Skip to main content

Advertisement

Log in

Gastroenteropancreatic neuroendocrine tumors: new insights in the diagnosis and therapy

  • Review
  • Published:
Endocrine Aims and scope Submit manuscript

Abstract

Gastroenteropancreatic neuroendocrine tumors (GEP-NETs) are relatively rare and heterogenous malignancies. Recent advances in histopathological classification according to the anatomical site of origin, proliferation rate, and extend of the disease have created a valid and powerful tool for the prognostic stratification of GEP-NETs. Chromogranin A is still the best available marker used for the biochemical confirmation of these tumors, but new more sensitive markers are urgently required. Although scintigraphy with 111In-octreotide has widely been applied for the localization and staging of GEP-NETs, newer imaging modalities based on the functional characteristics of these tumors are evolving aiming not only to facilitate the diagnosis but also prognosis and evaluation of treatment. Somatostatin receptors are the primary therapeutic targets through somatostatin analogs and peptide receptor radionuclide therapy (PRRT) producing symptomatic, biochemical and to a lesser extent antiproliferative effects. Due to the relatively limited and erratic response to chemotherapy, new molecular targeted therapies exploiting some of the biological properties of GEP-NETs such as increased vascularity and inhibition of pathways involved in downstream signal transduction have evolved. Some of these therapies, the mTOR inhibitor everolimus and the tyrosine kinase inhibitor sunitinib, have been recently validated in phase III studies producing practice changing outcomes. In addition, two oral chemotherapeutic agents temozolomide and capecitabine, show promising effects and may replace streptozotocin-based regimens whereas combination therapies with the angiogenesis inhibitor bevacizumab are being investigated. Although progression free survival is used as a feasible primary end point due to the long survival of patients even in the presence of extensive disease prolongation of overall survival following the introduction of new therapies needs to be established.

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.

Fig. 1

Similar content being viewed by others

Reference

  1. I.M. Modlin, M. Kidd, R. Pfragner, G.N. Eick, M.C. Champaneria, The functional characterization of normal and neoplastic human enterochromaffin cells. J. Clin. Endocrinol. Metab. 91, 2340–2348 (2006)

    Article  PubMed  CAS  Google Scholar 

  2. K. Oberg, Neuroendocrine gastrointestinal Tumors–a condensed overview of diagnosis and treatment. Ann. Oncol. 10(Suppl 2), S3–S8 (1999)

    Article  PubMed  Google Scholar 

  3. G.A. Kaltsas, G.M. Besser, A.B. Grossman, The diagnosis and medical management of advanced neuroendocrine tumors. Endocr. Rev. 25, 458–511 (2004)

    Article  PubMed  CAS  Google Scholar 

  4. J.C. Yao, M. Hassan, A. Phan, C. Dagohoy, C. Leary, J.E. Mares, E.K. Abdalla, J.B. Fleming, J.N. Vauthey, A. Rashid, D.B. Evans, One hundred years after carcinoid: epidemiology of and prognostic factors for neuroendocrine tumors in 35, 825 cases in the United States. J. Clin. Oncol. 26, 3063–3072 (2008)

    Article  PubMed  Google Scholar 

  5. M.B. Niederle, M. Hackl, K. Kaserer, B. Niederle, Gastroenteropancreatic neuroendocrine tumours: the current incidence and staging based on the WHO and European Neuroendocrine Tumour Society classification: an analysis based on prospectively collected parameters. Endocr. Relat. Cancer. 17, 909–918 (2010)

    Article  PubMed  Google Scholar 

  6. A. Sundin, M.P. Vullierme, G. Kaltsas, U. Plockinger, ENETS consensus guidelines for the standards of care in neuroendocrine tumors: radiological examinations. Neuroendocrinology. 90, 167–183 (2009)

    Article  PubMed  CAS  Google Scholar 

  7. I.M. Modlin, I. Latich, M. Kidd, M. Zikusoka, G. Eick, Therapeutic options for gastrointestinal carcinoids. Clin. Gastroenterol. Hepatol. 4, 526–547 (2006)

    Article  PubMed  CAS  Google Scholar 

  8. I.M. Modlin, K. Oberg, D.C. Chung, R.T. Jensen, W.W. de Herder, R.V. Thakker, M. Caplin, F.G. Delle, G.A. Kaltsas, E.P. Krenning, S.F. Moss, O. Nilsson, G. Rindi, R. Salazar, P. Ruszniewski, A. Sundin, Gastroenteropancreatic neuroendocrine tumours. Lancet Oncol. 9, 61–72 (2008)

    Article  PubMed  CAS  Google Scholar 

  9. H. Ahlman, O. Nilsson, A.M. McNicol, P. Ruszniewski, B. Niederle, J. Ricke, R. Jensen, B. Kos-Kudla, K. Oberg, J.M. O’Connor, M.E. Pavel, M.P. Vullierme, Poorly-differentiated endocrine carcinomas of midgut and hindgut origin. Neuroendocrinology. 87, 40–46 (2008)

    Article  PubMed  CAS  Google Scholar 

  10. O. Nilsson, C.E. Van, F.G. Delle, J.C. Yao, M.E. Pavel, A.M. McNicol, M.I. Sevilla Garcia, W.H. Knapp, F. Kelestimur, A. Sauvanet, S. Pauwels, D.J. Kwekkeboom, M. Caplin, Poorly differentiated carcinomas of the foregut (gastric, duodenal and pancreatic). Neuroendocrinology. 84, 212–215 (2006)

    Article  PubMed  CAS  Google Scholar 

  11. N. Ballian, A.G. Loeffler, V. Rajamanickam, P.A. Norstedt, S.M. Weber, C.S. Cho, A simplified prognostic system for resected pancreatic neuroendocrine neoplasms. HPB (Oxford). 11, 422–428 (2009)

    Article  Google Scholar 

  12. A. Couvelard, D. O’Toole, H. Turley, R. Leek, A. Sauvanet, C. Degott, P. Ruszniewski, J. Belghiti, A.L. Harris, K. Gatter, F. Pezzella, Microvascular density and hypoxia-inducible factor pathway in pancreatic endocrine tumours: negative correlation of microvascular density and VEGF expression with tumour progression. Br. J. Cancer. 92, 94–101 (2005)

    Article  PubMed  CAS  Google Scholar 

  13. A. Rinke, H.H. Muller, C. Schade-Brittinger, K.J. Klose, P. Barth, M. Wied, C. Mayer, B. Aminossadati, U.F. Pape, M. Blaker, J. Harder, C. Arnold, T. Gress, R. Arnold, Placebo-controlled, double-blind, prospective, randomized study on the effect of octreotide LAR in the control of tumor growth in patients with metastatic neuroendocrine midgut tumors: a report from the PROMID Study Group. J. Clin. Oncol. 27, 4656–4663 (2009)

    Article  PubMed  CAS  Google Scholar 

  14. C.G. Moertel, M. Lefkopoulo, S. Lipsitz, R.G. Hahn, D. Klaassen, Streptozocin-doxorubicin, streptozocin-fluorouracil or chlorozotocin in the treatment of advanced islet-cell carcinoma. N. Engl. J. Med. 326, 519–523 (1992)

    Article  PubMed  CAS  Google Scholar 

  15. C.G. Moertel, L.K. Kvols, M.J. O’Connell, J. Rubin, Treatment of neuroendocrine carcinomas with combined etoposide and cisplatin. Evidence of major therapeutic activity in the anaplastic variants of these neoplasms. Cancer. 68, 227–232 (1991)

    Article  PubMed  CAS  Google Scholar 

  16. M.A. Kouvaraki, J.A. Ajani, P. Hoff, R. Wolff, D.B. Evans, R. Lozano, J.C. Yao, Fluorouracil, doxorubicin, and streptozocin in the treatment of patients with locally advanced and metastatic pancreatic endocrine carcinomas. J. Clin. Oncol. 22, 4762–4771 (2004)

    Article  PubMed  CAS  Google Scholar 

  17. E. Vilar, R. Salazar, J. Perez-Garcia, J. Cortes, K. Oberg, J. Tabernero, Chemotherapy and role of the proliferation marker Ki-67 in digestive neuroendocrine tumors. Endocr. Relat. Cancer. 14, 221–232 (2007)

    Article  PubMed  CAS  Google Scholar 

  18. M.H. Kulke, L.L. Siu, J.E. Tepper, G. Fisher, D. Jaffe, D.G. Haller, L.M. Ellis, J.K. Benedetti, E.K. Bergsland, T.J. Hobday, C.E. Van, J. Pingpank, K. Oberg, S.J. Cohen, M.C. Posner, J.C. Yao, Future directions in the treatment of neuroendocrine tumors: consensus report of the National Cancer Institute Neuroendocrine Tumor clinical trials planning meeting. J. Clin. Oncol. 29, 934–943 (2011)

    Article  PubMed  CAS  Google Scholar 

  19. M.H. Kulke, J. Bendell, L. Kvols, J. Picus, R. Pommier, J. Yao, Evolving diagnostic and treatment strategies for pancreatic neuroendocrine tumors. J. Hematol. Oncol. 4, 29 (2011)

    Article  PubMed  Google Scholar 

  20. M.H. Kulke, L.B. Anthony, D.L. Bushnell, W.W. de Herder, S.J. Goldsmith, D.S. Klimstra, S.J. Marx, J.L. Pasieka, R.F. Pommier, J.C. Yao, R.T. Jensen, NANETS treatment guidelines: well-differentiated neuroendocrine tumors of the stomach and pancreas. Pancreas. 39, 735–752 (2010)

    Article  PubMed  Google Scholar 

  21. J.R. Strosberg, D. Coppola, D.S. Klimstra, A.T. Phan, M.H. Kulke, G.A. Wiseman, L.K. Kvols, The NANETS consensus guidelines for the diagnosis and management of poorly differentiated (high-grade) extrapulmonary neuroendocrine carcinomas. Pancreas. 39, 799–800 (2010)

    Article  PubMed  Google Scholar 

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

    Article  PubMed  Google Scholar 

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

    Article  PubMed  Google Scholar 

  24. G. Rindi, W.W. de Herder, D. O’Toole, B. Wiedenmann, Consensus guidelines for the management of patients with digestive neuroendocrine tumors: why such guidelines and how we went about It. Neuroendocrinology. 84, 155–157 (2006)

    Article  PubMed  CAS  Google Scholar 

  25. G. Rindi, G. Kloppel, H. Alhman, M. Caplin, A. Couvelard, W.W. de Herder, B. Erikssson, A. Falchetti, M. Falconi, P. Komminoth, M. Korner, J.M. Lopes, A.M. McNicol, O. Nilsson, A. Perren, A. Scarpa, J.Y. Scoazec, B. Wiedenmann, TNM staging of foregut (neuro)endocrine tumors: a consensus proposal including a grading system. Virchows Arch. 449, 395–401 (2006)

    Article  PubMed  CAS  Google Scholar 

  26. G. Rindi, G. Kloppel, A. Couvelard, P. Komminoth, M. Korner, J.M. Lopes, A.M. McNicol, O. Nilsson, A. Perren, A. Scarpa, J.Y. Scoazec, B. Wiedenmann, TNM staging of midgut and hindgut (neuro) endocrine tumors: a consensus proposal including a grading system. Virchows Arch. 451, 757–762 (2007)

    Article  PubMed  CAS  Google Scholar 

  27. B. Zarebczan, H. Chen, Signaling mechanisms in neuroendocrine tumors as targets for therapy. Endocrinol. Metab. Clin. North Am. 39, 801–810 (2010)

    Article  PubMed  CAS  Google Scholar 

  28. P.D. Leotlela, A. Jauch, H. Holtgreve-Grez, R.V. Thakker, Genetics of neuroendocrine and carcinoid tumours. Endocr. Relat. Cancer. 10, 437–450 (2003)

    Article  PubMed  CAS  Google Scholar 

  29. M.H. Kulke, E. Freed, D.Y. Chiang, J. Philips, D. Zahrieh, J.N. Glickman, R.A. Shivdasani, High-resolution analysis of genetic alterations in small bowel carcinoid tumors reveals areas of recurrent amplification and loss. Genes. Chromosomes Cancer. 47, 591–603 (2008)

    Article  PubMed  CAS  Google Scholar 

  30. D.H. Kim, Y. Nagano, I.S. Choi, J.A. White, J.C. Yao, A. Rashid, Allelic alterations in well-differentiated neuroendocrine tumors (carcinoid tumors) identified by genome-wide single nucleotide polymorphism analysis and comparison with pancreatic endocrine tumors. Genes Chromosomes Cancer. 47, 84–92 (2008)

    Article  CAS  Google Scholar 

  31. S. Faivre, M.P. Sablin, C. Dreyer, E. Raymond, Novel anticancer agents in clinical trials for well-differentiated neuroendocrine tumors. Endocrinol. Metab. Clin. North Am. 39, 811–826 (2010)

    Article  PubMed  CAS  Google Scholar 

  32. O. Casanovas, D.J. Hicklin, G. Bergers, D. Hanahan, Drug resistance by evasion of antiangiogenic targeting of VEGF signaling in late-stage pancreatic islet tumors. Cancer Cell. 8, 299–309 (2005)

    Article  PubMed  CAS  Google Scholar 

  33. M. Inoue, J.H. Hager, N. Ferrara, H.P. Gerber, D. Hanahan, VEGF-A has a critical, nonredundant role in angiogenic switching and pancreatic beta cell carcinogenesis. Cancer Cell. 1, 193–202 (2002)

    Article  PubMed  CAS  Google Scholar 

  34. B. Eriksson, New drugs in neuroendocrine tumors: rising of new therapeutic philosophies? Curr. Opin. Oncol. 22, 381–386 (2010)

    Article  PubMed  CAS  Google Scholar 

  35. E. Raymond, L. Dahan, J.L. Raoul, Y.J. Bang, I. Borbath, C. Lombard-Bohas, J. Valle, P. Metrakos, D. Smith, A. Vinik, J.S. Chen, D. Horsch, P. Hammel, B. Wiedenmann, C.E. Van, S. Patyna, D.R. Lu, C. Blanckmeister, R. Chao, P. Ruszniewski, Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. N. Engl. J. Med. 364, 501–513 (2011)

    Article  PubMed  CAS  Google Scholar 

  36. J.C. Yao, M.H. Shah, T. Ito, C.L. Bohas, E.M. Wolin, C.E. Van, T.J. Hobday, T. Okusaka, J. Capdevila, E.G. de Vries, P. Tomassetti, M.E. Pavel, S. Hoosen, T. Haas, J. Lincy, D. Lebwohl, K. Oberg, Everolimus for advanced pancreatic neuroendocrine tumors. N. Engl. J. Med. 364, 514–523 (2011)

    Article  PubMed  CAS  Google Scholar 

  37. G.W. Krystal, G. Sulanke, J. Litz, Inhibition of phosphatidylinositol 3-kinase-Akt signaling blocks growth, promotes apoptosis, and enhances sensitivity of small cell lung cancer cells to chemotherapy. Mol. Cancer Ther. 1, 913–922 (2002)

    PubMed  CAS  Google Scholar 

  38. S.C. Pitt, H. Chen, M. Kunnimalaiyaan, Inhibition of phosphatidylinositol 3-kinase/Akt signaling suppresses tumor cell proliferation and neuroendocrine marker expression in GI carcinoid tumors. Ann. Surg. Oncol. 16, 2936–2942 (2009)

    Article  PubMed  Google Scholar 

  39. R.K. Ravi, E. Weber, M. McMahon, J.R. Williams, S. Baylin, A. Mal, M.L. Harter, L.E. Dillehay, P.P. Claudio, A. Giordano, B.D. Nelkin, M. Mabry, Activated Raf-1 causes growth arrest in human small cell lung cancer cells. J. Clin. Invest. 101, 153–159 (1998)

    Article  PubMed  CAS  Google Scholar 

  40. M.R. Cook, S.N. Pinchot, R. Jaskula-Sztul, J. Luo, M. Kunnimalaiyaan, H. Chen, Identification of a novel Raf-1 pathway activator that inhibits gastrointestinal carcinoid cell growth. Mol. Cancer Ther. 9, 429–437 (2010)

    Article  PubMed  CAS  Google Scholar 

  41. B. Zarebczan, S.N. Pinchot, M. Kunnimalaiyaan, H. Chen, Hesperetin, a potential therapy for carcinoid cancer. Am. J. Surg. 201, 329–332 (2011)

    Article  PubMed  CAS  Google Scholar 

  42. G. Kaltsas, I.I. Androulakis, W.W. de Herder, A.B. Grossman, Paraneoplastic syndromes secondary to neuroendocrine tumours. Endocr. Relat. Cancer. 17, R173–R193 (2010)

    Article  PubMed  Google Scholar 

  43. S. Welin, M. Stridsberg, J. Cunningham, D. Granberg, B. Skogseid, K. Oberg, B. Eriksson, E.T. Janson, Elevated plasma chromogranin A is the first indication of recurrence in radically operated midgut carcinoid tumors. Neuroendocrinology. 89, 302–307 (2009)

    Article  PubMed  CAS  Google Scholar 

  44. S. Bhattacharyya, C. Toumpanakis, M.E. Caplin, J. Davar, Usefulness of N-terminal pro-brain natriuretic peptide as a biomarker of the presence of carcinoid heart disease. Am. J. Cardiol. 102, 938–942 (2008)

    Article  PubMed  CAS  Google Scholar 

  45. J.L. Cunningham, E.T. Janson, S. Agarwal, L. Grimelius, M. Stridsberg, Tachykinins in endocrine tumors and the carcinoid syndrome. Eur. J. Endocrinol. 159, 275–282 (2008)

    Article  PubMed  CAS  Google Scholar 

  46. S. Ekeblad, B. Skogseid, K. Dunder, K. Oberg, B. Eriksson, Prognostic factors and survival in 324 patients with pancreatic endocrine tumor treated at a single institution. Clin. Cancer Res. 14, 7798–7803 (2008)

    Article  PubMed  CAS  Google Scholar 

  47. I.M. Modlin, B.I. Gustafsson, I. Drozdov, B. Nadler, R. Pfragner, M. Kidd, Principal component analysis, hierarchical clustering, and decision tree assessment of plasma mRNA and hormone levels as an early detection strategy for small intestinal neuroendocrine (carcinoid) tumors. Ann. Surg. Oncol. 16, 487–498 (2009)

    Article  PubMed  Google Scholar 

  48. A. Sundin, U. Garske, H. Orlefors, Nuclear imaging of neuroendocrine tumours. Best Pract. Res. Clin. Endocrinol. Metab. 21, 69–85 (2007)

    Article  PubMed  CAS  Google Scholar 

  49. A.I. Vinik, E.A. Woltering, R.R. Warner, M. Caplin, T.M. O’Dorisio, G.A. Wiseman, D. Coppola, V.L. Go, NANETS consensus guidelines for the diagnosis of neuroendocrine tumor. Pancreas. 39, 713–734 (2010)

    Article  PubMed  Google Scholar 

  50. M. Gabriel, A. Oberauer, G. Dobrozemsky, C. Decristoforo, D. Putzer, D. Kendler, C. Uprimny, P. Kovacs, R. Bale, I.J. Virgolini, 68 Ga-DOTA-Tyr3-octreotide PET for assessing response to somatostatin-receptor-mediated radionuclide therapy. J. Nucl. Med. 50, 1427–1434 (2009)

    Article  PubMed  CAS  Google Scholar 

  51. M. Gabriel, C. Decristoforo, D. Kendler, G. Dobrozemsky, D. Heute, C. Uprimny, P. Kovacs, G.E. Von, R. Bale, I.J. Virgolini, 68 Ga-DOTA-Tyr3-octreotide PET in neuroendocrine tumors: comparison with somatostatin receptor scintigraphy and CT. J. Nucl. Med. 48, 508–518 (2007)

    Article  PubMed  CAS  Google Scholar 

  52. T. Binderup, U. Knigge, A. Loft, J. Mortensen, A. Pfeifer, B. Federspiel, C.P. Hansen, L. Hojgaard, A. Kjaer, Functional imaging of neuroendocrine tumors: a head-to-head comparison of somatostatin receptor scintigraphy, 123I-MIBG scintigraphy, and 18F-FDG PET. J. Nucl. Med. 51, 704–712 (2010)

    Article  PubMed  Google Scholar 

  53. A. Haug, C.J. Auernhammer, B. Wangler, R. Tiling, G. Schmidt, B. Goke, P. Bartenstein, G. Popperl, Intraindividual comparison of 68 Ga-DOTA-TATE and 18F-DOPA PET in patients with well-differentiated metastatic neuroendocrine tumours. Eur. J. Nucl. Med. Mol. Imaging. 36, 765–770 (2009)

    Article  PubMed  CAS  Google Scholar 

  54. E.A. Eisenhauer, P. Therasse, J. Bogaerts, L.H. Schwartz, D. Sargent, R. Ford, J. Dancey, S. Arbuck, S. Gwyther, M. Mooney, L. Rubinstein, L. Shankar, L. Dodd, R. Kaplan, D. Lacombe, J. Verweij, New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur. J. Cancer. 45, 228–247 (2009)

    Article  PubMed  CAS  Google Scholar 

  55. H. Choi, C. Charnsangavej, S.C. Faria, H.A. Macapinlac, M.A. Burgess, S.R. Patel, L.L. Chen, D.A. Podoloff, R.S. Benjamin, Correlation of computed tomography and positron emission tomography in patients with metastatic gastrointestinal stromal tumor treated at a single institution with imatinib mesylate: proposal of new computed tomography response criteria. J. Clin. Oncol. 25, 1753–1759 (2007)

    Article  PubMed  Google Scholar 

  56. X. Hong, H. Choi, E.M. Loyer, R.S. Benjamin, J.C. Trent, C. Charnsangavej, Gastrointestinal stromal tumor: role of CT in diagnosis and in response evaluation and surveillance after treatment with imatinib. Radiographics. 26, 481–495 (2006)

    Article  PubMed  Google Scholar 

  57. I. Kayani, J.B. Bomanji, A. Groves, G. Conway, S. Gacinovic, T. Win, J. Dickson, M. Caplin, P.J. Ell, Functional imaging of neuroendocrine tumors with combined PET/CT using 68 Ga-DOTATATE (DOTA-DPhe1, Tyr3-octreotate) and 18F-FDG. Cancer. 112, 2447–2455 (2008)

    Article  PubMed  Google Scholar 

  58. D.L. Bushnell, R.P. Baum 2011 Standard imaging techniques for neuroendocrine tumors. Endocrinol .Metab. Clin. North Am. 40,153–62, ix

    Google Scholar 

  59. T. Binderup, U. Knigge, A. Loft, B. Federspiel, A. Kjaer, 18F-fluorodeoxyglucose positron emission tomography predicts survival of patients with neuroendocrine tumors. Clin. Cancer Res. 16, 978–985 (2010)

    Article  PubMed  CAS  Google Scholar 

  60. E. Garin, J.F. Le, A. Devillers, M. Cuggia, A.S. de Lajarte-Thirouard, C. Bouriel, E. Boucher, J.L. Raoul, Predictive value of 18F-FDG PET and somatostatin receptor scintigraphy in patients with metastatic endocrine tumors. J. Nucl. Med. 50, 858–864 (2009)

    Article  PubMed  CAS  Google Scholar 

  61. I. Virgolini, V. Ambrosini, J.B. Bomanji, R.P. Baum, S. Fanti, M. Gabriel, N.D. Papathanasiou, G. Pepe, W. Oyen, C.C. De, A. Chiti, Procedure guidelines for PET/CT tumour imaging with 68 Ga-DOTA-conjugated peptides: 68 Ga-DOTA-TOC, 68 Ga-DOTA-NOC, 68 Ga-DOTA-TATE. Eur. J. Nucl. Med. Mol. Imaging. 37, 2004–2010 (2010)

    Article  PubMed  Google Scholar 

  62. C.S. Ng, D.L. Raunig, E.F. Jackson, E.A. Ashton, F. Kelcz, K.B. Kim, R. Kurzrock, T.M. McShane, Reproducibility of perfusion parameters in dynamic contrast-enhanced MRI of lung and liver tumors: effect on estimates of patient sample size in clinical trials and on individual patient responses. AJR. Am. J. Roentgenol. 194, W134–W140 (2010)

    Article  PubMed  Google Scholar 

  63. M.D. Pickles, P. Gibbs, M. Lowry, L.W. Turnbull, Diffusion changes precede size reduction in neoadjuvant treatment of breast cancer. Magn. Reson. Imaging. 24, 843–847 (2006)

    Article  PubMed  Google Scholar 

  64. T.L. Chenevert, C.R. Meyer, B.A. Moffat, A. Rehemtulla, S.K. Mukherji, S.S. Gebarski, D.J. Quint, P.L. Robertson, T.S. Lawrence, L. Junck, J.M. Taylor, T.D. Johnson, Q. Dong, K.M. Muraszko, J.A. Brunberg, B.D. Ross, Diffusion MRI: a new strategy for assessment of cancer therapeutic efficacy. Mol. Imaging. 1, 336–343 (2002)

    Article  PubMed  Google Scholar 

  65. E. Liapi, J.F. Geschwind, J.A. Vossen, M. Buijs, C.S. Georgiades, D.A. Bluemke, I.R. Kamel, Functional MRI evaluation of tumor response in patients with neuroendocrine hepatic metastasis treated with transcatheter arterial chemoembolization. AJR Am. J. Roentgenol. 190, 67–73 (2008)

    Article  PubMed  Google Scholar 

  66. Boudreaux JP 2011 Surgery for gastroenteropancreatic neuroendocrine tumors (GEPNETS). Endocrinol Metab Clin North Am 40:163-71, ix

    Google Scholar 

  67. J.A. Norton, M.L. Melcher, F. Gibril, R.T. Jensen, Gastric carcinoid tumors in multiple endocrine neoplasia-1 patients with Zollinger-Ellison syndrome can be symptomatic, demonstrate aggressive growth, and require surgical treatment. Surgery. 136, 1267–1274 (2004)

    Article  PubMed  Google Scholar 

  68. H. Gouya, O. Vignaux, J. Augui, B. Dousset, L. Palazzo, A. Louvel, S. Chaussade, P. Legmann, CT, endoscopic sonography, and a combined protocol for preoperative evaluation of pancreatic insulinomas. AJR Am. J. Roentgenol. 181, 987–992 (2003)

    PubMed  Google Scholar 

  69. T. Steinmuller, R. Kianmanesh, M. Falconi, A. Scarpa, B. Taal, D.J. Kwekkeboom, J.M. Lopes, A. Perren, G. Nikou, J. Yao, G.F. Delle Fave, D. O’Toole, Consensus guidelines for the management of patients with liver metastases from digestive (neuro)endocrine tumors: foregut, midgut, hindgut, and unknown primary. Neuroendocrinology. 87, 47–62 (2008)

    Article  PubMed  Google Scholar 

  70. S. Gupta, M.M. Johnson, R. Murthy, K. Ahrar, M.J. Wallace, D.C. Madoff, S.E. McRae, M.E. Hicks, S. Rao, J.N. Vauthey, J.A. Ajani, J.C. Yao, Hepatic arterial embolization and chemoembolization for the treatment of patients with metastatic neuroendocrine tumors: variables affecting response rates and survival. Cancer. 104, 1590–1602 (2005)

    Article  PubMed  Google Scholar 

  71. A.S. Kennedy, W.A. Dezarn, P. McNeillie, D. Coldwell, C. Nutting, D. Carter, R. Murthy, S. Rose, R.R. Warner, D. Liu, H. Palmedo, C. Overton, B. Jones, R. Salem, Radioembolization for unresectable neuroendocrine hepatic metastases using resin 90Y-microspheres: early results in 148 patients. Am. J. Clin. Oncol. 31, 271–279 (2008)

    Article  PubMed  Google Scholar 

  72. J. King, R. Quinn, D.M. Glenn, J. Janssen, D. Tong, W. Liaw, D.L. Morris, Radioembolization with selective internal radiation microspheres for neuroendocrine liver metastases. Cancer. 113, 921–929 (2008)

    Article  PubMed  Google Scholar 

  73. M. Kalinowski, M. Dressler, A. Konig, M. El-Sheik, A. Rinke, H. Hoffken, T.M. Gress, R. Arnold, K.J. Klose, H.J. Wagner, Selective internal radiotherapy with Yttrium-90 microspheres for hepatic metastatic neuroendocrine tumors: a prospective single center study. Digestion. 79, 137–142 (2009)

    Article  PubMed  Google Scholar 

  74. I.M. Modlin, M. Pavel, M. Kidd, B.I. Gustafsson, Review article: somatostatin analogues in the treatment of gastroenteropancreatic neuroendocrine (carcinoid) tumours. Aliment. Pharmacol. Ther. 31, 169–188 (2010)

    PubMed  CAS  Google Scholar 

  75. Oberg K, Kvols L, Caplin M, Delle FG, de HW, Rindi G, Ruszniewski P, Woltering EA, Wiedenmann B 2004 Consensus report on the use of somatostatin analogs for the management of neuroendocrine tumors of the gastroenteropancreatic system. Ann Oncol 15:966-973

    Google Scholar 

  76. H.A. Schmid, P. Schoeffter, Functional activity of the multiligand analog SOM230 at human recombinant somatostatin receptor subtypes supports its usefulness in neuroendocrine tumors. Neuroendocrinology. 80(Suppl 1), 47–50 (2004)

    Article  PubMed  CAS  Google Scholar 

  77. P. Jaquet, G. Gunz, A. Saveanu, A. Barlier, H. Dufour, J. Taylor, J. Dong, S. Kim, J.P. Moreau, M.D. Culler, BIM-23A760, a chimeric molecule directed towards somatostatin and dopamine receptors, vs universal somatostatin receptors ligands in GH-secreting pituitary adenomas partial responders to octreotide. J. Endocrinol. Invest. 28, 21–27 (2005)

    PubMed  CAS  Google Scholar 

  78. G. Kaltsas, A. Rockall, D. Papadogias, R. Reznek, A.B. Grossman, Recent advances in radiological and radionuclide imaging and therapy of neuroendocrine tumours. Eur. J. Endocrinol. 151, 15–27 (2004)

    Article  PubMed  CAS  Google Scholar 

  79. Kwekkeboom DJ, Kam BL, van EM, Teunissen JJ, van Eijck CH, Valkema R, de JM, de Herder WW, Krenning EP 2010 Somatostatin-receptor-based imaging and therapy of gastroenteropancreatic neuroendocrine tumors. Endocr Relat Cancer 17:R53-R73

  80. Fazio N, Cinieri S, Lorizzo K, Squadroni M, Orlando L, Spada F, Maiello E, Bodei L, Paganelli G, Delle FG, de BF 2010 Biological targeted therapies in patients with advanced enteropancreatic neuroendocrine carcinomas. Cancer Treat Rev 36 (Suppl 3):S87-S94

  81. Kwekkeboom DJ, de Herder WW, Kam BL, van Eijck CH, van EM, Kooij PP, Feelders RA, van Aken MO, Krenning EP 2008 Treatment with the radiolabeled somatostatin analog [177 Lu-DOTA 0,Tyr3]octreotate: toxicity, efficacy, and survival. J Clin Oncol 26:2124-2130

  82. D.L. Bushnell Jr, T.M. O’Dorisio, M.S. O’Dorisio, Y. Menda, R.J. Hicks, C.E. Van, J.L. Baulieu, F. Borson-Chazot, L. Anthony, A.B. Benson, K. Oberg, A.B. Grossman, M. Connolly, H. Bouterfa, Y. Li, K.A. Kacena, N. LaFrance, S.A. Pauwels, 90Y-edotreotide for metastatic carcinoid refractory to octreotide. J. Clin. Oncol. 28, 1652–1659 (2010)

    Article  PubMed  CAS  Google Scholar 

  83. S. Faivre, G. Demetri, W. Sargent, E. Raymond, Molecular basis for sunitinib efficacy and future clinical development. Nat. Rev. Drug Discov. 6, 734–745 (2007)

    Article  PubMed  CAS  Google Scholar 

  84. S. Faivre, C. Delbaldo, K. Vera, C. Robert, S. Lozahic, N. Lassau, C. Bello, S. Deprimo, N. Brega, G. Massimini, J.P. Armand, P. Scigalla, E. Raymond, Safety, pharmacokinetic, and antitumor activity of SU11248, a novel oral multitarget tyrosine kinase inhibitor, in patients with cancer. J. Clin. Oncol. 24, 25–35 (2006)

    Article  PubMed  CAS  Google Scholar 

  85. M.H. Kulke, H.J. Lenz, N.J. Meropol, J. Posey, D.P. Ryan, J. Picus, E. Bergsland, K. Stuart, L. Tye, X. Huang, J.Z. Li, C.M. Baum, C.S. Fuchs, Activity of sunitinib in patients with advanced neuroendocrine tumors. J. Clin. Oncol. 26, 3403–3410 (2008)

    Article  PubMed  CAS  Google Scholar 

  86. L. Ciuffreda, S.C. Di, U.C. Incani, M. Milella, The mTOR pathway: a new target in cancer therapy. Curr. Cancer. Drug Targets. 10, 484–495 (2010)

    Article  PubMed  CAS  Google Scholar 

  87. Faggiano A, Ramundo V, Dicitore A, Castiglioni S, Borghi MO, Severino R, Ferolla P, Crino L, Abbruzzese A, Sperlongano P, Caraglia M, Ferone D, Hofland L, Colao A, Vitale G 2011 Everolimus is an active agent in medullary thyroid cancer: a clinical and in vitro study. J Cell Mol Med

  88. J.C. Yao, A.T. Phan, D.Z. Chang, R.A. Wolff, K. Hess, S. Gupta, C. Jacobs, J.E. Mares, A.N. Landgraf, A. Rashid, F. Meric-Bernstam, 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, 4311–4318 (2008)

    Article  PubMed  Google Scholar 

  89. A. O’Donnell, S. Faivre, H.A. Burris III, D. Rea, V. Papadimitrakopoulou, N. Shand, H.A. Lane, K. Hazell, U. Zoellner, J.M. Kovarik, C. Brock, S. Jones, E. Raymond, I. Judson, Phase I pharmacokinetic and pharmacodynamic study of the oral mammalian target of rapamycin inhibitor everolimus in patients with advanced solid tumors. J. Clin. Oncol. 26, 1588–1595 (2008)

    Article  PubMed  Google Scholar 

  90. J.C. Yao, A. Phan, P.M. Hoff, H.X. Chen, C. Charnsangavej, S.C. Yeung, K. Hess, C. Ng, J.L. Abbruzzese, J.A. Ajani, Targeting vascular endothelial growth factor in advanced carcinoid tumor: a random assignment phase II study of depot octreotide with bevacizumab and pegylated interferon alpha-2b. J. Clin. Oncol. 26, 1316–1323 (2008)

    Article  PubMed  CAS  Google Scholar 

  91. M.H. Kulke, J.A. Chan, J.A. Meyerhardt, A.X. Zhu, T.A. Abrams, L.S. Blaszkowsky, E. Regan, C. Sidor, C.S. Fuchs, A prospective phase II study of 2-methoxyestradiol administered in combination with bevacizumab in patients with metastatic carcinoid tumors. Cancer Chemother. Pharmacol. 68, 293–300 (2011)

    Article  PubMed  CAS  Google Scholar 

  92. R. Arnold, A. Rinke, C. Schmidt, L. Hofbauer, Endocrine tumours of the gastrointestinal tract: chemotherapy. Best Pract. Res. Clin. Gastroenterol. 19, 649–656 (2005)

    Article  PubMed  CAS  Google Scholar 

  93. J.R. Strosberg, L.K. Kvols, A review of the current clinical trials for gastroenteropancreatic neuroendocrine tumours. Expert Opin. Investig. Drugs. 16, 219–224 (2007)

    Article  PubMed  CAS  Google Scholar 

  94. M.H. Kulke, K. Stuart, P.C. Enzinger, D.P. Ryan, J.W. Clark, A. Muzikansky, M. Vincitore, A. Michelini, C.S. Fuchs, Phase II study of temozolomide and thalidomide in patients with metastatic neuroendocrine tumors. J. Clin. Oncol. 24, 401–406 (2006)

    Article  PubMed  CAS  Google Scholar 

  95. M.H. Kulke, J.L. Hornick, C. Frauenhoffer, S. Hooshmand, D.P. Ryan, P.C. Enzinger, J.A. Meyerhardt, J.W. Clark, K. Stuart, C.S. Fuchs, M.S. Redston, O6-methylguanine DNA methyltransferase deficiency and response to temozolomide-based therapy in patients with neuroendocrine tumors. Clin. Cancer Res. 15, 338–345 (2009)

    Article  PubMed  CAS  Google Scholar 

  96. S. Ekeblad, A. Sundin, E.T. Janson, S. Welin, D. Granberg, H. Kindmark, K. Dunder, G. Kozlovacki, H. Orlefors, M. Sigurd, K. Oberg, B. Eriksson, B. Skogseid, Temozolomide as monotherapy is effective in treatment of advanced malignant neuroendocrine tumors. Clin. Cancer Res. 13, 2986–2991 (2007)

    Article  PubMed  CAS  Google Scholar 

  97. J.R. Strosberg, R.L. Fine, J. Choi, A. Nasir, D. Coppola, D.T. Chen, J. Helm, L. Kvols, First-line chemotherapy with capecitabine and temozolomide in patients with metastatic pancreatic endocrine carcinomas. Cancer 117, 268–275 (2011)

    Article  PubMed  CAS  Google Scholar 

  98. M. Caraglia, M. Marra, P. Tagliaferri, S.W. Lamberts, S. Zappavigna, G. Misso, F. Cavagnini, G. Facchini, A. Abbruzzese, L.J. Hofland, G. Vitale, Emerging strategies to strengthen the anti-tumour activity of type I interferons: overcoming survival pathways. Curr. Cancer Drug Targets. 9, 690–704 (2009)

    Article  PubMed  CAS  Google Scholar 

  99. G. Vitale, W.W. de Herder, P.M. van Koetsveld, M. Waaijers, W. Schoordijk, E. Croze, A. Colao, S.W. Lamberts, L.J. Hofland, IFN-beta is a highly potent inhibitor of gastroenteropancreatic neuroendocrine tumor cell growth in vitro. Cancer Res. 66, 554–562 (2006)

    Article  PubMed  CAS  Google Scholar 

  100. G. Vitale, P.M. van Koetsveld, W.W. de Herder, K. van der Wansem, J.A. Janssen, A. Colao, G. Lombardi, S.W. Lamberts, L.J. Hofland, Effects of type I interferons on IGF-mediated autocrine/paracrine growth of human neuroendocrine tumor cells. Am J Physiol Endocrinol Metab. 296, E559–E566 (2009)

    Article  PubMed  CAS  Google Scholar 

  101. A.W. Tolcher, J. Sarantopoulos, A. Patnaik, K. Papadopoulos, C.C. Lin, J. Rodon, B. Murphy, B. Roth, I. McCaffery, K.S. Gorski, B. Kaiser, M. Zhu, H. Deng, G. Friberg, I. Puzanov, Phase I, pharmacokinetic, and pharmacodynamic study of AMG 479, a fully human monoclonal antibody to insulin-like growth factor receptor 1. J. Clin. Oncol. 27, 5800–5807 (2009)

    Article  PubMed  CAS  Google Scholar 

  102. E. Allen, I.B. Walters, D. Hanahan, Brivanib, a dual FGF/VEGF inhibitor, is active both first and second line against mouse pancreatic neuroendocrine tumors developing adaptive/evasive resistance to VEGF inhibition. Clin. Cancer Res. 17, 5299–5310 (2011)

    Article  PubMed  CAS  Google Scholar 

  103. McDermott AM, Heneghan HM, Miller N, Kerin MJ (2011) The Therapeutic Potential of MicroRNAs: Disease Modulators and Drug Targets. Pharm Res

  104. C. Huynh, M.F. Segura, A. Gaziel-Sovran, S. Menendez, F. Darvishian, L. Chiriboga, B. Levin, D. Meruelo, I. Osman, J. Zavadil, E.G. Marcusson, E. Hernando, Efficient in vivo microRNA targeting of liver metastasis. Oncogene. 30, 1481–1488 (2011)

    Article  PubMed  CAS  Google Scholar 

  105. M.F. Segura, D. Hanniford, S. Menendez, L. Reavie, X. Zou, S. Alvarez-Diaz, J. Zakrzewski, E. Blochin, A. Rose, D. Bogunovic, D. Polsky, J. Wei, P. Lee, I. Belitskaya-Levy, N. Bhardwaj, I. Osman, E. Hernando, Aberrant miR-182 expression promotes melanoma metastasis by repressing FOXO3 and microphthalmia-associated transcription factor. Proc. Natl. Acad. Sci. USA. 106, 1814–1819 (2009)

    Article  PubMed  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Gregory Kaltsas.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Alexandraki, K.I., Kaltsas, G. Gastroenteropancreatic neuroendocrine tumors: new insights in the diagnosis and therapy. Endocrine 41, 40–52 (2012). https://doi.org/10.1007/s12020-011-9562-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12020-011-9562-2

Keywords

Navigation