater with multiple gastrointestinal stromal tumors in a patient with von Recklinghausen's disease. J Gastroenterol2002;37:947. CrossRef

269. YoshidaA, HatanakaS, OhiY, et al.von Recklinghausen's disease associated with somatostatin‐rich duodenal carcinoid (somatostatinoma), medullary thyroid carcinoma and diffuse adrenal medullary hyperplasia. Acta Pathol Jpn1991;41:847.

270. OhtsukiY, SonobeH, MizobuchiT, et al.Duodenal carcinoid (somatostatinoma) combined with von Recklinghausen's disease. A case report and review of the literature. Acta Pathol Jpn1989;39:141.

271. LeeW‐S, KohY‐S, KimJ‐C, et al.Zollinger‐Ellison syndrome associated with neurofibromatosis type 1: a case report. BMC Cancer2005;5:85. CrossRef

272. PerrenA, WiesliP, SchmidS, et al.Pancreatic endocrine tumors are a rare manifestation of the neurofibromatosis type 1 phenotype: molecular analysis of a malignant insulinoma in a NF‐1 patient. Am J Surg Pathol2006;30:1047. CrossRef

273. FujisawaT, OsugaT, MaedaM, et al.Malignant endocrine tumor of the pancreas associated with von Recklinghausen's disease. J Gastroenterol2002;37:59. CrossRef

274. DworakowskaD, GrossmanA. Are neuroendocrine tumours a feature of tuberous sclerosis? A systematic review. Endocr Relat Cancer2009;16:45. CrossRef

275. MerrittJ, DavisD, PittelkowM, et al.Extensive acrochordons and pancreatic islet‐cell tumors in tuberous sclerosis associated with TSC2 mutations. Am J Med Genet A2006;140:1669. CrossRef

276. VezzosiD, WalterT, LaplancheA, et al.Chromogranin A measurement in metastatic well‐differentiated gastroenteropancreatic neuroendocrine carcinoma: screening for false positives and a prospective follow‐up study. Int J Biol Markers2011;26:94. CrossRef

277. ShahT, SrirajaskanthanR, BhogalM, et al.Alpha‐fetoprotein and human chorionic gonadotrophin‐beta as prognostic markers in neuroendocrine tumour patients. Br J Cancer2008;99:72. CrossRef

278. JensenR, CadiotG, BrandiM, et al.ENETS Consensus Guidelines for the management of patients with digestive neuroendocrine neoplasms: functional pancreatic endocrine tumor syndromes. Neuroendocrinology2012;95:98. CrossRef

279. CryerP, AxelrodL, GrossmanA, et al.Evaluation and management of adult hypoglycemic disorders: an endocrine society clinical practice guideline. J Clin Endocrinol Metab2009;94:709. CrossRef

280. VezzosiD, BennetA, FauvelJ, et al.Insulin, C‐peptide and proinsulin for the biochemical diagnosis of hypoglycaemia related to endogenous hyperinsulinism. Eur J Endocrinol2007;157:75. CrossRef

281. LupsaB, ChongA, CochranE, et al.Autoimmune forms of hypoglycemia. Medicine2009;88:141. CrossRef

282. MullansE, CohenP. Iatrogenic necrolytic migratory erythema: a case report and review of nonglucagonoma‐associated necrolytic migratory erythema. J Am Acad Dermatol1998;38:866. CrossRef

283. TierneyE, BadgerJ. Etiology and pathogenesis of necrolytic migratory erythema: review of the literature. Medgenmed2004;6:4.

284. KukuS, JaspanJ, EmmanouelD, et al.Heterogeneity of plasma glucagon. Circulating components in normal subjects and patients with chronic renal failure. J Clin Invest1976;58:742. CrossRef

285. MarcoJ, DiegoJ, VillanuevaM, et al.Elevated plasma glucagon levels in cirrhosis of the liver. N Engl J Med1973;289:1107. CrossRef

286. SchillerL, RiveraL, SantangeloW, et al.Diagnostic value of fasting plasma peptide concentrations in patients with chronic diarrhea. Dig Dis Sci1994;39:2216. CrossRef

287. MeijerW, KemaI, VolmerM, et al.Discriminating capacity of indole markers in the diagnosis of carcinoid tumors. Clin Chem2000;46:1588.

288. O'TooleD, GrossmanA, GrossD, et al.ENETS consensus guidelines for the standards of care in neuroendocrine tumors: biochemical markers. Neuroendocrinology2009;90:194. CrossRef

289. StrosbergJ. Neuroendocrine tumours of the small intestine. Best Pract Res Clin Gastroenterol2012;26:755. CrossRef

290. KemaI, SchellingsA, MeiborgG, et al.Influence of a serotonin‐ and dopamine‐rich diet on platelet serotonin content and urinary excretion of biogenic amines and their metabolites. Clin Chem1992;38:1730.

291. MashigeF, MatsushimaY, KanazawaH, et al.Acidic catecholamine metabolites and 5‐hydroxyindoleacetic acid in urine: the influence of diet. Ann Clin Biochem1996;33:43. CrossRef

292. CleareA, KeatingJ, EalingJ, et al.A case of coeliac disease detected via raised 5‐hydroxytryptamine and 5‐hydroxyindoleacetic acid. Ann Clin Biochem1997;34:440. CrossRef

293. Abu‐QurshinR, NaschitzJ, ZuckermannE, et al.Crohn's disease associated with pellagra and increased excretion of 5‐hydroxyindolacetic acid. Am J Med Sci1997;313:111. CrossRef

294. ColemanN, FoleyS, DunlopS, et al.Abnormalities of serotonin metabolism and their relation to symptoms in untreated celiac disease. Clin Gastroenterol Hepatol2006;4:874. CrossRef

295. GolaM, DogaM, BonadonnaS, et al.Neuroendocrine tumors secreting growth hormone‐releasing hormone: pathophysiological and clinical aspects. Pituitary2006;9:221. CrossRef

296. MachensA, SchaafL, KargesW, et al.Age‐related penetrance of endocrine tumours in multiple endocrine neoplasia type 1 (MEN1): a multicentre study of 258 gene carriers. Clin Endocrinol (Oxf)2007;67:613.

297. NeweyP, JeyabalanJ, WallsG, et al.Asymptomatic children with multiple endocrine neoplasia type 1 mutations may harbor nonfunctioning pancreatic neuroendocrine tumors. J Clin Endocrinol Metab2009;94:3640. CrossRef

298. TriponezF, DossehD, GoudetP, et al.Epidemiology data on 108 MEN 1 patients from the GTE with isolated nonfunctioning tumors of the pancreas. Ann Surg2006;243:265. CrossRef

299. StarkD, MossA, GoldbergH, et al.CT of pancreatic islet cell tumors. Radiology1984;150:491. CrossRef

300. WankS, DoppmanJ, MillerD, et al.Prospective study of the ability of computed axial tomography to localize gastrinomas in patients with Zollinger‐Ellison syndrome. Gastroenterology1987;92:905.

301. FidlerJ, FletcherJ, ReadingC, et al.Preoperative detection of pancreatic insulinomas on multiphasic helical CT. AJR Am J Roentgenol2003;181:775. CrossRef

302. RockallA, ReznekR. Imaging of neuroendocrine tumours (CT/MR/US). Best Pract Res Clin Endocrinol Metab2007;21:43. CrossRef

303. KhashabM, YongE, LennonA, et al.EUS is still superior to multidetector computerized tomography for detection of pancreatic neuroendocrine tumors. Gastrointest Endosc2011;73:691. CrossRef

304. MatonP, MillerD, DoppmanJ, et al.Role of selective angiography in the management of patients with Zollinger‐Ellison syndrome. Gastroenterology1987;92:913.

305. IchikawaT, PetersonM, FederleM, et al.Islet cell tumor of the pancreas: biphasic CT versus MR imaging in tumor detection. Radiology2000;216:163. CrossRef

306. OwenN, SohaibS, PeppercornP, et al.MRI of pancreatic neuroendocrine tumours. Br J Radiol2001;74:968. CrossRef

307. SemelkaR, CustodioC, Cem BalciN, et al.Neuroendocrine tumors of the pancreas: spectrum of appearances on MRI. J Magn Reson Imaging2000;11:141. CrossRef

308. SrirajaskanthanR, WatkinsJ, MarelliL, et al.Expression of somatostatin and dopamine 2 receptors in neuroendocrine tumours and the potential role for new biotherapies. Neuroendocrinology2009;89:308. CrossRef

309. ReubiJ, WaserB, SchaerJ, et al.Somatostatin receptor sst1‐sst5 expression in normal and neoplastic human tissues using receptor autoradiography with subtype‐selective ligands. Eur J Nucl Med2001;28:836. CrossRef

310. PapottiM, BongiovanniM, VolanteM, et al.Expression of somatostatin receptor types 1‐5 in 81 cases of gastrointestinal and pancreatic endocrine tumors. A correlative immunohistochemical and reverse‐transcriptase polymerase chain reaction analysis. Virchows Arch2002;440:461. CrossRef

311. KrenningE, KwekkeboomD, BakkerW, et al.Somatostatin receptor scintigraphy with [111In‐DTPA‐D‐Phe1]‐ and [123I‐Tyr3]‐octreotide: the Rotterdam experience with more than 1000 patients. Eur J Nucl Med1993;20:716. CrossRef

312. SchillaciO, SpanuA, ScopinaroF, et al.Somatostatin receptor scintigraphy with 111In‐pentetreotide in non‐functioning gastroenteropancreatic neuroendocrine tumors. Int J Oncol2003;23:1687.

313. GibrilF, ReynoldsJ, DoppmanJ, et al.Somatostatin receptor scintigraphy: its sensitivity compared with that of other imaging methods in detecting primary and metastatic gastrinomas. A prospective study. Ann Intern Med1996;125:26. CrossRef

314. GibrilF, DoppmanJ, ReynoldsJ, et al.Bone metastases in patients with gastrinomas: a prospective study of bone scanning, somatostatin receptor scanning, and magnetic resonance image in their detection, frequency, location, and effect of their detection on management. J Clin Oncol1998;16:1040.

315. LeboulleuxS, DromainC, VataireA, et al.Prediction and diagnosis of bone 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 Metab2008;93:3021. CrossRef

316. GibrilF, ReynoldsJ, ChenC, et al.Specificity of somatostatin receptor scintigraphy: a prospective study and effects of false‐positive localizations on management in patients with gastrinomas. J Nucl Med1999;40:539.

317. TermaniniB, GibrilF, ReynoldsJ, et al.Value of somatostatin receptor scintigraphy: a prospective study in gastrinoma of its effect on clinical management. Gastroenterology1997;112:335. CrossRef

318. JamarF, FiasseR, LenersN, et al.Somatostatin receptor imaging with indium‐111‐pentetreotide in gastroenteropancreatic neuroendocrine tumors: safety, efficacy and impact on patient management. J Nucl Med1995;36:542.

319. AhlmanH, TisellL, WängbergB, et al.Fjälling M. Somatostatin receptor imaging in patients with neuroendocrine tumors: preoperative and postoperative scintigraphy and intraoperative use of a scintillation detector. Semin Oncol1994;21(5 Suppl 13):21.

320. ChitiA, FantiS, SavelliG, et al.Comparison of somatostatin receptor imaging, computed tomography and ultrasound in the clinical management of neuroendocrine gastro‐entero‐pancreatic tumours. Eur J Nucl Med1998;25:1396. CrossRef

321. BinderupT, KniggeU, LoftA, et al.Functional imaging of neuroendocrine tumors: a head‐to‐head comparison of somatostatin receptor scintigraphy, 123I‐MIBG scintigraphy, and 18F‐FDG PET. J Nucl Med2010;51:704. CrossRef

322. KaltsasG, KorbonitsM, HeintzE, et al.Comparison of somatostatin analog and meta‐iodobenzylguanidine radionuclides in the diagnosis and localization of advanced neuroendocrine tumors. J Clin Endocrinol Metab2001;86:895. CrossRef

323. HoegerleS, AltehoeferC, GhanemN, et al.Whole‐body 18F dopa PET for detection of gastrointestinal carcinoid tumors. Radiology2001;220:373. CrossRef

324. BechererA, SzabóM, KaranikasG, et al.Imaging of advanced neuroendocrine tumors with (18)F‐FDOPA PET. J Nucl Med2004;45:1161.

325. KoopmansK, de VriesE, KemaI, et al.Staging of carcinoid tumours with 18F‐DOPA PET: a prospective, diagnostic accuracy study. Lancet Oncol2006;7:728. CrossRef

326. KauhanenS, SeppänenM, MinnH, et al.Clinical PET imaging of insulinoma and beta‐cell hyperplasia. Curr Pharm Des2010;16:1550. CrossRef

327. TessonnierL, SebagF, GhanderC, et al.Limited value of 18F‐F‐DOPA PET to localize pancreatic insulin‐secreting tumors in adults with hyperinsulinemic hypoglycemia. J Clin Endocrinol Metab2010;95:303. CrossRef

328. OrleforsH, SundinA, GarskeU, et al.Whole‐body (11)C‐5‐hydroxytryptophan positron emission tomography as a universal imaging technique for neuroendocrine tumors: comparison with somatostatin receptor scintigraphy and computed tomography. J Clin Endocrinol Metab2005;90:3392. CrossRef

329. KoopmansK, NeelsO, KemaI, et al.Improved staging of patients with carcinoid and islet cell tumors with 18F‐dihydroxy‐phenyl‐alanine and 11C‐5‐hydroxy‐tryptophan positron emission tomography. J Clin Oncol2008;26:1489. CrossRef

330. AmbrosiniV, TomassettiP, CastellucciP, et al.Comparison between 68Ga‐DOTA‐NOC and 18F‐DOPA PET for the detection of gastro‐entero‐pancreatic and lung neuro‐endocrine tumours. Eur J Nucl Med Mol Imaging2008;35:1431. CrossRef

331. KumarR, SharmaP, GargP, et al.Role of (68)Ga‐DOTATOC PET‐CT in the diagnosis and staging of pancreatic neuroendocrine tumours. Eur Radiol2011;21:2408. CrossRef

332. VersariA, CamelliniL, CarlinfanteG, et al.Ga‐68 DOTATOC PET, endoscopic ultrasonography, and multidetector CT in the diagnosis of duodenopancreatic neuroendocrine tumors: a single‐centre retrospective study. Clin Nucl Med2010;35:321. CrossRef

333. TregliaG, CastaldiP, RindiG, et al.Diagnostic performance of Gallium‐68 somatostatin receptor PET and PET/CT in patients with thoracic and gastroenteropancreatic neuroendocrine tumours: a meta‐analysis. Endocrine2012;42:80. CrossRef

334. GeijerH, BreimerLH. Somatostatin receptor PET/CT in neuroendocrine tumours: update on systematic review and meta‐analysis. Eur J Nucl Med Mol Imaging2013;40:1770. CrossRef

335. AmbrosiniV, CampanaD, BodeiL, et al.68Ga‐DOTANOC PET/CT clinical impact in patients with neuroendocrine tumors. J Nucl Med2010;51:669. CrossRef

336. WildD, MäckeH, ChristE, et al.Glucagon‐like peptide 1‐receptor scans to localize occult insulinomas. N Engl J Med2008;359:766. CrossRef

337. ChristE, WildD, ForrerF, et al.Glucagon‐like peptide‐1 receptor imaging for localization of insulinomas. J Clin Endocrinol Metab2009;94:4398. CrossRef

338. DoppmanJ, ChangR, FrakerD, et al.Localization of insulinomas to regions of the pancreas by intra‐arterial stimulation with calcium. Ann Intern Med1995;123:269. CrossRef

339. RöschT, LightdaleC, BotetJ, et al.Localization of pancreatic endocrine tumors by endoscopic ultrasonography. N Engl J Med1992;326:1721. CrossRef

340. FeinJ, GerdesH. Localization of islet cell tumors by endoscopic ultrasonography. Gastroenterology1992;103:711.

341. McLeanA, FaircloughP. Endoscopic ultrasound in the localisation of pancreatic islet cell tumours. Best Pract Res Clin Endocrinol Metab2005;19:177. CrossRef

342. GaugerP, ScheimanJ, WamstekerEJ, et al.Role of endoscopic ultrasonography in screening and treatment of pancreatic endocrine tumours in asymptomatic patients with multiple endocrine neoplasia type 1. Br J Surg2003;90:748. CrossRef

343. WongM, IsaS, ZahiahM, et al.Intraoperative ultrasound with palpation is still superior to intra‐arterial calcium stimulation test in localising insulinoma. World J Surg2007;31:586. CrossRef

344. HellmanP, AnderssonM, RastadJ, et al.Surgical strategy for large or malignant endocrine pancreatic tumors. World J Surg2000;24:1353. CrossRef

345. NortonJ, KivlenM, LiM, et al.Morbidity and mortality of aggressive resection in patients with advanced neuroendocrine tumors. Arch Surg2003;138:859. CrossRef

346. NortonJ, WarrenR, KellyM, et al.Aggressive surgery for metastatic liver neuroendocrine tumors. Surgery2003;134:1057. CrossRef

347. KniggeU, HansenC. Surgery for GEP‐NETs. Best Pract Res Clin Gastroenterol2012;26:819. CrossRef

348. HackertT, HinzU, FritzS, et al.Enucleation in pancreatic surgery: indications, technique, and outcome compared to standard pancreatic resections. Langenbecks Arch Surg2011;396:1197. CrossRef

349. HouseM, CameronJ, LillemoeK, et al.Differences in survival for patients with resectable versus unresectable metastases from pancreatic islet cell cancer. J Gastrointest Surg2006;10:138. CrossRef

350. ChenH, HardacreJ, UzarA, et al.Isolated liver metastases from neuroendocrine tumors: does resection prolong survival?J Am Coll Surg1998;187:88. CrossRef

351. ChamberlainR, CanesD, BrownK, et al.Hepatic neuroendocrine metastases: does intervention alter outcomes?J Am Coll Surg2000;190:432. CrossRef

352. TouziosJ, KielyJ, PittS, et al.Neuroendocrine hepatic metastases: does aggressive management improve survival?Ann Surg2005;241:776. CrossRef

353. FischerL, KleeffJ, EspositoI, et al.Clinical outcome and long‐term survival in 118 consecutive patients with neuroendocrine tumours of the pancreas. Br J Surg2008;95:627. CrossRef

354. NortonJ, FangT, JensenR. Surgery for gastrinoma and insulinoma in multiple endocrine neoplasia type 1. J Natl Compr Canc Netw2006;4:148.

355. ShimonI, YanX, TaylorJ, et al.Somatostatin receptor (SSTR) subtype‐selective analogues differentially suppress in vitro growth hormone and prolactin in human pituitary adenomas. Novel potential therapy for functional pituitary tumors. J Clin Invest1997;100:2386. CrossRef

356. KvolsL, ObergK, O'DorisioT, et al.Pasireotide (SOM230) shows efficacy and tolerability in the treatment of patients with advanced neuroendocrine tumors refractory or resistant to octreotide LAR: results from a phase II study. Endocr Relat Cancer2012;19:657. CrossRef

357. BrunsC, LewisI, BrinerU, et al.SOM230: a novel somatostatin peptidomimetic with broad somatotropin release inhibiting factor (SRIF) receptor binding and a unique antisecretory profile. Eur J Endocrinol2002;146:707. CrossRef

358. MariotP, GilonP, NenquinM, et al.Tolbutamide and diazoxide influence insulin secretion by changing the concentration but not the action of cytoplasmic Ca2+ in beta‐cells. Diabetes1998;47:365. CrossRef

359. GoodeP, FarndonJ, AndersonJ, et al.Diazoxide in the management of patients with insulinoma. World J Surg1986;10:586. CrossRef

360. GillG, RaufO, MacFarlaneI. Diazoxide treatment for insulinoma: a national UK survey. Postgrad Med J1997;73:640. CrossRef

361. VezzosiD, BennetA, RochaixP, et al.Octreotide in insulinoma patients: efficacy on hypoglycemia, relationships with Octreoscan scintigraphy and immunostaining with anti‐sst2A and anti‐sst5 antibodies. Eur J Endocrinol2005;152:757. CrossRef

362. BernardV, Lombard‐BohasC, TaquetM‐C, et al.Efficacy of everolimus in patients with metastatic insulinoma and refractory hypoglycemia. Eur J Endocrinol2013;168:665. CrossRef

363. KulkeM, BergslandE, YaoJ. Glycemic control in patients with insulinoma treated with everolimus. N Engl J Med2009;360:195. CrossRef

364. BlumI, AderkaD, DoronM, et al.Suppression of hypoglycemia by DL‐propranolol in malignant insulinoma. N Engl J Med1978;299:487.

365. ScandellariC, ZaccariaM, De PaloC, et al.The effect of propranolol on hypoglycaemia. Observations in five insulinoma patients. Diabetologia1978;15:297. CrossRef

366. TaniguchiH, MurakamiK, MoritaS, et al.Calcium antagonist (diltiazem) for reversal of hypoglycaemic symptoms in insulinoma. Lancet1977;2:501. CrossRef

367. CohenM, BowerR, FidlerS, et al.Inhibition of insulin release by diphenylhydantoin and diazoxide in a patient with benign insulinoma. Lancet1973;1:40. CrossRef

368. NovotnyJ, JankuF, MaresP, et al.Symptomatic control of hypoglycaemia with prednisone in refractory metastatic pancreatic insulinoma. Support Care Cancer2005;13:760. CrossRef

369. BodenG, RyanI, EisenschmidB, et al.Treatment of inoperable glucagonoma with the long‐acting somatostatin analogue SMS 201‐995. N Engl J Med1986;314:1686. CrossRef

370. RosenbaumA, FlourieB, ChagnonS, et al.Octreotide (SMS 201‐995) in the treatment of metastatic glucagonoma: report of one case and review of the literature. Digestion1989;42:116. CrossRef

371. MatonP, O'DorisioT, HoweB, et al.Effect of a long‐acting somatostatin analogue (SMS 201‐995) in a patient with pancreatic cholera. N Engl J Med1985;312:17. CrossRef

372. ItoT, IgarashiH, JensenR. Pancreatic neuroendocrine tumors: clinical features, diagnosis and medical treatment: advances. Best Pract Res Clin Gastroenterol2012;26:737. CrossRef

373. AngelettiS, CorletoV, SchillaciO, et al.Use of the somatostatin analogue octreotide to localise and manage somatostatin‐producing tumours. Gut1998;42:792. CrossRef

374. ObergK, KvolsL, CaplinM, et al.Consensus report on the use of somatostatin analogs for the management of neuroendocrine tumors of the gastroenteropancreatic system. Ann Oncol2004;15:966. CrossRef

375. KvolsL, MoertelC, O'ConnellM, et al.Treatment of the malignant carcinoid syndrome. Evaluation of a long‐acting somatostatin analogue. N Engl J Med1986;315:663. CrossRef

376. O'TooleD, DucreuxM, BommelaerG, et al.Treatment of carcinoid syndrome: a prospective crossover evaluation of lanreotide versus octreotide in terms of efficacy, patient acceptability, and tolerance. Cancer2000;88:770. CrossRef

377. di BartolomeoM, BajettaE, BuzzoniR, et al.Clinical efficacy of octreotide in the treatment of metastatic neuroendocrine tumors. A study by the Italian Trials in Medical Oncology Group. Cancer1996;77:402. CrossRef

378. KvolsLK, ObergKE, O'DorisioTM, et al.Pasireotide (SOM230) shows efficacy and tolerability in the treatment of patients with advanced neuroendocrine tumors refractory or resistant to octreotide LAR: results from a phase II study. Endocr Relat Cancer2012;19:657. CrossRef

379. WymengaA, de VriesE, LeijsmaM, et al.Effects of ondansetron on gastrointestinal symptoms in carcinoid syndrome. Eur J Cancer (Oxford, England: 1990)1998;34:1293. CrossRef

380. ObergK. The action of interferon alpha on human carcinoid tumours. Semin Cancer Biol1992;3:35.

381. DetjenK, WelzelM, FarwigK, et al.Molecular mechanism of interferon alfa‐mediated growth inhibition in human neuroendocrine tumor cells. Gastroenterology2000;118:735. CrossRef

382. LewisM, JaramilloS, RobertsL, et al.Hepatic artery embolization for neuroendocrine tumors: postprocedural management and complications. Oncologist2012;17:725. CrossRef

383. KinneyM, WarnerM, NagorneyD, et al.Perianaesthetic risks and outcomes of abdominal surgery for metastatic carcinoid tumours. Br J Anaesth2001;87:447. CrossRef

384. SeymourN, SawhS. Mega‐dose intravenous octreotide for the treatment of carcinoid crisis: a systematic review. Can J Anaesth2013;60:492. CrossRef

385. GillamsA, CassoniA, ConwayG, et al.Radiofrequency ablation of neuroendocrine liver metastases: the Middlesex experience. Abdom Imaging2005;30:435. CrossRef

386. AkyildizH, MitchellJ, MilasM, et al.Laparoscopic radiofrequency thermal ablation of neuroendocrine hepatic metastases: long‐term follow‐up. Surgery2010;148:1288. CrossRef

387. SteinmüllerT, KianmaneshR, FalconiM, et al.Consensus guidelines for the management of patients with liver metastases from digestive (neuro)endocrine tumors: foregut, midgut, hindgut, and unknown primary. Neuroendocrinology2008;87:47. CrossRef

388. MaireF, Lombard‐BohasC, O'TooleD, et al.Hepatic arterial embolization versus chemoembolization in the treatment of liver metastases from well‐differentiated midgut endocrine tumors: a prospective randomized study. Neuroendocrinology2012;96:294. CrossRef

389. RickeJ, WustP, WienersG, et al.Liver malignancies: CT‐guided interstitial brachytherapy in patients with unfavorable lesions for thermal ablation. J Vasc Interv Radiol2004;15:1279. CrossRef

390. MittyH, WarnerR, NewmanL, et al.Control of carcinoid syndrome with hepatic artery embolization. Radiology1985;155:623. CrossRef

391. RocheA, GirishB, de BaèreT, et al.Trans‐catheter arterial chemoembolization as first‐line treatment for hepatic metastases from endocrine tumors. Eur Radiol2003;13:136.

392. MarracheF, VulliermeM, RoyC, et al.Arterial phase enhancement and body mass index are predictors of response to chemoembolisation for liver metastases of endocrine tumours. Br J Cancer2007;96:49. CrossRef

393. RuszniewskiP, RougierP, RocheA, et al.Hepatic arterial chemoembolization in patients with liver metastases of endocrine tumors. A prospective phase II study in 24 patients. Cancer1993;71:2624. CrossRef

394. TherasseE, BreittmayerF, RocheA, et al.Transcatheter chemoembolization of progressive carcinoid liver metastasis. Radiology1993;189:541. CrossRef

395. PerryL, StuartK, StokesK, et al.Hepatic arterial chemoembolization for metastatic neuroendocrine tumors. Surgery1994;116:1111.

396. LehnertT. Liver transplantation for metastatic neuroendocrine carcinoma: an analysis of 103 patients. Transplantation1998;66:1307. CrossRef

397. GedalyR, DailyM, DavenportD, et al.Liver transplantation for the treatment of liver metastases from neuroendocrine tumors: an analysis of the UNOS database. Arch Surg2011;146:953. CrossRef

398. RosenauJ, BahrM, von WasielewskiR, et al.Ki67, E‐cadherin, and p53 as prognostic indicators of long‐term outcome after liver transplantation for metastatic neuroendocrine tumors. Transplantation2002;73:386. CrossRef

399. MoertelC, HanleyJ, JohnsonL. Streptozocin alone compared with streptozocin plus fluorouracil in the treatment of advanced islet‐cell carcinoma. N Engl J Med1980;303:1189. CrossRef

400. MoertelC, LefkopouloM, LipsitzS, et al.Streptozocin‐doxorubicin, streptozocin‐fluorouracil or chlorozotocin in the treatment of advanced islet‐cell carcinoma. N Engl J Med1992;326:519. CrossRef

401. DelaunoitT, DucreuxM, BoigeV, et al.The doxorubicin‐streptozotocin combination for the treatment of advanced well‐differentiated pancreatic endocrine carcinoma; a judicious option?Eur J Cancer2004;40:515. CrossRef

402. FjallskogM‐LH, JansonE, FalkmerU, et al.Treatment with combined streptozotocin and liposomal doxorubicin in metastatic endocrine pancreatic tumors. Neuroendocrinology2008;88:53. CrossRef

403. KulkeM, StuartK, EnzingerP, et al.Phase II study of temozolomide and thalidomide in patients with metastatic neuroendocrine tumors. J Clin Oncol2006;24:401. CrossRef

404. StrosbergJ, FineR, ChoiJ, et al.First‐line chemotherapy with capecitabine and temozolomide in patients with metastatic pancreatic endocrine carcinomas. Cancer2011;117:268. CrossRef

405. ChanJ, StuartK, EarleC, et al.Prospective study of bevacizumab plus temozolomide in patients with advanced neuroendocrine tumors. J Clin Oncol2012;30:2963. CrossRef

406. KulkeM, HornickJ, FrauenhofferC, et al.O6‐methylguanine DNA methyltransferase deficiency and response to temozolomide‐based therapy in patients with neuroendocrine tumors. Clin Cancer Res2009;15:338. CrossRef

407. PavelM, BaudinE, CouvelardA, 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. Neuroendocrinology2011;95:157. CrossRef

408. MoertelC, KvolsL, O'ConnellM, et al.Treatment of neuroendocrine carcinomas with combined etoposide and cisplatin. Evidence of major therapeutic activity in the anaplastic variants of these neoplasms. Cancer1991;68:227. CrossRef

409. MitryE, BaudinE, DucreuxM, et al.Treatment of poorly differentiated neuroendocrine tumours with etoposide and cisplatin. Br J Cancer1999;81:1351. CrossRef

410. FjällskogM, GranbergD, WelinS, et al.Treatment with cisplatin and etoposide in patients with neuroendocrine tumors. Cancer2001;92:1101. CrossRef

411. SorbyeH, WelinS, LangerS, 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 Oncol2013;24:152. CrossRef

412. WelinS, SorbyeH, SebjornsenS, et al.Clinical effect of temozolomide‐based chemotherapy in poorly differentiated endocrine carcinoma after progression on first‐line chemotherapy. Cancer2011;117:4617. CrossRef

413. BajettaE, CatenaL, ProcopioG, et al.Are capecitabine and oxaliplatin (XELOX) suitable treatments for progressing low‐grade and high‐grade neuroendocrine tumours?Cancer Chemother Pharmacol2007;59:637. CrossRef

414. ErikssonB, ObergK. Summing up 15 years of somatostatin analog therapy in neuroendocrine tumors: future outlook. Ann Oncol1999;10(Suppl 2):8. CrossRef

415. AparicioT, DucreuxM, BaudinE, et al.Antitumour activity of somatostatin analogues in progressive metastatic neuroendocrine tumours. Eur J Cancer (Oxford, England: 1990)2001;37:1014. CrossRef

416. ArnoldR, TrautmannM, CreutzfeldtW, et al.Somatostatin analogue octreotide and inhibition of tumour growth in metastatic endocrine gastroenteropancreatic tumours. Gut1996;38:430. CrossRef

417. WymengaA, ErikssonB, SalmelaP, et al.Efficacy and safety of prolonged‐release lanreotide in patients with gastrointestinal neuroendocrine tumors and hormone‐related symptoms. J Clin Oncol1999;17:1111.

418. WelinS, JansonE, SundinA, et al.High‐dose treatment with a long‐acting somatostatin analogue in patients with advanced midgut carcinoid tumours. Eur J Endocrinol2004;151:107. CrossRef

419. FaissS, RäthU, MansmannU, et al.Ultra‐high‐dose lanreotide treatment in patients with metastatic neuroendocrine gastroenteropancreatic tumors. Digestion1999;60:469. CrossRef

420. RinkeA, MüllerH‐H, Schade‐BrittingerC, 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 Oncol2009;27:4656. CrossRef

421. CaplinME, PavelM, ĆwikłaJB, et al.Lanreotide in metastatic enteropancreatic neuroendocrine tumors. N Engl J Med2014;371:224. CrossRef

422. ObergK, FunaK, AlmG. Effects of leukocyte interferon on clinical symptoms and hormone levels in patients with mid‐gut carcinoid tumors and carcinoid syndrome. N Engl J Med1983;309:129. CrossRef

423. MoertelC, RubinJ, KvolsL. Therapy of metastatic carcinoid tumor and the malignant carcinoid syndrome with recombinant leukocyte A interferon. J Clin Oncol1989;7:865.

424. CreutzfeldtW, BartschH, JacubaschkeU, et al.Treatment of gastrointestinal endocrine tumours with interferon‐alpha and octreotide. Acta Oncol1991;30:529. CrossRef

425. PavelM, BaumU, HahnE, et al.Efficacy and tolerability of pegylated IFN‐alpha in patients with neuroendocrine gastroenteropancreatic carcinomas. J Interferon Cytokine Res2006;26:8. CrossRef

426. ArnoldR, RinkeA, KloseK‐J, et al.Octreotide versus octreotide plus interferon‐alpha in endocrine gastroenteropancreatic tumors: a randomized trial. Clin Gastroenterol Hepatol2005;3:761. CrossRef

427. JansonE, ObergK. Long‐term management of the carcinoid syndrome. Treatment with octreotide alone and in combination with alpha‐interferon. Acta Oncol1993;32:225. CrossRef

428. KölbyL, PerssonG, FranzénS, et al.Randomized clinical trial of the effect of interferon alpha on survival in patients with disseminated midgut carcinoid tumours. Br J Surg2003;90:687. CrossRef

429. FaissS, PapeU‐F, BöhmigM, et al.Prospective, randomized, multicenter trial on the antiproliferative effect of lanreotide, interferon alfa, and their combination for therapy of metastatic neuroendocrine gastroenteropancreatic tumors–the International Lanreotide and Interferon Alfa Study Group. J Clin Oncol2003;21:2689. CrossRef

430. RaymondE, DahanL, RaoulJ‐LL, et al.Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. N Engl J Med2011;364:501. CrossRef

431. YaoJ, ShahM, ItoT, et al.Everolimus for advanced pancreatic neuroendocrine tumors. N Engl J Med2011;364:514. CrossRef

432. PavelM, HainsworthJ, BaudinE, 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. Lancet2011;378:2005. CrossRef

433. OtteA, HerrmannR, HeppelerA, et al.Yttrium‐90 DOTATOC: first clinical results. Eur J Nucl Med1999;26:1439. CrossRef

434. WaldherrC, PlessM, MaeckeH, et al.The clinical value of [90Y‐DOTA]‐D‐Phe1‐Tyr3‐octreotide (90Y‐DOTATOC) in the treatment of neuroendocrine tumours: a clinical phase II study. Ann Oncol2001;12:941. CrossRef

435. WaldherrC, PlessM, MaeckeH, et al.Tumor response and clinical benefit in neuroendocrine tumors after 7.4 GBq (90)Y‐DOTATOC. J Nucl Med2002;43:610.

436. BodeiL, CremonesiM, ZoboliS, et al.Receptor‐mediated radionuclide therapy with 90Y‐DOTATOC in association with amino acid infusion: a phase I study. Eur J Nucl Med Mol Imaging2003;30:207. CrossRef

437. ValkemaR, PauwelsS, KvolsL, et al.Survival and response after peptide receptor radionuclide therapy with [90Y‐DOTA0,Tyr3]octreotide in patients with advanced gastroenteropancreatic neuroendocrine tumors. Semin Nucl Med2006;36:147. CrossRef

438. BushnellD, O'DorisioT, O'DorisioM, et al.90Y‐edotreotide for metastatic carcinoid refractory to octreotide. J Clin Oncol2010;28:1652. CrossRef

439. PfeiferA, GregersenT, GrønbækH, et al.Peptide receptor radionuclide therapy with Y‐DOTATOC and (177)Lu‐DOTATOC in advanced neuroendocrine tumors: results from a Danish cohort treated in Switzerland. Neuroendocrinology2011;93:189. CrossRef

440. VirgoliniI, BrittonK, BuscombeJ, et al.In‐ and Y‐DOTA‐lanreotide: results and implications of the MAURITIUS trial. Semin Nucl Med2002;32:148. CrossRef

441. CwiklaJ, SankowskiA, SekleckaN, et al.Efficacy of radionuclide treatment DOTATATE Y‐90 in patients with progressive metastatic gastroenteropancreatic neuroendocrine carcinomas (GEP‐NETs): a phase II study. Ann Oncol2010;21:787. CrossRef

442. KwekkeboomD, de HerderW, KamB, et al.Treatment with the radiolabeled somatostatin analog [177 Lu‐DOTA 0,Tyr3]octreotate: toxicity, efficacy, and survival. J Clin Oncol2008;26:2124. CrossRef

443. SwärdC, BernhardtP, AhlmanH, et al.[177Lu‐DOTA 0‐Tyr 3]‐octreotate treatment in patients with disseminated gastroenteropancreatic neuroendocrine tumors: the value of measuring absorbed dose to the kidney. World J Surg2010;34:1368. CrossRef

444. GarkavijM, NickelM, Sjögreen‐GleisnerK, et al.177Lu‐[DOTA0,Tyr3] octreotate therapy in patients with disseminated neuroendocrine tumors: analysis of dosimetry with impact on future therapeutic strategy. Cancer2010;116(4 Suppl):1084. CrossRef

445. BodeiL, CremonesiM, GranaC, et al.Peptide receptor radionuclide therapy with 177Lu‐DOTATATE: the IEO phase I‐II study. Eur J Nucl Med Mol Imaging2011;38:2125. CrossRef