Skip to main content

Advertisement

Log in

The diversity and commonalities of gastroenteropancreatic neuroendocrine tumors

  • Review Article
  • Published:
Langenbeck's Archives of Surgery Aims and scope Submit manuscript

Abstract

Background

Recent data demonstrate that the incidence of gastroenteropancreatic neuroendocrine tumors (GEP-NETs) has increased exponentially (overall ~500%) over the last three decades, thus refuting the erroneous concept of rarity. GEP-NETs comprise 2% of all malignancies and in terms of prevalence, are the second commonest gastrointestinal malignancy after colorectal cancer. Diagnosis is usually late since there is no biochemical screening test and symptoms are protean and overlooked. As a consequence, 60–80% exhibit metastases with a consequent suboptimal outcome.

Discussion

The gastrointestinal tract and pancreas exhibit ~17 different neuroendocrine cell types, but neither the cell of origin nor the biological basis of GEP-NETs is understood. This review examines GEP-NETs from the cellular and molecular perspective and addresses the distinct patterns of functional tumor biology pertinent to clinicians. Although grouped as a neoplastic entity (NETs), each lesion is derived from distinct cell precursors, produces specific bioactive products, exhibits distinct chromosomal abnormalities and somatic mutation events and has uniquely dissimilar clinical presentations. GEP-NETs demonstrate very different survival rates reflecting the intrinsic differences in malignant potential and variations in proliferative regulation. Apart from the identification of the inhibitory role of the somatostatin receptors, there is limited biological knowledge of the key regulators of proliferation and hence a paucity of successful targeted therapeutic agents. IGF-I, TGFβ and a variety of tyrosine kinases have been postulated as key regulatory elements; rigorous data is still required to define predictably effective and rational therapeutic strategy in an individual tumor. A critical issue in the clinical management of GEP-NETs is the need to appreciate both the neuroendocrine commonalities of the disease as well as the unique characteristics of each tumor. The further acquisition of a detailed biological and molecular appreciation of GEP-NETs is vital to the development of effective management strategy.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Institutional subscriptions

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7
Fig. 8
Fig. 9
Fig. 10
Fig. 11

Similar content being viewed by others

References

  1. Modlin IM, Oberg K, Chung DC, Jensen RT, de Herder WW, Thakker RV, Caplin M, Delle Fave G, Kaltsas GA, Krenning EP, Moss SF, Nilsson O, Rindi G, Salazar R, Ruszniewski P, Sundin A (2008) Gastroenteropancreatic neuroendocrine tumours. Lancet Oncol 9(1):61–72. doi:10.1016/S1470-2045(07)70410-2

    Article  PubMed  CAS  Google Scholar 

  2. Modlin IM, Gustafsson BI, Moss SF, Pavel M, Tsolakis AV, Kidd M (2010) Chromogranin A-biological function and clinical utility in neuro endocrine tumor disease. Ann Surg Oncol. doi:10.1245/s10434-010-1006-3

    PubMed  Google Scholar 

  3. Calender A (2000) Molecular genetics of neuroendocrine tumors. Digestion 62(Suppl 1):3–18

    Article  PubMed  CAS  Google Scholar 

  4. Kloppel G, Perren A, Heitz PU (2004) The gastroenteropancreatic neuroendocrine cell system and its tumors: the WHO classification. Ann NY Acad Sci 1014:13–27

    Article  PubMed  Google Scholar 

  5. Andrew A, Kramer B, Rawdon B (1998) The origin of gut and pancreatic neuroendocrine (APUD) cells—the last word? J Pathol 186:117–118

    Article  PubMed  CAS  Google Scholar 

  6. Waldum HL, Rorvik H, Falkmer S, Kawase S (1999) Neuroendocrine (ECL cell) differentiation of spontaneous gastric carcinomas of cotton rats (Sigmodon hispidus). Lab Anim Sci 49(3):241–247

    PubMed  CAS  Google Scholar 

  7. Wright NA (1999) Letter from Waldum et al. commenting on the editorial by Andrew et al. and responses. J Pathol 189(3):439–440

    Article  PubMed  CAS  Google Scholar 

  8. Pearse AG (1969) The cytochemistry and ultrastructure of polypeptide hormone-producing cells of the APUD series and the embryologic, physiologic and pathologic implications of the concept. J Histochem Cytochem 17(5):303–313

    PubMed  CAS  Google Scholar 

  9. Schonhoff SE, Giel-Moloney M, Leiter AB (2004) Minireview: development and differentiation of gut endocrine cells. Endocrinology 145(6):2639–2644

    Article  PubMed  CAS  Google Scholar 

  10. Novelli MR, Williamson JA, Tomlinson IP, Elia G, Hodgson SV, Talbot IC, Bodmer WF, Wright NA (1996) Polyclonal origin of colonic adenomas in an XO/XY patient with FAP. Science 272(5265):1187–1190

    Article  PubMed  CAS  Google Scholar 

  11. Taylor RW, Barron MJ, Borthwick GM, Gospel A, Chinnery PF, Samuels DC, Taylor GA, Plusa SM, Needham SJ, Greaves LC, Kirkwood TB, Turnbull DM (2003) Mitochondrial DNA mutations in human colonic crypt stem cells. J Clin Invest 112(9):1351–1360

    PubMed  CAS  Google Scholar 

  12. Greaves LC, Preston SL, Tadrous PJ, Taylor RW, Barron MJ, Oukrif D, Leedham SJ, Deheragoda M, Sasieni P, Novelli MR, Jankowski JA, Turnbull DM, Wright NA, McDonald SA (2006) Mitochondrial DNA mutations are established in human colonic stem cells, and mutated clones expand by crypt fission. Proc Natl Acad Sci USA 103(3):714–719

    Article  PubMed  CAS  Google Scholar 

  13. McDonald SA, Greaves LC, Gutierrez-Gonzalez L, Rodriguez-Justo M, Deheragoda M, Leedham SJ, Taylor RW, Lee CY, Preston SL, Lovell M, Hunt T, Elia G, Oukrif D, Harrison R, Novelli MR, Mitchell I, Stoker DL, Turnbull DM, Jankowski JA, Wright NA (2008) Mechanisms of field cancerization in the human stomach: the expansion and spread of mutated gastric stem cells. Gastroenterology 134(2):500–510

    Article  PubMed  CAS  Google Scholar 

  14. Bouwens L, Kloppel G (1996) Islet cell neogenesis in the pancreas. Virchows Arch 427(6):553–560

    Article  PubMed  CAS  Google Scholar 

  15. Paris M, Tourrel-Cuzin C, Plachot C, Ktorza A (2004) Review: pancreatic beta-cell neogenesis revisited. Exp Diabesity Res 5(2):111–121. doi:10.1080/15438600490455079

    Article  PubMed  CAS  Google Scholar 

  16. Wang J, Cortina G, Wu SV, Tran R, Cho JH, Tsai MJ, Bailey TJ, Jamrich M, Ament ME, Treem WR, Hill ID, Vargas JH, Gershman G, Farmer DG, Reyen L, Martin MG (2006) Mutant neurogenin-3 in congenital malabsorptive diarrhea. N Engl J Med 355(3):270–280

    Article  PubMed  CAS  Google Scholar 

  17. Barrett P, Hobbs RC, Coates PJ, Risdon RA, Wright NA, Hall PA (1995) Endocrine cells of the human gastrointestinal tract have no proliferative capacity. Histochem J 27(6):482–486

    PubMed  CAS  Google Scholar 

  18. Karam S, Leblond CP (1995) Origin and migratory pathways of the eleven epithelial cell types present in the body of the mouse stomach. Microsc Res Tech 31(3):193–214. doi:10.1002/jemt.1070310304

    Article  PubMed  CAS  Google Scholar 

  19. Rindi G, Leiter AB, Kopin AS, Bordi C, Solcia E (2004) The “normal” endocrine cell of the gut: changing concepts and new evidences. Ann NY Acad Sci 1014:1–12

    Article  PubMed  CAS  Google Scholar 

  20. Kidd M, Modlin IM, Tang LH (1998) Gastrin and the enterochromaffin-like cell: an acid update. Dig Surg 15(3):209–217

    Article  PubMed  CAS  Google Scholar 

  21. Wiedenmann B, John M, Ahnert-Hilger G, Riecken EO (1998) Molecular and cell biological aspects of neuroendocrine tumors of the gastroenteropancreatic system. J Mol Med 76(9):637–647

    Article  PubMed  CAS  Google Scholar 

  22. Bloom SR (1978) Gut hormones. Proc Nutr Soc 37(3):259–271

    Article  PubMed  CAS  Google Scholar 

  23. Zanner R, Gratzl M, Prinz C (2002) Circle of life of secretory vesicles in gastric enterochromaffin-like cells. Ann NY Acad Sci 971:389–396

    Article  PubMed  CAS  Google Scholar 

  24. Amara SG, Kuhar MJ (1993) Neurotransmitter transporters: recent progress. Annu Rev Neurosci 16:73–93. doi:10.1146/annurev.ne.16.030193.000445

    Article  PubMed  CAS  Google Scholar 

  25. Erickson JD, Schafer MK, Bonner TI, Eiden LE, Weihe E (1996) Distinct pharmacological properties and distribution in neurons and endocrine cells of two isoforms of the human vesicular monoamine transporter. Proc Natl Acad Sci USA 93(10):5166–5171

    Article  PubMed  CAS  Google Scholar 

  26. Kidd M, Modlin IM, Gustafsson BI, Drozdov I, Hauso O, Pfragner R (2008) Luminal regulation of normal and neoplastic human EC cell serotonin release is mediated by bile salts, amines, tastants, and olfactants. Am J Physiol Gastrointest Liver Physiol 295(2):G260–G272. doi:10.1152/ajpgi.00056.2008

    Article  PubMed  CAS  Google Scholar 

  27. Jahn R, Lang T, Sudhof TC (2003) Membrane fusion. Cell 112(4):519–533

    Article  PubMed  CAS  Google Scholar 

  28. Stenmark H, Olkkonen VM (2001) The Rab GTPase family. Genome Biol 2 (5):REVIEWS3007

    Google Scholar 

  29. Helpap B, Kollermann J (2001) Immunohistochemical analysis of the proliferative activity of neuroendocrine tumors from various organs. Are there indications for a neuroendocrine tumor-carcinoma sequence? Virchows Arch 438(1):86–91

    Article  PubMed  CAS  Google Scholar 

  30. Leotlela PD, Jauch A, Holtgreve-Grez H, Thakker RV (2003) Genetics of neuroendocrine and carcinoid tumours. Endocr Relat Cancer 10(4):437–450

    Article  PubMed  CAS  Google Scholar 

  31. Hiripi E, Bermejo JL, Sundquist J, Hemminki K (2009) Familial gastrointestinal carcinoid tumours and associated cancers. Ann Oncol 20(5):950–954

    Article  PubMed  CAS  Google Scholar 

  32. Wermer P (1954) Genetic aspects of adenomatosis of endocrine glands. Am J Med 16(3):363–371

    Article  PubMed  CAS  Google Scholar 

  33. Larsson C, Skogseid B, Oberg K, Nakamura Y, Nordenskjold M (1988) Multiple endocrine neoplasia type 1 gene maps to chromosome 11 and is lost in insulinoma. Nature 332(6159):85–87. doi:10.1038/332085a0

    Article  PubMed  CAS  Google Scholar 

  34. Knudson AG Jr (1971) Mutation and cancer: statistical study of retinoblastoma. Proc Natl Acad Sci USA 68(4):820–823

    Article  PubMed  Google Scholar 

  35. Boissan M, De Wever O, Lizarraga F, Wendum D, Poincloux R, Chignard N, Desbois-Mouthon C, Dufour S, Nawrocki-Raby B, Birembaut P, Bracke M, Chavrier P, Gespach C, Lacombe ML (2010) Implication of metastasis suppressor NM23-H1 in maintaining adherens junctions and limiting the invasive potential of human cancer cells. Cancer Res 70(19):7710–7722. doi:10.1158/0008-5472.CAN-10-1887

    Article  PubMed  CAS  Google Scholar 

  36. Dimou AT, Syrigos KN, Saif MW (2010) Neuroendocrine tumors of the pancreas: what’s new. Highlights from the “2010 ASCO Gastrointestinal Cancers Symposium”. Orlando, FL, USA. JOP 11(2):135–138

    PubMed  Google Scholar 

  37. Donis-Keller H, Dou S, Chi D, Carlson KM, Toshima K, Lairmore TC, Howe JR, Moley JF, Goodfellow P, Wells SA Jr (1993) Mutations in the RET proto-oncogene are associated with MEN 2A and FMTC. Hum Mol Genet 2(7):851–856

    Article  PubMed  CAS  Google Scholar 

  38. Vasen HF, van der Feltz M, Raue F, Kruseman AN, Koppeschaar HP, Pieters G, Seif FJ, Blum WF, Lips CJ (1992) The natural course of multiple endocrine neoplasia type IIb. A study of 18 cases. Arch Intern Med 152(6):1250–1252

    Article  PubMed  CAS  Google Scholar 

  39. Richard S, Giraud S, Beroud C, Caron J, Penfornis F, Baudin E, Niccoli-Sire P, Murat A, Schlumberger M, Plouin PF, Conte-Devolx B (1998) Von Hippel-Lindau disease: recent genetic progress and patient management. Francophone Study Group of von Hippel-Lindau Disease (GEFVH). Ann Endocrinol (Paris) 59(6):452–458

    CAS  Google Scholar 

  40. Neumann HP, Dinkel E, Brambs H, Wimmer B, Friedburg H, Volk B, Sigmund G, Riegler P, Haag K, Schollmeyer P et al (1991) Pancreatic lesions in the von Hippel-Lindau syndrome. Gastroenterology 101(2):465–471

    PubMed  CAS  Google Scholar 

  41. Latif F, Tory K, Gnarra J, Yao M, Duh FM, Orcutt ML, Stackhouse T, Kuzmin I, Modi W, Geil L et al (1993) Identification of the von Hippel-Lindau disease tumor suppressor gene. Science 260(5112):1317–1320

    Article  PubMed  CAS  Google Scholar 

  42. Kibel A, Iliopoulos O, DeCaprio JA, Kaelin WG Jr (1995) Binding of the von Hippel-Lindau tumor suppressor protein to Elongin B and C. Science 269(5229):1444–1446

    Article  PubMed  CAS  Google Scholar 

  43. Bluyssen HA, Lolkema MP, van Beest M, Boone M, Snijckers CM, Los M, Gebbink MF, Braam B, Holstege FC, Giles RH, Voest EE (2004) Fibronectin is a hypoxia-independent target of the tumor suppressor VHL. FEBS Lett 556(1–3):137–142

    Article  PubMed  CAS  Google Scholar 

  44. Ruggieri M, Huson SM (1999) The neurofibromatoses. An overview. Ital J Neurol Sci 20(2):89–108

    Article  PubMed  CAS  Google Scholar 

  45. Au KS, Williams AT, Gambello MJ, Northrup H (2004) Molecular genetic basis of tuberous sclerosis complex: from bench to bedside. J Child Neurol 19(9):699–709

    PubMed  Google Scholar 

  46. Carney JA, Gordon H, Carpenter PC, Shenoy BV, Go VL (1985) The complex of myxomas, spotty pigmentation, and endocrine overactivity. Medicine (Baltimore) 64(4):270–283

    CAS  Google Scholar 

  47. Kirschner LS, Carney JA, Pack SD, Taymans SE, Giatzakis C, Cho YS, Cho-Chung YS, Stratakis CA (2000) Mutations of the gene encoding the protein kinase A type I-alpha regulatory subunit in patients with the Carney complex. Nat Genet 26(1):89–92. doi:10.1038/79238

    Article  PubMed  CAS  Google Scholar 

  48. Modlin IM, Lye KD, Kidd M (2003) Carcinoid tumors of the stomach. Surg Oncol 12(2):153–172

    Article  PubMed  Google Scholar 

  49. Rindi G, Solcia E (2007) Endocrine hyperplasia and dysplasia in the pathogenesis of gastrointestinal and pancreatic endocrine tumors. Gastroenterol Clin North Am 36(4):851–865. doi:10.1016/j.gtc.2007.08.006, vi

    Article  PubMed  Google Scholar 

  50. Rindi G, Bordi C, Rappel S, La Rosa S, Stolte M, Solcia E (1996) Gastric carcinoids and neuroendocrine carcinomas: pathogenesis, pathology, and behavior. World J Surg 20(2):168–172

    Article  PubMed  CAS  Google Scholar 

  51. Borch K, Ahren B, Ahlman H, Falkmer S, Granerus G, Grimelius L (2005) Gastric carcinoids: biologic behavior and prognosis after differentiated treatment in relation to type. Ann Surg 242(1):64–73

    Article  PubMed  Google Scholar 

  52. Rindi G, Luinetti O, Cornaggia M, Capella C, Solcia E (1993) Three subtypes of gastric argyrophil carcinoid and the gastric neuroendocrine carcinoma: a clinicopathologic study. Gastroenterology 104(4):994–1006

    PubMed  CAS  Google Scholar 

  53. Nakata K, Aishima S, Ichimiya H, Yao T, Matsuura T, Seo M, Nagai E, Okido M, Kato M, Nakagaki M, Tsuneyoshi M, Tanaka M (2010) Unusual multiple gastric carcinoids with hypergastrinemia: report of a case. Surg Today 40(3):267–271. doi:10.1007/s00595-009-4032-7

    Article  PubMed  Google Scholar 

  54. Lawrence B, Kidd M, Svejda B, Modlin I (2010) A clinical perspective on gastric neuroendocrine neoplasia. Curr Gastroenterol Rep. doi:10.1007/s11894-010-0158-4

    Google Scholar 

  55. Thomson AB, Sauve MD, Kassam N, Kamitakahara H (2010) Safety of the long-term use of proton pump inhibitors. World J Gastroenterol 16(19):2323–2330

    Article  PubMed  CAS  Google Scholar 

  56. Cao L, Mizoshita T, Tsukamoto T, Takenaka Y, Toyoda T, Cao X, Ban H, Nozaki K, Tatematsu M (2008) Development of carcinoid tumors of the glandular stomach and effects of eradication in Helicobacter pylori-infected Mongolian gerbils. Asian Pac J Cancer Prev 9(1):25–30

    PubMed  Google Scholar 

  57. Jensen RT (1998) Management of the Zollinger-Ellison syndrome in patients with multiple endocrine neoplasia type 1. J Intern Med 243(6):477–488

    Article  PubMed  CAS  Google Scholar 

  58. Berna MJ, Annibale B, Marignani M, Luong TV, Corleto V, Pace A, Ito T, Liewehr D, Venzon DJ, Delle Fave G, Bordi C, Jensen RT (2008) A prospective study of gastric carcinoids and enterochromaffin-like cell changes in multiple endocrine neoplasia type 1 and Zollinger-Ellison syndrome: identification of risk factors. J Clin Endocrinol Metab 93(5):1582–1591. doi:10.1210/jc.2007-2279

    Article  PubMed  CAS  Google Scholar 

  59. Bordi C, Yu JY, Baggi MT, Davoli C, Pilato FP, Baruzzi G, Gardini G, Zamboni G, Franzin G, Papotti M et al (1991) Gastric carcinoids and their precursor lesions. A histologic and immunohistochemical study of 23 cases. Cancer 67(3):663–672

    Article  PubMed  CAS  Google Scholar 

  60. Kim BS, Oh ST, Yook JH, Kim KC, Kim MG, Jeong JW (2010) Typical carcinoids and neuroendocrine carcinomas of the stomach: differing clinical courses and prognoses. Am J Surg 200(3):328–333. doi:10.1016/j.amjsurg.2009.10.028

    Article  PubMed  Google Scholar 

  61. Kaltsas GA, Cunningham J, Falkmer S, Grimelius L, Tsolakis A (2010) Expression of connective tissue growth factor and insulin growth factor 1 in normal and neoplastic gastrointestinal neuroendocrine cells and their clinicopathological significance. Endocr Relat Cancer. doi:10.1677/ERC-10-0026

    Google Scholar 

  62. Kidd M, Modlin IM, Eick GN, Camp RL, Mane SM (2007) Role of CCN2/CTGF in the proliferation of Mastomys enterochromaffin-like cells and gastric carcinoid development. Am J Physiol Gastrointest Liver Physiol 292(1):G191–G200. doi:10.1152/ajpgi.00131.2006

    Article  PubMed  CAS  Google Scholar 

  63. Lauffer JM, Tang LH, Zhang T, Hinoue T, Rahbar S, Odo M, Modlin IM, Kidd M (2001) PACAP mediates the neural proliferative pathway of Mastomys enterochromaffin-like cell transformation. Regul Pept 102(2–3):157–164

    Article  PubMed  CAS  Google Scholar 

  64. Tang LH, Modlin IM, Lawton GP, Kidd M, Chinery R (1996) The role of transforming growth factor alpha in the enterochromaffin-like cell tumor autonomy in an African rodent mastomys. Gastroenterology 111(5):1212–1223

    Article  PubMed  CAS  Google Scholar 

  65. Lawrence B, Gustafsson B, Chan A, Svejda B, Kidd M, Modlin I (2010) The epidemiology of gastroenteropancreatic neuroendocrine tumors. Endocrinol Metab Clin N Am (in press)

  66. Landry CS, Brock G, Scoggins CR, McMasters KM, Martin RC 2nd (2009) A proposed staging system for gastric carcinoid tumors based on an analysis of 1, 543 patients. Ann Surg Oncol 16(1):51–60. doi:10.1245/s10434-008-0192-8

    Article  PubMed  Google Scholar 

  67. Nilsson O, Van Cutsem E, Delle Fave G, Yao JC, Pavel ME, McNicol AM, Sevilla Garcia MI, Knapp WH, Kelestimur F, Sauvanet A, Pauwels S, Kwekkeboom DJ, Caplin M (2006) Poorly differentiated carcinomas of the foregut (gastric, duodenal and pancreatic). Neuroendocrinology 84(3):212–215. doi:10.1159/000098013

    Article  PubMed  CAS  Google Scholar 

  68. Kidd M, Modlin IM, Mane SM, Camp RL, Eick GN, Latich I, Zikusoka MN (2006) Utility of molecular genetic signatures in the delineation of gastric neoplasia. Cancer 106(7):1480–1488. doi:10.1002/cncr.21758

    Article  PubMed  CAS  Google Scholar 

  69. Jensen RT, Niederle B, Mitry E, Ramage JK, Steinmuller T, Lewington V, Scarpa A, Sundin A, Perren A, Gross D, O'Connor JM, Pauwels S, Kloppel G (2006) Gastrinoma (duodenal and pancreatic). Neuroendocrinology 84(3):173–182. doi:10.1159/000098009

    Article  PubMed  CAS  Google Scholar 

  70. Jensen RT, Rindi G, Arnold R, Lopes JM, Brandi ML, Bechstein WO, Christ E, Taal BG, Knigge U, Ahlman H, Kwekkeboom DJ, O'Toole D (2006) Well-differentiated duodenal tumor/carcinoma (excluding gastrinomas). Neuroendocrinology 84(3):165–172. doi:10.1159/000098008

    Article  PubMed  CAS  Google Scholar 

  71. Scherubl H, Faiss S, Jahn HU, Liehr RM, Schwertner C, Steinberg J, Stolzel U, Weinke T, Zimmer T, Kloppel G (2009) Neuroendocrine tumors of the stomach (gastric carcinoids) are on the rise: good prognosis with early detection. Dtsch Med Wochenschr 134(30):1529–1535. doi:10.1055/s-0029-1233975

    Article  PubMed  CAS  Google Scholar 

  72. Warner RR (2005) Enteroendocrine tumors other than carcinoid: a review of clinically significant advances. Gastroenterology 128(6):1668–1684

    Article  PubMed  Google Scholar 

  73. Jonkers YM, Ramaekers FC, Speel EJ (2007) Molecular alterations during insulinoma tumorigenesis. Biochim Biophys Acta 1775(2):313–332. doi:10.1016/j.bbcan.2007.05.004

    PubMed  CAS  Google Scholar 

  74. Stabile BE, Morrow DJ, Passaro E Jr (1984) The gastrinoma triangle: operative implications. Am J Surg 147(1):25–31

    Article  PubMed  CAS  Google Scholar 

  75. Winter TC 3rd, Freeny PC, Nghiem HV (1996) Extrapancreatic gastrinoma localization: value of arterial-phase helical CT with water as an oral contrast agent. AJR Am J Roentgenol 166(1):51–52

    PubMed  Google Scholar 

  76. Yu F, Venzon DJ, Serrano J, Goebel SU, Doppman JL, Gibril F, Jensen RT (1999) Prospective study of the clinical course, prognostic factors, causes of death, and survival in patients with long-standing Zollinger-Ellison syndrome. J Clin Oncol 17(2):615–630

    PubMed  CAS  Google Scholar 

  77. Weber HC, Venzon DJ, Lin JT, Fishbein VA, Orbuch M, Strader DB, Gibril F, Metz DC, Fraker DL, Norton JA et al (1995) Determinants of metastatic rate and survival in patients with Zollinger-Ellison syndrome: a prospective long-term study. Gastroenterology 108(6):1637–1649

    Article  PubMed  CAS  Google Scholar 

  78. Zhuang Z, Vortmeyer AO, Pack S, Huang S, Pham TA, Wang C, Park WS, Agarwal SK, Debelenko LV, Kester M, Guru SC, Manickam P, Olufemi SE, Yu F, Heppner C, Crabtree JS, Skarulis MC, Venzon DJ, Emmert-Buck MR, Spiegel AM, Chandrasekharappa SC, Collins FS, Burns AL, Marx SJ, Lubensky IA et al (1997) Somatic mutations of the MEN1 tumor suppressor gene in sporadic gastrinomas and insulinomas. Cancer Res 57(21):4682–4686

    PubMed  CAS  Google Scholar 

  79. Debelenko LV, Zhuang Z, Emmert-Buck MR, Chandrasekharappa SC, Manickam P, Guru SC, Marx SJ, Skarulis MC, Spiegel AM, Collins FS, Jensen RT, Liotta LA, Lubensky IA (1997) Allelic deletions on chromosome 11q13 in multiple endocrine neoplasia type 1-associated and sporadic gastrinomas and pancreatic endocrine tumors. Cancer Res 57(11):2238–2243

    PubMed  CAS  Google Scholar 

  80. Serrano J, Goebel SU, Peghini PL, Lubensky IA, Gibril F, Jensen RT (2000) Alterations in the p16INK4a/CDKN2A tumor suppressor gene in gastrinomas. J Clin Endocrinol Metab 85(11):4146–4156

    Article  PubMed  CAS  Google Scholar 

  81. Evers BM, Rady PL, Sandoval K, Arany I, Tyring SK, Sanchez RL, Nealon WH, Townsend CM Jr, Thompson JC (1994) Gastrinomas demonstrate amplification of the HER-2/neu proto-oncogene. Ann Surg 219(6):596–601, discussion 602–594

    Article  PubMed  CAS  Google Scholar 

  82. Peghini PL, Iwamoto M, Raffeld M, Chen YJ, Goebel SU, Serrano J, Jensen RT (2002) Overexpression of epidermal growth factor and hepatocyte growth factor receptors in a proportion of gastrinomas correlates with aggressive growth and lower curability. Clin Cancer Res 8(7):2273–2285

    PubMed  CAS  Google Scholar 

  83. Cappelli C, Agosti B, Braga M, Cumetti D, Gandossi E, Rizzoni D, Agabiti Rosei E (2004) Von Recklinghausen's neurofibromatosis associated with duodenal somatostatinoma. A case report and review of the literature. Minerva Endocrinol 29(1):19–24

    PubMed  CAS  Google Scholar 

  84. Hamy A, Heymann MF, Bodic J, Visset J, Le Borgne J, Leneel JC, Le Bodic MF (2001) Duodenal somatostatinoma. Anatomic/clinical study of 12 operated cases. Ann Chir 126(3):221–226

    Article  PubMed  CAS  Google Scholar 

  85. Garbrecht N, Anlauf M, Schmitt A, Henopp T, Sipos B, Raffel A, Eisenberger CF, Knoefel WT, Pavel M, Fottner C, Musholt TJ, Rinke A, Arnold R, Berndt U, Plockinger U, Wiedenmann B, Moch H, Heitz PU, Komminoth P, Perren A, Kloppel G (2008) Somatostatin-producing neuroendocrine tumors of the duodenum and pancreas: incidence, types, biological behavior, association with inherited syndromes, and functional activity. Endocr Relat Cancer 15(1):229–241. doi:10.1677/ERC-07-0157

    Article  PubMed  Google Scholar 

  86. Soga J, Yakuwa Y (1999) Somatostatinoma/inhibitory syndrome: a statistical evaluation of 173 reported cases as compared to other pancreatic endocrinomas. J Exp Clin Cancer Res 18(1):13–22

    PubMed  CAS  Google Scholar 

  87. Nesi G, Marcucci T, Rubio CA, Brandi ML, Tonelli F (2008) Somatostatinoma: clinico-pathological features of three cases and literature reviewed. J Gastroenterol Hepatol 23(4):521–526. doi:10.1111/j.1440-1746.2007.05053.x

    Article  PubMed  CAS  Google Scholar 

  88. Pernet C, Kluger N, Du-Thanh A, Guillon F, Dereure O, Bessis D, Guillot B (2010) Somatostatin-producing endocrine tumour of the duodenum associated with type 1 neurofibromatosis. Acta Derm Venereol 90(3):320–321. doi:10.2340/00015555-0844

    PubMed  Google Scholar 

  89. Marion-Audibert AM, Barel C, Gouysse G, Dumortier J, Pilleul F, Pourreyron C, Hervieu V, Poncet G, Lombard-Bohas C, Chayvialle JA, Partensky C, Scoazec JY (2003) Low microvessel density is an unfavorable histoprognostic factor in pancreatic endocrine tumors. Gastroenterology 125(4):1094–1104

    Article  PubMed  Google Scholar 

  90. Modlin IM, Kidd M, Pfragner R, Eick GN, Champaneria MC (2006) The functional characterization of normal and neoplastic human enterochromaffin cells. J Clin Endocrinol Metab 91(6):2340–2348. doi:10.1210/jc.2006-0110

    Article  PubMed  CAS  Google Scholar 

  91. Panzuto F, Nasoni S, Falconi M, Corleto VD, Capurso G, Cassetta S, Di Fonzo M, Tornatore V, Milione M, Angeletti S, Cattaruzza MS, Ziparo V, Bordi C, Pederzoli P, Delle Fave G (2005) Prognostic factors and survival in endocrine tumor patients: comparison between gastrointestinal and pancreatic localization. Endocr Relat Cancer 12(4):1083–1092. doi:10.1677/erc.1.01017

    Article  PubMed  Google Scholar 

  92. Heymann MF, Joubert M, Nemeth J, Franc B, Visset J, Hamy A, le Borgne J, le Neel JC, Murat A, Cordel S, le Bodic MF (2000) Prognostic and immunohistochemical validation of the capella classification of pancreatic neuroendocrine tumours: an analysis of 82 sporadic cases. Histopathology 36(5):421–432

    Article  PubMed  CAS  Google Scholar 

  93. La Rosa S, Sessa F, Capella C, Riva C, Leone BE, Klersy C, Rindi G, Solcia E (1996) Prognostic criteria in nonfunctioning pancreatic endocrine tumours. Virchows Arch 429(6):323–333

    Article  PubMed  Google Scholar 

  94. Brissova M, Fowler MJ, Nicholson WE, Chu A, Hirshberg B, Harlan DM, Powers AC (2005) Assessment of human pancreatic islet architecture and composition by laser scanning confocal microscopy. J Histochem Cytochem 53(9):1087–1097. doi:10.1369/jhc.5C6684.2005

    Article  PubMed  CAS  Google Scholar 

  95. Ehehalt F, Saeger HD, Schmidt CM, Grutzmann R (2009) Neuroendocrine tumors of the pancreas. Oncologist 14(5):456–467. doi:10.1634/theoncologist.2008-0259

    Article  PubMed  CAS  Google Scholar 

  96. Eriksson B, Oberg K, Skogseid B (1989) Neuroendocrine pancreatic tumors. Clinical findings in a prospective study of 84 patients. Acta Oncol 28(3):373–377

    Article  PubMed  CAS  Google Scholar 

  97. Finegood DT, Tobin BW, Lewis JT (1992) Dynamics of glycemic normalization following transplantation of incremental islet masses in streptozotocin-diabetic rats. Transplantation 53(5):1033–1037

    Article  PubMed  CAS  Google Scholar 

  98. Bouwens L, Braet F, Heimberg H (1995) Identification of rat pancreatic duct cells by their expression of cytokeratins 7, 19, and 20 in vivo and after isolation and culture. J Histochem Cytochem 43(3):245–253

    PubMed  CAS  Google Scholar 

  99. Bouwens L, Wang RN, De Blay E, Pipeleers DG, Kloppel G (1994) Cytokeratins as markers of ductal cell differentiation and islet neogenesis in the neonatal rat pancreas. Diabetes 43(11):1279–1283

    Article  PubMed  CAS  Google Scholar 

  100. Bell DA (1987) Cytologic features of islet-cell tumors. Acta Cytol 31(4):485–492

    PubMed  CAS  Google Scholar 

  101. Kloppel G, Heitz PU (1988) Pancreatic endocrine tumors. Pathol Res Pract 183(2):155–168

    PubMed  CAS  Google Scholar 

  102. Schaffalitzky De Muckadell OB, Aggestrup S, Stentoft P (1986) Flushing and plasma substance P concentration during infusion of synthetic substance P in normal man. Scand J Gastroenterol 21(4):498–502

    Article  PubMed  CAS  Google Scholar 

  103. Oberg K, Eriksson B (2005) Endocrine tumours of the pancreas. Best Pract Res Clin Gastroenterol 19(5):753–781. doi:10.1016/j.bpg.2005.06.002

    Article  PubMed  CAS  Google Scholar 

  104. Rindi G, Candusso ME, Solcia E (1999) Molecular aspects of the endocrine tumours of the pancreas and the gastrointestinal tract. Ital J Gastroenterol Hepatol 31(Suppl 2):S135–S138

    PubMed  Google Scholar 

  105. Yoshimoto K, Iwahana H, Fukuda A, Sano T, Katsuragi K, Kinoshita M, Saito S, Itakura M (1992) ras mutations in endocrine tumors: mutation detection by polymerase chain reaction-single strand conformation polymorphism. Jpn J Cancer Res 83(10):1057–1062

    PubMed  CAS  Google Scholar 

  106. Hu W, Feng Z, Modica I, Klimstra DS, Song L, Allen PJ, Brennan MF, Levine AJ, Tang LH (2010) Gene Amplifications in Well-Differentiated Pancreatic Neuroendocrine Tumors Inactivate the p53 Pathway. Genes Cancer 1(4):360–368. doi:10.1177/1947601910371979

    Article  PubMed  CAS  Google Scholar 

  107. Wang Q, Fang WH, Krupinski J, Kumar S, Slevin M, Kumar P (2008) Pax genes in embryogenesis and oncogenesis. J Cell Mol Med 12(6A):2281–2294

    Article  PubMed  CAS  Google Scholar 

  108. Long KB, Srivastava A, Hirsch MS, Hornick JL, PAX8 Expression in well-differentiated pancreatic endocrine tumors: correlation with clinicopathologic features and comparison with gastrointestinal and pulmonary carcinoid tumors. Am J Surg Pathol 34(5):723–729

  109. Speel EJ, Richter J, Moch H, Egenter C, Saremaslani P, Rutimann K, Zhao J, Barghorn A, Roth J, Heitz PU, Komminoth P (1999) Genetic differences in endocrine pancreatic tumor subtypes detected by comparative genomic hybridization. Am J Pathol 155(6):1787–1794

    Article  PubMed  CAS  Google Scholar 

  110. Speel EJ, Scheidweiler AF, Zhao J, Matter C, Saremaslani P, Roth J, Heitz PU, Komminoth P (2001) Genetic evidence for early divergence of small functioning and nonfunctioning endocrine pancreatic tumors: gain of 9Q34 is an early event in insulinomas. Cancer Res 61(13):5186–5192

    PubMed  CAS  Google Scholar 

  111. Rigaud G, Missiaglia E, Moore PS, Zamboni G, Falconi M, Talamini G, Pesci A, Baron A, Lissandrini D, Rindi G, Grigolato P, Pederzoli P, Scarpa A (2001) High resolution allelotype of nonfunctional pancreatic endocrine tumors: identification of two molecular subgroups with clinical implications. Cancer Res 61(1):285–292

    PubMed  CAS  Google Scholar 

  112. Zhao J, Moch H, Scheidweiler AF, Baer A, Schaffer AA, Speel EJ, Roth J, Heitz PU, Komminoth P (2001) Genomic imbalances in the progression of endocrine pancreatic tumors. Genes Chromosom Cancer 32(4):364–372. doi:10.1002/gcc.1201

    Article  PubMed  CAS  Google Scholar 

  113. Chung DC, Brown SB, Graeme-Cook F, Seto M, Warshaw AL, Jensen RT, Arnold A (2000) Overexpression of cyclin D1 occurs frequently in human pancreatic endocrine tumors. J Clin Endocrinol Metab 85(11):4373–4378

    Article  PubMed  CAS  Google Scholar 

  114. Chung DC, Brown SB, Graeme-Cook F, Tillotson LG, Warshaw AL, Jensen RT, Arnold A (1998) Localization of putative tumor suppressor loci by genome-wide allelotyping in human pancreatic endocrine tumors. Cancer Res 58(16):3706–3711

    PubMed  CAS  Google Scholar 

  115. Perren A, Komminoth P, Saremaslani P, Matter C, Feurer S, Lees JA, Heitz PU, Eng C (2000) Mutation and expression analyses reveal differential subcellular compartmentalization of PTEN in endocrine pancreatic tumors compared to normal islet cells. Am J Pathol 157(4):1097–1103

    Article  PubMed  CAS  Google Scholar 

  116. Missiaglia E, Dalai I, Barbi S, Beghelli S, Falconi M, della Peruta M, Piemonti L, Capurso G, Di Florio A, delle Fave G, Pederzoli P, Croce CM, Scarpa A (2010) Pancreatic endocrine tumors: expression profiling evidences a role for AKT-mTOR pathway. J Clin Oncol 28(2):245–255. doi:10.1200/JCO.2008.21.5988

    Article  PubMed  CAS  Google Scholar 

  117. D'Adda T, Bottarelli L, Azzoni C, Pizzi S, Bongiovanni M, Papotti M, Pelosi G, Maisonneuve P, Antonetti T, Rindi G, Bordi C (2005) Malignancy-associated X chromosome allelic losses in foregut endocrine neoplasms: further evidence from lung tumors. Mod Pathol 18(6):795–805. doi:10.1038/modpathol.3800353

    Article  PubMed  CAS  Google Scholar 

  118. Ghimenti C, Lonobile A, Campani D, Bevilacqua G, Caligo MA (1999) Microsatellite instability and allelic losses in neuroendocrine tumors of the gastro-entero-pancreatic system. Int J Oncol 15(2):361–366

    PubMed  CAS  Google Scholar 

  119. Arnold CN, Sosnowski A, Blum HE (2004) Analysis of molecular pathways in neuroendocrine cancers of the gastroenteropancreatic system. Ann NY Acad Sci 1014:218–219

    Article  PubMed  Google Scholar 

  120. Mallinson CN, Bloom SR, Warin AP, Salmon PR, Cox B (1974) A glucagonoma syndrome. Lancet 2(7871):1–5

    Article  PubMed  CAS  Google Scholar 

  121. Anlauf M, Schlenger R, Perren A, Bauersfeld J, Koch CA, Dralle H, Raffel A, Knoefel WT, Weihe E, Ruszniewski P, Couvelard A, Komminoth P, Heitz PU, Kloppel G (2006) Microadenomatosis of the endocrine pancreas in patients with and without the multiple endocrine neoplasia type 1 syndrome. Am J Surg Pathol 30(5):560–574. doi:10.1097/01.pas.0000194044.01104.25

    Article  PubMed  Google Scholar 

  122. Pellegata NS, Quintanilla-Martinez L, Siggelkow H, Samson E, Bink K, Hofler H, Fend F, Graw J, Atkinson MJ (2006) Germ-line mutations in p27Kip1 cause a multiple endocrine neoplasia syndrome in rats and humans. Proc Natl Acad Sci USA 103(42):15558–15563. doi:10.1073/pnas.0603877103

    Article  PubMed  CAS  Google Scholar 

  123. Henopp T, Anlauf M, Schmitt A, Schlenger R, Zalatnai A, Couvelard A, Ruszniewski P, Schaps KP, Jonkers YM, Speel EJ, Pellegata NS, Heitz PU, Komminoth P, Perren A, Kloppel G (2009) Glucagon cell adenomatosis: a newly recognized disease of the endocrine pancreas. J Clin Endocrinol Metab 94(1):213–217. doi:10.1210/jc.2008-1300

    Article  PubMed  CAS  Google Scholar 

  124. Yu R, Nissen NN, Dhall D, Heaney AP (2008) Nesidioblastosis and hyperplasia of alpha cells, microglucagonoma, and nonfunctioning islet cell tumor of the pancreas: review of the literature. Pancreas 36(4):428–431. doi:10.1097/MPA.0b013e31815ceb23

    Article  PubMed  CAS  Google Scholar 

  125. Stacpoole PW (1981) The glucagonoma syndrome: clinical features, diagnosis, and treatment. Endocr Rev 2(3):347–361

    Article  PubMed  CAS  Google Scholar 

  126. Wermers RA, Fatourechi V, Wynne AG, Kvols LK, Lloyd RV (1996) The glucagonoma syndrome. Clinical and pathologic features in 21 patients. Medicine (Baltimore) 75(2):53–63

    Article  CAS  Google Scholar 

  127. Bloom SR (1972) An enteroglucagon tumour. Gut 13(7):520–523

    Article  PubMed  CAS  Google Scholar 

  128. Gleeson MH, Bloom SR, Polak JM, Henry K, Dowling RH (1971) Endocrine tumour in kidney affecting small bowel structure, motility, and absorptive function. Gut 12(10):773–782

    Article  PubMed  CAS  Google Scholar 

  129. Mansour JC, Chen H (2004) Pancreatic endocrine tumors. J Surg Res 120(1):139–161. doi:10.1016/j.jss.2003.12.007

    Article  PubMed  CAS  Google Scholar 

  130. Soga J, Yakuwa Y (1998) Glucagonomas/diabetico-dermatogenic syndrome (DDS): a statistical evaluation of 407 reported cases. J Hepatobiliary Pancreat Surg 5(3):312–319

    Article  PubMed  CAS  Google Scholar 

  131. Perren A, Roth J, Muletta-Feurer S, Saremaslani P, Speel EJ, Heitz PU, Komminoth P (1998) Clonal analysis of sporadic pancreatic endocrine tumours. J Pathol 186(4):363–371. doi:10.1002/(SICI)1096-9896(199812)186:4<363::AID-PATH197>3.0.CO;2-W

    Article  PubMed  CAS  Google Scholar 

  132. Alexakis N, Neoptolemos JP (2008) Pancreatic neuroendocrine tumours. Best Pract Res Clin Gastroenterol 22(1):183–205. doi:10.1016/j.bpg.2007.10.008

    Article  PubMed  CAS  Google Scholar 

  133. Jensen RT (2006) Pancreatic neuroendocrine tumors: overview of recent advances and diagnosis. J Gastrointest Surg 10(3):324–326

    Article  PubMed  Google Scholar 

  134. Nikfarjam M, Warshaw AL, Axelrod L, Deshpande V, Thayer SP, Ferrone CR, Fernandez-del Castillo C (2008) Improved contemporary surgical management of insulinomas: a 25-year experience at the Massachusetts General Hospital. Ann Surg 247(1):165–172. doi:10.1097/SLA.0b013e31815792ed

    Article  PubMed  Google Scholar 

  135. Schindl M, Kaczirek K, Kaserer K, Niederle B (2000) Is the new classification of neuroendocrine pancreatic tumors of clinical help? World J Surg 24(11):1312–1318. doi:10.1007/s002680010217

    Article  PubMed  CAS  Google Scholar 

  136. Vaidakis D, Karoubalis J, Pappa T, Piaditis G, Zografos GN (2010) Pancreatic insulinoma: current issues and trends. Hepatobiliary Pancreat Dis Int 9(3):234–241

    PubMed  Google Scholar 

  137. Dong J, Asa SL, Drucker DJ (1991) Islet cell and extrapancreatic expression of the LIM domain homeobox gene isl-1. Mol Endocrinol 5(11):1633–1641

    Article  PubMed  CAS  Google Scholar 

  138. Schmitt AM, Riniker F, Anlauf M, Schmid S, Soltermann A, Moch H, Heitz PU, Kloppel G, Komminoth P, Perren A (2008) Islet 1 (Isl1) expression is a reliable marker for pancreatic endocrine tumors and their metastases. Am J Surg Pathol 32(3):420–425. doi:10.1097/PAS.0b013e318158a397

    Article  PubMed  Google Scholar 

  139. Vinik AI, Strodel WE, Eckhauser FE, Moattari AR, Lloyd R (1987) Somatostatinomas, PPomas, neurotensinomas. Semin Oncol 14(3):263–281

    PubMed  CAS  Google Scholar 

  140. Thompson GB, van Heerden JA, Grant CS, Carney JA, Ilstrup DM (1988) Islet cell carcinomas of the pancreas: a twenty-year experience. Surgery 104(6):1011–1017

    PubMed  CAS  Google Scholar 

  141. Mignon M (2000) Natural history of neuroendocrine enteropancreatic tumors. Digestion 62(Suppl 1):51–58

    Article  PubMed  Google Scholar 

  142. Maton PN, Gardner JD, Jensen RT (1986) Cushing's syndrome in patients with the Zollinger-Ellison syndrome. N Engl J Med 315(1):1–5. doi:10.1056/NEJM198607033150101

    Article  PubMed  CAS  Google Scholar 

  143. Sano T, Asa SL, Kovacs K (1988) Growth hormone-releasing hormone-producing tumors: clinical, biochemical, and morphological manifestations. Endocr Rev 9(3):357–373

    Article  PubMed  CAS  Google Scholar 

  144. Ahlman H (1985) Serotonin and carcinoid tumors. J Cardiovasc Pharmacol 7(Suppl 7):S79–S85

    Article  PubMed  CAS  Google Scholar 

  145. Nilsson O, Ericson LE, Dahlstrom A, Ekholm R, Steinbusch HW, Ahlman H (1985) Subcellular localization of serotonin immunoreactivity in rat enterochromaffin cells. Histochemistry 82(4):351–355

    Article  PubMed  CAS  Google Scholar 

  146. Heitz P, Polak JM, Timson DM, Pearse AG (1976) Enterochromaffin cells as the endocrine source of gastrointestinal substance P. Histochemistry 49(4):343–347

    Article  PubMed  CAS  Google Scholar 

  147. Pearse AG, Polak JM, Bloom SR, Adams C, Dryburgh JR, Brown JC (1974) Enterochromaffin cells of the mammalian small intestine as the source of motilin. Virchows Arch B Cell Pathol 16(2):111–120

    Article  PubMed  CAS  Google Scholar 

  148. Barter R, Pearse AG (1955) Mammalian enterochromaffin cells as the source of serotonin (5-hydroxytryptamine). J Pathol Bacteriol 69(1–2):25–31

    Article  PubMed  CAS  Google Scholar 

  149. Cetin Y, Kuhn M, Kulaksiz H, Adermann K, Bargsten G, Grube D, Forssmann WG (1994) Enterochromaffin cells of the digestive system: cellular source of guanylin, a guanylate cyclase-activating peptide. Proc Natl Acad Sci USA 91(8):2935–2939

    Article  PubMed  CAS  Google Scholar 

  150. Modlin IM, Kidd M, Latich I, Zikusoka MN, Shapiro MD (2005) Current status of gastrointestinal carcinoids. Gastroenterology 128(6):1717–1751

    Article  PubMed  Google Scholar 

  151. van der Horst-Schrivers AN, Wymenga AN, Links TP, Willemse PH, Kema IP, de Vries EG (2004) Complications of midgut carcinoid tumors and carcinoid syndrome. Neuroendocrinology 80(Suppl 1):28–32

    Article  PubMed  CAS  Google Scholar 

  152. Kunnimalaiyaan M, Chen H (2007) Tumor suppressor role of Notch-1 signaling in neuroendocrine tumors. Oncologist 12(5):535–542. doi:10.1634/theoncologist.12-5-535

    Article  PubMed  CAS  Google Scholar 

  153. Yantiss RK, Odze RD, Farraye FA, Rosenberg AE (2003) Solitary versus multiple carcinoid tumors of the ileum: a clinical and pathologic review of 68 cases. Am J Surg Pathol 27(6):811–817

    Article  PubMed  Google Scholar 

  154. Hodges JR, Isaacson P, Wright R (1981) Diffuse enterochromaffin-like (ECL) cell hyperplasia and multiple gastric carcinoids: a complication of pernicious anaemia. Gut 22(3):237–241

    Article  PubMed  CAS  Google Scholar 

  155. Moyana TN, Satkunam N (1992) A comparative immunohistochemical study of jejunoileal and appendiceal carcinoids. Implications for histogenesis and pathogenesis. Cancer 70(5):1081–1088

    Article  PubMed  CAS  Google Scholar 

  156. Slade MJ, Smith BM, Sinnett HD, Cross NC, Coombes RC (1999) Quantitative polymerase chain reaction for the detection of micrometastases in patients with breast cancer. J Clin Oncol 17(3):870–879

    PubMed  CAS  Google Scholar 

  157. Modlin I, Sandor A (1997) An analysis of 8305 cases of carcinoid tumors. Cancer 79:813–829

    Article  PubMed  CAS  Google Scholar 

  158. Modlin I, Lye K, Kidd M (2003) A 5-decade analysis of 13, 715 carcinoid tumors. Cancer 97(4):934–959

    Article  PubMed  Google Scholar 

  159. Saha S, Hoda S, Godfrey R, Sutherland C, Raybon K (1989) Carcinoid tumors of the gastrointestinal tract: a 44-year experience. South Med J 82(12):1501–1505

    PubMed  CAS  Google Scholar 

  160. Lawrence B, Gustafsson B, Chan A, Svejda B, Kidd M, Modlin I (2010) The epidemiology of gastroenteropancreatic tumors. Endocrinol Metab Clin N Am (in press)

  161. Burke AP, Thomas RM, Elsayed AM, Sobin LH (1997) Carcinoids of the jejunum and ileum: an immunohistochemical and clinicopathologic study of 167 cases. Cancer 79(6):1086–1093

    Article  PubMed  CAS  Google Scholar 

  162. Johnson LA, Lavin P, Moertel CG, Weiland L, Dayal Y, Doos WG, Geller SA, Cooper HS, Nime F, Masse S, Simson IW, Sumner H, Folsch E, Engstrom P (1983) Carcinoids: the association of histologic growth pattern and survival. Cancer 51(5):882–889

    Article  PubMed  CAS  Google Scholar 

  163. Tonnies H, Toliat MR, Ramel C, Pape UF, Neitzel H, Berger W, Wiedenmann B (2001) Analysis of sporadic neuroendocrine tumours of the enteropancreatic system by comparative genomic hybridisation. Gut 48(4):536–541

    Article  PubMed  CAS  Google Scholar 

  164. Kytola S, Hoog A, Nord B, Cedermark B, Frisk T, Larsson C, Kjellman M (2001) Comparative genomic hybridization identifies loss of 18q22-qter as an early and specific event in tumorigenesis of midgut carcinoids. Am J Pathol 158(5):1803–1808

    Article  PubMed  CAS  Google Scholar 

  165. Kytola S, Nord B, Elder EE, Carling T, Kjellman M, Cedermark B, Juhlin C, Hoog A, Isola J, Larsson C (2002) Alterations of the SDHD gene locus in midgut carcinoids, Merkel cell carcinomas, pheochromocytomas, and abdominal paragangliomas. Genes Chromosom Cancer 34(3):325–332. doi:10.1002/gcc.10081

    Article  PubMed  CAS  Google Scholar 

  166. Lollgen RM, Hessman O, Szabo E, Westin G, Akerstrom G (2001) Chromosome 18 deletions are common events in classical midgut carcinoid tumors. Int J Cancer 92(6):812–815. doi:10.1002/ijc.127610.1002/ijc.1276

    Article  PubMed  CAS  Google Scholar 

  167. Andersson E, Sward C, Stenman G, Ahlman H, Nilsson O (2009) High-resolution genomic profiling reveals gain of chromosome 14 as a predictor of poor outcome in ileal carcinoids. Endocr Relat Cancer 16(3):953–966. doi:10.1677/ERC-09-0052

    Article  PubMed  Google Scholar 

  168. Kulke MH, Freed E, Chiang DY, Philips J, Zahrieh D, Glickman JN, Shivdasani RA (2008) High-resolution analysis of genetic alterations in small bowel carcinoid tumors reveals areas of recurrent amplification and loss. Genes Chromosom Cancer 47(7):591–603. doi:10.1002/gcc.20561

    Article  PubMed  CAS  Google Scholar 

  169. Fearon ER, Pierceall WE (1995) The deleted in colorectal cancer (DCC) gene: a candidate tumour suppressor gene encoding a cell surface protein with similarity to neural cell adhesion molecules. Cancer Surv 24:3–17

    PubMed  CAS  Google Scholar 

  170. Petzmann S, Ullmann R, Halbwedl I, Popper HH (2004) Analysis of chromosome-11 aberrations in pulmonary and gastrointestinal carcinoids: an array comparative genomic hybridization-based study. Virchows Arch 445(2):151–159. doi:10.1007/s00428-004-1052-y

    Article  PubMed  CAS  Google Scholar 

  171. Walsh KM, Choi M, Oberg KE, Kulke MH, Yao JC, Wu C, Jurkiewicz M, Hsu LI, Hooshmand SM, Hassan M, Janson ET, Cunningham J, Vosburgh E, Sackler RS, Lifton RP, Dewan AT, Hoh J (2010) A pilot genome-wide association study shows genomic variants enriched in the non-tumor cells of patients with well-differentiated neuroendocrine tumors of the ileum. Endocr Relat Cancer. doi:10.1677/ERC-10-0248

    Google Scholar 

  172. Kidd M, Eick G, Shapiro MD, Camp RL, Mane SM, Modlin IM (2005) Microsatellite instability and gene mutations in transforming growth factor-beta type II receptor are absent in small bowel carcinoid tumors. Cancer 103(2):229–236. doi:10.1002/cncr.20750

    Article  PubMed  CAS  Google Scholar 

  173. Muneyuki T, Watanabe M, Yamanaka M, Isaji S, Kawarada Y, Yatani R (2000) Combination analysis of genetic alterations and cell proliferation in small intestinal carcinomas. Dig Dis Sci 45(10):2022–2028

    Article  PubMed  CAS  Google Scholar 

  174. Planck M, Ericson K, Piotrowska Z, Halvarsson B, Rambech E, Nilbert M (2003) Microsatellite instability and expression of MLH1 and MSH2 in carcinomas of the small intestine. Cancer 97(6):1551–1557. doi:10.1002/cncr.11197

    Article  PubMed  CAS  Google Scholar 

  175. Kidd M, Modlin IM, Mane SM, Camp RL, Eick G, Latich I (2006) The role of genetic markers—NAP1L1, MAGE-D2, and MTA1—in defining small-intestinal carcinoid neoplasia. Ann Surg Oncol 13(2):253–262. doi:10.1245/ASO.2006.12.011

    Article  PubMed  Google Scholar 

  176. Drozdov I, Kidd M, Nadler B, Camp RL, Mane SM, Hauso O, Gustafsson BI, Modlin IM (2009) Predicting neuroendocrine tumor (carcinoid) neoplasia using gene expression profiling and supervised machine learning. Cancer 115(8):1638–1650. doi:10.1002/cncr.24180

    Article  PubMed  CAS  Google Scholar 

  177. Leja J, Essaghir A, Essand M, Wester K, Oberg K, Totterman TH, Lloyd R, Vasmatzis G, Demoulin JB, Giandomenico V (2009) Novel markers for enterochromaffin cells and gastrointestinal neuroendocrine carcinomas. Mod Pathol 22(2):261–272. doi:10.1038/modpathol.2008.174

    Article  PubMed  CAS  Google Scholar 

  178. Hofer MD, Tapia C, Browne TJ, Mirlacher M, Sauter G, Rubin MA (2006) Comprehensive analysis of the expression of the metastasis-associated gene 1 in human neoplastic tissue. Arch Pathol Lab Med 130(7):989–996

    PubMed  CAS  Google Scholar 

  179. Ruebel K, Leontovich AA, Stilling GA, Zhang S, Righi A, Jin L, Lloyd RV (2010) MicroRNA expression in ileal carcinoid tumors: downregulation of microRNA-133a with tumor progression. Mod Pathol 23(3):367–375. doi:10.1038/modpathol.2009.161

    Article  PubMed  CAS  Google Scholar 

  180. Hemminki K, Li X (2001) Familial carcinoid tumors and subsequent cancers: a nation-wide epidemiologic study from Sweden. Int J Cancer 94(3):444–448

    Article  PubMed  CAS  Google Scholar 

  181. Hassan MM, Phan A, Li D, Dagohoy CG, Leary C, Yao JC (2008) Family history of cancer and associated risk of developing neuroendocrine tumors: a case-control study. Cancer Epidemiol Biomarkers Prev 17(4):959–965

    Article  PubMed  Google Scholar 

  182. Jarhult J, Landerholm K, Falkmer S, Nordenskjold M, Sundler F, Wierup N, First report on metastasizing small bowel carcinoids in first-degree relatives in three generations. Neuroendocrinology 91(4):318–323

  183. Zikusoka MN, Kidd M, Eick G, Latich I, Modlin IM (2005) The molecular genetics of gastroenteropancreatic neuroendocrine tumors. Cancer 104(11):2292–2309. doi:10.1002/cncr.21451

    Article  PubMed  CAS  Google Scholar 

  184. Kidd M, Modlin IM, Pfragner R, Eick GN, Champaneria MC, Chan AK, Camp RL, Mane SM (2007) Small bowel carcinoid (enterochromaffin cell) neoplasia exhibits transforming growth factor-beta1-mediated regulatory abnormalities including up-regulation of C-Myc and MTA1. Cancer 109(12):2420–2431

    Article  PubMed  CAS  Google Scholar 

  185. Vogelsang H, Siewert JR (2005) Endocrine tumours of the hindgut. Best Pract Res Clin Gastroenterol 19(5):739–751. doi:10.1016/j.bpg.2005.06.001

    Article  PubMed  CAS  Google Scholar 

  186. Ramage JK, Goretzki PE, Manfredi R, Komminoth P, Ferone D, Hyrdel R, Kaltsas G, Kelestimur F, Kvols L, Scoazec JY, Garcia MI, Caplin ME (2008) Consensus guidelines for the management of patients with digestive neuroendocrine tumours: well-differentiated colon and rectum tumour/carcinoma. Neuroendocrinology 87(1):31–39. doi:10.1159/000111036

    Article  PubMed  CAS  Google Scholar 

  187. Federspiel BH, Burke AP, Sobin LH, Shekitka KM (1990) Rectal and colonic carcinoids. A clinicopathologic study of 84 cases. Cancer 65(1):135–140

    Article  PubMed  CAS  Google Scholar 

  188. Kimura N, Pilichowska M, Okamoto H, Kimura I, Aunis D (2000) Immunohistochemical expression of chromogranins A and B, prohormone convertases 2 and 3, and amidating enzyme in carcinoid tumors and pancreatic endocrine tumors. Mod Pathol 13(2):140–146. doi:10.1038/modpathol.3880026

    Article  PubMed  CAS  Google Scholar 

  189. Yao JC, Phan AT, Chang DZ, Wolff RA, Hess K, Gupta S, Jacobs C, Mares JE, Landgraf AN, Rashid A, Meric-Bernstam F (2008) Efficacy of RAD001 (everolimus) and octreotide LAR in advanced low- to intermediate-grade neuroendocrine tumors: results of a phase II study. J Clin Oncol 26(26):4311–4318. doi:10.1200/JCO.2008.16.7858

    Article  PubMed  Google Scholar 

  190. Modlin IM, Lye KD, Kidd M (2003) A 5-decade analysis of 13, 715 carcinoid tumors. Cancer 97(4):934–959. doi:10.1002/cncr.11105

    Article  PubMed  Google Scholar 

  191. DiSario JA, Burt RW, Kendrick ML, McWhorter WP (1994) Colorectal cancers of rare histologic types compared with adenocarcinomas. Dis Colon Rectum 37(12):1277–1280

    Article  PubMed  CAS  Google Scholar 

  192. Saclarides TJ, Szeluga D, Staren ED (1994) Neuroendocrine cancers of the colon and rectum. Results of a ten-year experience. Dis Colon Rectum 37(7):635–642

    Article  PubMed  CAS  Google Scholar 

  193. Mani S, Modlin IM, Ballantyne G, Ahlman H, West B (1994) Carcinoids of the rectum. J Am Coll Surg 179(2):231–248

    PubMed  CAS  Google Scholar 

  194. Naunheim KS, Zeitels J, Kaplan EL, Sugimoto J, Shen KL, Lee CH, Straus FH 2nd (1983) Rectal carcinoid tumors—treatment and prognosis. Surgery 94(4):670–676

    PubMed  CAS  Google Scholar 

  195. Modlin IM, Sandor A (1997) An analysis of 8305 cases of carcinoid tumors. Cancer 79(4):813–829. doi:10.1002/(SICI)1097-0142(19970215)79:4<813::AID-CNCR19>3.0.CO;2-2

    Article  PubMed  CAS  Google Scholar 

  196. Grabowski P, Schonfelder J, Ahnert-Hilger G, Foss HD, Stein H, Berger G, Zeitz M, Scherubl H (2004) Heterogeneous expression of neuroendocrine marker proteins in human undifferentiated carcinoma of the colon and rectum. Ann NY Acad Sci 1014:270–274

    Article  PubMed  CAS  Google Scholar 

  197. Eriksson B, Oberg K, Stridsberg M (2000) Tumor markers in neuroendocrine tumors. Digestion 62(Suppl 1):33–38

    Article  PubMed  CAS  Google Scholar 

  198. O'Connor DT, Deftos LJ (1986) Secretion of chromogranin A by peptide-producing endocrine neoplasms. N Engl J Med 314(18):1145–1151. doi:10.1056/NEJM198605013141803

    Article  PubMed  Google Scholar 

  199. Norheim I, Oberg K, Theodorsson-Norheim E, Lindgren PG, Lundqvist G, Magnusson A, Wide L, Wilander E (1987) Malignant carcinoid tumors. An analysis of 103 patients with regard to tumor localization, hormone production, and survival. Ann Surg 206(2):115–125

    Article  PubMed  CAS  Google Scholar 

  200. Hotta K, Shimoda T, Nakanishi Y, Saito D (2006) Usefulness of Ki-67 for predicting the metastatic potential of rectal carcinoids. Pathol Int 56(10):591–596. doi:10.1111/j.1440-1827.2006.02013.x

    Article  PubMed  Google Scholar 

  201. Bosman F, Carneiro F, Hruban RH, Theise ND (eds) (2010) WHO classification of tumours of the digestive system. WHO

Download references

Conflicts of interest

None.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Irvin M. Modlin.

Additional information

Supported by NIH: DK080871

Rights and permissions

Reprints and permissions

About this article

Cite this article

Schimmack, S., Svejda, B., Lawrence, B. et al. The diversity and commonalities of gastroenteropancreatic neuroendocrine tumors. Langenbecks Arch Surg 396, 273–298 (2011). https://doi.org/10.1007/s00423-011-0739-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00423-011-0739-1

Keywords

Navigation