open access

Vol 68, No 2 (2017)
Review paper
Submitted: 2017-04-04
Accepted: 2017-04-05
Published online: 2017-05-08
Get Citation

The content of this article is also available in the following languages:
Polski

Pancreatic neuroendocrine neoplasms — management guidelines (recommended by the Polish Network of Neuroendocrine Tumours)

Beata Kos-Kudła, Violetta Rosiek, Małgorzata Borowska, Agata Bałdys-Waligórska, Tomasz Bednarczuk, Jolanta Blicharz-Dorniak, Marek Bolanowski, Agnieszka Boratyn-Nowicka, Andrzej Cichocki, Jarosław B. Ćwikła, Massimo Falconi, Wanda Foltyn, Foltyn Handkiewicz-Junak, Alicja Hubalewska-Dydejczyk, Barbara Jarząb, Michał Jarząb, Roman Junik, Dariusz Kajdaniuk, Grzegorz Kamiński, Agnieszka Kolasińska-Ćwikła, Aldona Kowalska, Robert Król, Leszek Królicki, Jolanta Kunikowska, Katarzyna Kuśnierz, Paweł Lampe, Dariusz Lange, Anna Lewczuk-Myślicka, Andrzej Lewiński, Michał Lipiński, Magdalena Londzin-Olesik, Bogdan Marek, Anna Nasierowska-Guttmejer, Ewa Nowakowska-Duława, Joanna Pilch-Kowalczyk, Marek Ruchała, Lucyna Siemińska, Anna Sowa-Staszczak, Teresa Starzyńska, Katarzyna Steinhof-Radwańska, Janusz Strzelczyk, Krzysztof Sworczak, Anhelli Syrenicz1, Andrzej Szawłowski, Marek Szczepkowski, Ewa Wachuła, Wojciech Zajęcki, Anna Zemczak, Wojciech Zgliczyński
·
Pubmed: 28540973
·
Endokrynol Pol 2017;68(2):169-197.
Affiliations
  1. Klinika Endokrynologii, Chorób Metabolicznych i Chorób Wewnętrznych, Pomorski Uniwersytet Medyczny, ul. Unii Lubelskiej 1, 71-252 Szczecin, Poland

open access

Vol 68, No 2 (2017)
Reviews — Postgraduate Education
Submitted: 2017-04-04
Accepted: 2017-04-05
Published online: 2017-05-08

Abstract

This article presents updated diagnostic and therapeutic guidelines for the management of pancreatic neuroendocrine tumours (PNEN), proposed by the Polish Network of Neuroendocrine Tumours. The guidelines contain new data received in the years 2013–2016, which confirm previous recommendations, and have led to modification of previous guidelines or have resulted in the formulation of new guidelines. Biochemical and imaging (anatomical and functional) tests are of great importance in diagnostics, as well as histopathological diagnosis to determine the management of PNEN patients, but they must be confirmed by an immunohistochemical examination. PNEN therapy requires collaboration among the members a multidisciplinary team of specialists experienced in the management of these neoplasms. Surgery is the basic form of treatment in many cases. Further therapy requires a multidirectional procedure; therefore, the rules of biotherapy, peptide receptor radionuclide therapy, molecular targeted therapy, and chemotherapy are discussed.

Abstract

This article presents updated diagnostic and therapeutic guidelines for the management of pancreatic neuroendocrine tumours (PNEN), proposed by the Polish Network of Neuroendocrine Tumours. The guidelines contain new data received in the years 2013–2016, which confirm previous recommendations, and have led to modification of previous guidelines or have resulted in the formulation of new guidelines. Biochemical and imaging (anatomical and functional) tests are of great importance in diagnostics, as well as histopathological diagnosis to determine the management of PNEN patients, but they must be confirmed by an immunohistochemical examination. PNEN therapy requires collaboration among the members a multidisciplinary team of specialists experienced in the management of these neoplasms. Surgery is the basic form of treatment in many cases. Further therapy requires a multidirectional procedure; therefore, the rules of biotherapy, peptide receptor radionuclide therapy, molecular targeted therapy, and chemotherapy are discussed.

Get Citation

Keywords

pancreatic neuroendocrine neoplasms; functional; non-functional; diagnostics; therapy; guidelines

About this article
Title

Pancreatic neuroendocrine neoplasms — management guidelines (recommended by the Polish Network of Neuroendocrine Tumours)

Journal

Endokrynologia Polska

Issue

Vol 68, No 2 (2017)

Article type

Review paper

Pages

169-197

Published online

2017-05-08

Page views

6806

Article views/downloads

23571

DOI

10.5603/EP.2017.2017

Pubmed

28540973

Bibliographic record

Endokrynol Pol 2017;68(2):169-197.

Keywords

pancreatic neuroendocrine neoplasms
functional
non-functional
diagnostics
therapy
guidelines

Authors

Beata Kos-Kudła
Violetta Rosiek
Małgorzata Borowska
Agata Bałdys-Waligórska
Tomasz Bednarczuk
Jolanta Blicharz-Dorniak
Marek Bolanowski
Agnieszka Boratyn-Nowicka
Andrzej Cichocki
Jarosław B. Ćwikła
Massimo Falconi
Wanda Foltyn
Foltyn Handkiewicz-Junak
Alicja Hubalewska-Dydejczyk
Barbara Jarząb
Michał Jarząb
Roman Junik
Dariusz Kajdaniuk
Grzegorz Kamiński
Agnieszka Kolasińska-Ćwikła
Aldona Kowalska
Robert Król
Leszek Królicki
Jolanta Kunikowska
Katarzyna Kuśnierz
Paweł Lampe
Dariusz Lange
Anna Lewczuk-Myślicka
Andrzej Lewiński
Michał Lipiński
Magdalena Londzin-Olesik
Bogdan Marek
Anna Nasierowska-Guttmejer
Ewa Nowakowska-Duława
Joanna Pilch-Kowalczyk
Marek Ruchała
Lucyna Siemińska
Anna Sowa-Staszczak
Teresa Starzyńska
Katarzyna Steinhof-Radwańska
Janusz Strzelczyk
Krzysztof Sworczak
Anhelli Syrenicz
Andrzej Szawłowski
Marek Szczepkowski
Ewa Wachuła
Wojciech Zajęcki
Anna Zemczak
Wojciech Zgliczyński

References (252)
  1. Capelli P, Fassan M, Scarpa A. Pathology - grading and staging of GEP-NETs. Best Pract Res Clin Gastroenterol. 2012; 26(6): 705–717.
  2. Ito T, Igarashi H, Nakamura K, et al. Epidemiological trends of pancreatic and gastrointestinal neuroendocrine tumors in Japan: a nationwide survey analysis. J Gastroenterol. 2015; 50(1): 58–64.
  3. Cho MY, Kim JM, Sohn JH, et al. Gastrointestinal Pathology Study Group of Korean Society of Pathologists. Current Trends of the Incidence and Pathological Diagnosis of Gastroenteropancreatic Neuroendocrine Tumors (GEP-NETs) in Korea 2000-2009: Multicenter Study. Cancer Res Treat. 2012; 44(3): 157–165.
  4. Tsai HJ, Wu CC, Tsai CR, et al. The epidemiology of neuroendocrine tumors in Taiwan: a nation-wide cancer registry-based study. PLoS One. 2013; 8(4): e62487.
  5. Scherübl H, Streller B, Stabenow R, et al. Clinically detected gastroenteropancreatic neuroendocrine tumors are on the rise: epidemiological changes in Germany. World J Gastroenterol. 2013; 19(47): 9012–9019.
  6. Kos-Kudła B, Hubalewska-Dydejczyk A, Kuśnierz K, et al. Pancreatic endocrine tumors — management guidelines (recommended by the Polish Network of Neuroendocrine Tumors). Endokrynol Pol 2013; 64 (6): 459-479.
  7. Öberg K, Knigge U, Kwekkeboom D, et al. ESMO Guidelines Working Group. Neuroendocrine gastro-entero-pancreatic tumors: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2012; 23 Suppl 7: vii124–vii130.
  8. Falconi M, Bartsch DK, Eriksson B, et al. Barcelona Consensus Conference participants. ENETS Consensus Guidelines for the management of patients with digestive neuroendocrine neoplasms of the digestive system: well-differentiated pancreatic non-functioning tumors. Neuroendocrinology. 2012; 95(2): 120–134.
  9. Jensen RT, Cadiot G, Brandi ML, et al. Barcelona Consensus Conference participants. ENETS Consensus Guidelines for the management of patients with digestive neuroendocrine neoplasms: functional pancreatic endocrine tumor syndromes. Neuroendocrinology. 2012; 95(2): 98–119.
  10. Oberg K. Pancreatic endocrine tumors. Semin Oncol 2010; 37: 594–618.
  11. Falconi M, Eriksson B, Kaltsas G, et al. Vienna Consensus Conference participants. ENETS Consensus Guidelines Update for the Management of Patients with Functional Pancreatic Neuroendocrine Tumors and Non-Functional Pancreatic Neuroendocrine Tumors. Neuroendocrinology. 2016; 103(2): 153–171.
  12. Jensen RT, Berna MJ, Bingham DB, et al. Inherited pancreatic endocrine tumor syndromes: advances in molecular pathogenesis, diagnosis, management, and controversies. Cancer. 2008; 113(7 Suppl): 1807–1843.
  13. Ito T, Igarashi H, Nakamura K, et al. Causes of death and prognostic factors in multiple endocrine neoplasia type 1: a prospective study: comparison of 106 MEN1/Zollinger-Ellison syndrome patients with 1613 literature MEN1 patients with or without pancreatic endocrine tumors. Medicine (Baltimore). 2013; 92(3): 135–181.
  14. Lévy-Bohbot N, Merle C, Goudet P, et al. Groupe des Tumeurs Endocrines. Prevalence, characteristics and prognosis of MEN 1-associated glucagonomas, VIPomas, and somatostatinomas: study from the GTE (Groupe des Tumeurs Endocrines) registry. Gastroenterol Clin Biol. 2004; 28(11): 1075–1081.
  15. Thakker RV, Newey PJ, Walls GV, et al. Endocrine Society. Clinical practice guidelines for multiple endocrine neoplasia type 1 (MEN1). J Clin Endocrinol Metab. 2012; 97(9): 2990–3011.
  16. Le Roith D. Tumor-induced hypoglycemia. N Engl J Med. 1999; 341(10): 757–758.
  17. Vanderveen K, Grant C. Insulinoma. Cancer Treat Res. 2010; 153: 235–252.
  18. Zhao YP, Zhan HX, Zhang TP, et al. Surgical management of patients with insulinomas: Result of 292 cases in a single institution. J Surg Oncol. 2011; 103(2): 169–174.
  19. Gnacńska M, Lewczuk A, Sworczak K. [Insulinoma misdiagnosed and treated as epilepsy]. Pol Merkur Lekarski. 2008; 24(141): 251–253.
  20. Placzkowski KA, Vella A, Thompson GB, et al. Secular trends in the presentation and management of functioning insulinoma at the Mayo Clinic, 1987-2007. J Clin Endocrinol Metab. 2009; 94(4): 1069–1073.
  21. Toaiari M, Davì MV, Dalle Carbonare L, et al. Presentation, diagnostic features and glucose handling in a monocentric series of insulinomas. J Endocrinol Invest. 2013; 36(9): 753–758.
  22. Iida K, Ohara T, Hino Y, et al. Glucose-responsive insulinoma in a patient with postprandial hypoglycemia in the morning. Intern Med. 2010; 49(19): 2123–2127.
  23. de Herder WW, Niederle B, Scoazec JY, et al. Frascati Consensus Conference, European Neuroendocrine Tumor Society. Well-differentiated pancreatic tumor/carcinoma: insulinoma. Neuroendocrinology. 2006; 84(3): 183–188.
  24. Marek B, Kajdaniuk D, Kos-Kudła B, et al. Insulinoma - diagnosis and treatment. Endokrynol Pol. 2007; 58(1): 58–62.
  25. Ekeblad S. Islet cell tumours. Adv Exp Med Biol. 2010; 654: 771–789.
  26. O’Toole D, Salazar R, Falconi M, et al. Rare Functioning Pancreatic Endocrine Tumors. Neuroendocrinology. 2007; 84(3): 189–195.
  27. Metz DC, Jensen RT. Gastrointestinal neuroendocrine tumors: pancreatic endocrine tumors. Gastroenterology. 2008; 135(5): 1469–1492.
  28. Vagefi PA, Razo O, Deshpande V, et al. Evolving patterns in the detection and outcomes of pancreatic neuroendocrine neoplasms: the Massachusetts General Hospital experience from 1977 to 2005. Arch Surg. 2007; 142(4): 347–354.
  29. Halfdanarson TR, Rabe KG, Rubin J, et al. Pancreatic neuroendocrine tumors (PNETs): incidence, prognosis and recent trend toward improved survival. Ann Oncol. 2008; 19(10): 1727–1733.
  30. Yao JC, Hassan M, Phan A, et al. One hundred years after. J Clin Oncol. 2008; 26(18): 3063–3072.
  31. Triponez F, Dosseh D, Goudet P, et al. Epidemiology data on 108 MEN 1 patients from the GTE with isolated nonfunctioning tumors of the pancreas. Ann Surg. 2006; 243(2): 265–272.
  32. Blansfield JA, Choyke L, Morita SY, et al. Clinical, genetic and radiographic analysis of 108 patients with von Hippel-Lindau disease (VHL) manifested by pancreatic neuroendocrine neoplasms (PNETs). Surgery. 2007; 142(6): 814–8; discussion 818.e1.
  33. Cheslyn-Curtis S, Sitaram V, Williamson RC. Management of non-functioning neuroendocrine tumours of the pancreas. Br J Surg. 1993; 80(5): 625–627.
  34. Madura JA, Cummings OW, Wiebke EA, et al. Nonfunctioning islet cell tumors of the pancreas: a difficult diagnosis but one worth the effort. Am Surg. 1997; 63(7): 573–7; discussion 577.
  35. Chu QD, Hill HC, Douglass HO, et al. Predictive factors associated with long-term survival in patients with neuroendocrine tumors of the pancreas. Ann Surg Oncol. 2002; 9(9): 855–862.
  36. Zerbi A, Falconi M, Rindi G, et al. AISP-Network Study Group. Clinicopathological features of pancreatic endocrine tumors: a prospective multicenter study in Italy of 297 sporadic cases. Am J Gastroenterol. 2010; 105(6): 1421–1429.
  37. Garcia-Carbonero R, Capdevila J, Crespo-Herrero G, et al. Incidence, patterns of care and prognostic factors for outcome of gastroenteropancreatic neuroendocrine tumors (GEP-NETs): results from the National Cancer Registry of Spain (RGETNE). Ann Oncol. 2010; 21(9): 1794–1803.
  38. Kazanjian KK, Reber HA, Hines OJ. Resection of pancreatic neuroendocrine tumors: results of 70 cases. Arch Surg. 2006; 141(8): 765–9; discussion 769.
  39. Pape UF, Böhmig M, Berndt U, et al. Survival and clinical outcome of patients with neuroendocrine tumors of the gastroenteropancreatic tract in a german referral center. Ann N Y Acad Sci. 2004; 1014: 222–233.
  40. Ferrone CR, Tang LH, Tomlinson J, et al. Determining prognosis in patients with pancreatic endocrine neoplasms: can the WHO classification system be simplified? J Clin Oncol. 2007; 25(35): 5609–5615.
  41. Bilimoria KY, Talamonti MS, Tomlinson JS, et al. Prognostic score predicting survival after resection of pancreatic neuroendocrine tumors: analysis of 3851 patients. Ann Surg. 2008; 247(3): 490–500.
  42. Poultsides GA, Huang LC, Chen Y, et al. Pancreatic neuroendocrine tumors: radiographic calcifications correlate with grade and metastasis. Ann Surg Oncol. 2012; 19(7): 2295–2303.
  43. Panzuto F, Merola E, Rinzivillo M, et al. Advanced digestive neuroendocrine tumors: metastatic pattern is an independent factor affecting clinical outcome. Pancreas. 2014; 43(2): 212–218.
  44. Panzuto F, Boninsegna L, Fazio N, et al. Metastatic and locally advanced pancreatic endocrine carcinomas: analysis of factors associated with disease progression. J Clin Oncol. 2011; 29(17): 2372–2377.
  45. Halfdanarson TR, Rubin J, Farnell MB, et al. Pancreatic endocrine neoplasms: epidemiology and prognosis of pancreatic endocrine tumors. Endocr Relat Cancer. 2008; 15(2): 409–427.
  46. Ilias I, Torpy DJ, Pacak K, et al. Cushing's syndrome due to ectopic corticotropin secretion: twenty years' experience at the National Institutes of Health. J Clin Endocrinol Metab. 2005; 90(8): 4955–4962.
  47. Qiao XW, Qiu L, Chen YJ, et al. Chromogranin A is a reliable serum diagnostic biomarker for pancreatic neuroendocrine tumors but not for insulinomas. BMC Endocr Disord. 2014; 14: 64.
  48. Guettier JM, Lungu A, Goodling A, et al. The role of proinsulin and insulin in the diagnosis of insulinoma: a critical evaluation of the Endocrine Society clinical practice guideline. J Clin Endocrinol Metab. 2013; 98(12): 4752–4758.
  49. Sakurai A, Yamazaki M, Suzuki S, et al. Clinical features of insulinoma in patients with multiple endocrine neoplasia type 1: analysis of the database of the MEN Consortium of Japan. Endocr J 2012;59:859-866.
  50. Gibril F, Schumann M, Pace A, 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. Medicine (Baltimore). 2004; 83(1): 43–83.
  51. Gonçalves TD, Toledo RA, Sekiya T, et al. Penetrance of functioning and nonfunctioning pancreatic neuroendocrine tumors in multiple endocrine neoplasia type 1 in the second decade of life. J Clin Endocrinol Metab. 2014; 99(1): E89–E96.
  52. Oberg K, Eriksson B. Endocrine tumours of the pancreas. Best Pract Res Clin Gastroenterol. 2005; 19(5): 753–781.
  53. Warner RRP. Enteroendocrine tumors other than carcinoid: a review of clinically significant advances. Gastroenterology. 2005; 128(6): 1668–1684.
  54. Cryer PE, Axelrod L, Grossman AB, et al. Endocrine Society. Evaluation and management of adult hypoglycemic disorders: an Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab. 2009; 94(3): 709–728.
  55. Ardill JES. Circulating markers for endocrine tumours of the gastroenteropancreatic tract. Ann Clin Biochem. 2008; 45(Pt 6): 539–559.
  56. Kos-Kudła B, Zemczak A. Diagnostyka biochemiczna guzów neuroendokrynnych układu pokarmowego. W: . (red.). Guzy neroendokrynne układu pokarmowego. Via Medica, Gdańsk. ; 2010: 17–24.
  57. Blicharz-Dorniak J, Kos-Kudła B, Foltyn W, et al. Is determination of matrix metalloproteinases and their tissue inhibitors serum concentrations useful in patients with gastroenteropancreatic and bronchopulmonary neuroendocrine neoplasms? Endokrynol Pol. 2012; 63(6): 470–476.
  58. Stridsberg M, Eriksson B, Fellström B, et al. Measurements of chromogranin B can serve as a complement to chromogranin A. Regul Pept. 2007; 139(1-3): 80–83.
  59. Modlin IM, Gustafsson BI, Moss SF, et al. Chromogranin A--biological function and clinical utility in neuro endocrine tumor disease. Ann Surg Oncol. 2010; 17(9): 2427–2443.
  60. Glinicki P, Jeske W, Kapuścińska R, et al. Comparison of chromogranin A (CgA) levels in serum and plasma (EDTA2K) and the respective reference ranges in healthy males. Endokrynol Pol. 2015; 66(1): 53–56.
  61. Baudin E, Gigliotti A, Ducreux M, et al. Neuron-specific enolase and chromogranin A as markers of neuroendocrine tumours. Br J Cancer. 1998; 78(8): 1102–1107.
  62. Korse CM, Taal BG, Vincent A, et al. Choice of tumour markers in patients with neuroendocrine tumours is dependent on the histological grade. A marker study of Chromogranin A, Neuron specific enolase, Progastrin-releasing peptide and cytokeratin fragments. Eur J Cancer. 2012; 48(5): 662–671.
  63. WHO Classification of Tumours of the Digestive System. IARC: Lyon 2017 (in press)
  64. Vagefi PA, Razo O, Deshpande V, et al. Evolving patterns in the detection and outcomes of pancreatic neuroendocrine neoplasms: the Massachusetts General Hospital experience from 1977 to 2005. Arch Surg. 2007; 142(4): 347–354.
  65. Bergsland EK, Woltering EA, Rindi G, et al. Neuroendocrine Tumors of the Pancreas. American Joint Committee on Cancer 2017. In: Amin M. B. et al (eds) AJCC Cancer Staging Manual. Eight Edition. Springer 2017: 407-419. DOI 10.1007/978-3-319-40618-3_30.
  66. Basturk O, Yang Z, Tang LH, et al. The high-grade (WHO G3) pancreatic neuroendocrine tumor category is morphologically and biologically heterogenous and includes both well differentiated and poorly differentiated neoplasms. Am J Surg Pathol. 2015; 39(5): 683–690.
  67. Crippa S, Partelli S, Belfiori G, et al. Management of neuroendocrine carcinomas of the pancreas (WHO G3): A tailored approach between proliferation and morphology. World J Gastroenterol. 2016; 22(45): 9944–9953.
  68. Milione M, Maisonneuve P, Spada F, et al. The Clinicopathologic Heterogeneity of Grade 3 Gastroenteropancreatic Neuroendocrine Neoplasms: Morphological Differentiation and Proliferation Identify Different Prognostic Categories. Neuroendocrinology. 2017; 104(1): 85–93.
  69. Rindi G, Klöppel G, Alhman H, et al. all other Frascati Consensus Conference participants, European Neuroendocrine Tumor Society (ENETS). TNM staging of foregut (neuro)endocrine tumors: a consensus proposal including a grading system. Virchows Arch. 2006; 449(4): 395–401.
  70. Brierley J, Gospodarowicz M, Wittekind C. Pancreas. TNM Online. 2017: 93–95.
  71. Nasierowska-Guttmejer A. Przyczyny opóźnionej diagnostyki i leczenia guzów neuroendokrynnych trzustki. Komentarz. Przegląd Gastroenterologiczny. 2009; 4: 221–223.
  72. Capelli P, Martignoni G, Pedica F, et al. Endocrine neoplasms of the pancreas: pathologic and genetic features. Arch Pathol Lab Med. 2009; 133(3): 350–364.
  73. Foltyn W, Zajęcki W, Marek B, et al. The value of the Ki-67 proliferation marker as a prognostic factor in gastroenteropancreatic neuroendocrine tumours. Endokrynol Pol. 2012; 63(5): 362–366.
  74. Chathadi KV, Khashab MA, Acosta RD, et al. ASGE Standards of Practice Committee. The role of endoscopy in ampullary and duodenal adenomas. Gastrointest Endosc. 2015; 82(5): 773–781.
  75. Eloubeidi MA, Decker GA, Chandrasekhara V, et al. ASGE Standards of Practice Committee. The role of endoscopy in the evaluation and management of patients with solid pancreatic neoplasia. Gastrointest Endosc. 2016; 83(1): 17–28.
  76. Kaltsas G, Rockall A, Papadogias D, et al. Recent advances in radiological and radionuclide imaging and therapy of neuroendocrine tumours. Eur J Endocrinol. 2004; 151(1): 15–27.
  77. Zimmer T, Stölzel U, Bäder M, et al. Endoscopic ultrasonography and somatostatin receptor scintigraphy in the preoperative localisation of insulinomas and gastrinomas. Gut. 1996; 39(4): 562–568.
  78. Ramage JK, Davies AHG, Ardill J, et al. UKNETwork for Neuroendocrine Tumours. Guidelines for the management of gastroenteropancreatic neuroendocrine (including carcinoid) tumours. Gut. 2005; 54 Suppl 4: iv1–i16.
  79. Hocke M, Schulze E, Gottschalk P, et al. Contrast-enhanced endoscopic ultrasound in discrimination between focal pancreatitis and pancreatic cancer. World J Gastroenterol. 2006; 12(2): 246–250.
  80. Hawes RH, Fockens P. Endosonography. Saunders Elsevier, Philadelphia. 2006: USA.
  81. Goldberg J, Rosenblat J, Khatri G, et al. Complementary roles of CT and endoscopic ultrasound in evaluating a pancreatic mass. AJR Am J Roentgenol. 2010; 194(4): 984–992.
  82. Ito T, Hijioka S, Masui T, et al. Advances in the diagnosis and treatment of pancreatic neuroendocrine neoplasms in Japan. J Gastroenterol. 2017; 52(1): 9–18.
  83. Gouya H, Vignaux O, Augui J, et al. CT, endoscopic sonography, and a combined protocol for preoperative evaluation of pancreatic insulinomas. AJR Am J Roentgenol. 2003; 181(4): 987–992.
  84. Zimmer T, Scherübl H, Faiss S, et al. Endoscopic ultrasonography of neuroendocrine tumours. Digestion. 2000; 62 Suppl 1: 45–50.
  85. Anderson MA, Carpenter S, Thompson NW, et al. Endoscopic ultrasound is highly accurate and directs management in patients with neuroendocrine tumors of the pancreas. Am J Gastroenterol. 2000; 95(9): 2271–2277.
  86. Manta R, Nardi E, Pagano N, et al. Pre-operative Diagnosis of Pancreatic Neuroendocrine Tumors with Endoscopic Ultrasonography and Computed Tomography in a Large Series. J Gastrointestin Liver Dis. 2016; 25(3): 317–321.
  87. Sugiyama M, Abe N, Izumisato Y, et al. Differential diagnosis of benign versus malignant nonfunctioning islet cell tumors of the pancreas: the roles of EUS and ERCP. Gastrointest Endosc. 2002; 55(1): 115–119.
  88. Gonçalves B, Soares J, Bastos P. Endoscopic Ultrasound in the Diagnosis and Staging of Pancreatic Cancer. GE Portuguese Journal of Gastroenterology. 2015; 22(4): 161–171.
  89. Sugimoto M, Takagi T, Hikichi T, et al. Efficacy of endoscopic ultrasonography-guided fine needle aspiration for pancreatic neuroendocrine tumor grading. World J Gastroenterol. 2015; 21(26): 8118–8124.
  90. Horiguchi S, Kato H, Shiraha H, et al. Dynamic computed tomography is useful for prediction of pathological grade in pancreatic neuroendocrine neoplasm. J Gastroenterol Hepatol. 2017; 32(4): 925–931.
  91. Lennon AM, Newman N, Makary MA, et al. EUS-guided tattooing before laparoscopic distal pancreatic resection (with video). Gastrointest Endosc. 2010; 72(5): 1089–1094.
  92. Newman NA, Lennon AM, Edil BH, et al. Preoperative endoscopic tattooing of pancreatic body and tail lesions decreases operative time for laparoscopic distal pancreatectomy. Surgery. 2010; 148(2): 371–377.
  93. Doi R. Determinants of surgical resection for pancreatic neuroendocrine tumors. J Hepatobiliary Pancreat Sci. 2015; 22(8): 610–617.
  94. Ricke J, Klose KJ, Mignon M, et al. Standardisation of imaging in neuroendocrine tumours: results of a European delphi process. Eur J Radiol. 2001; 37(1): 8–17.
  95. Ćwikła JB, Walecki J. Diagnostyka obrazowa guzów neuroendokrynnych trzustki z elementami leczenia radioizotopowego. Przegl Gastroenterol. 2006; 1: 31–44.
  96. Sahani DV, Bonaffini PA, Fernández-Del Castillo C, et al. Gastroenteropancreatic neuroendocrine tumors: role of imaging in diagnosis and management. Radiology. 2013; 266(1): 38–61.
  97. Takumi K, Fukukura Y, Higashi M, et al. Pancreatic neuroendocrine tumors: Correlation between the contrast-enhanced computed tomography features and the pathological tumor grade. Eur J Radiol. 2015; 84(8): 1436–1443.
  98. Tatsumoto S, Kodama Y, Sakurai Y, et al. Pancreatic neuroendocrine neoplasm: correlation between computed tomography enhancement patterns and prognostic factors of surgical and endoscopic ultrasound-guided fine-needle aspiration biopsy specimens. Abdom Imaging. 2013; 38(2): 358–366.
  99. Delrue L, Blanckaert P, Mertens D, et al. Tissue perfusion in pathologies of the pancreas: assessment using 128-slice computed tomography. Abdom Imaging. 2012; 37(4): 595–601.
  100. Kim JH, Eun HW, Kim YJ, et al. Pancreatic neuroendocrine tumour (PNET): Staging accuracy of MDCT and its diagnostic performance for the differentiation of PNET with uncommon CT findings from pancreatic adenocarcinoma. Eur Radiol. 2016; 26(5): 1338–1347.
  101. Pilch-Kowalczyk J. Anatomia radiologiczna i metody obrazowania trzustki w Leszczyński S, Pilch-Kowalczyk J (red.). Diagnostyka obrazowa Układ trawienny. PZWL, Warszawa 2012.
  102. Prokop M, Galanski M. Spiralna wielorzedowa tomografia komputerowa człowieka. Medipage, Warszawa 2015.
  103. Gouya H, Vignaux O, Augui J, et al. CT, endoscopic sonography, and a combined protocol for preoperative evaluation of pancreatic insulinomas. AJR Am J Roentgenol. 2003; 181(4): 987–992.
  104. Al-Hawary MM, Francis IR, Chari ST, et al. Pancreatic ductal adenocarcinoma radiology reporting template: consensus statement of the Society of Abdominal Radiology and the American Pancreatic Association. Radiology. 2014; 270(1): 248–260.
  105. Kartalis N, Lindholm TL, Aspelin P, et al. Diffusion-weighted magnetic resonance imaging of pancreas tumours. Eur Radiol. 2009; 19(8): 1981–1990.
  106. Kim JH, Eun HW, Kim YJ, et al. Staging accuracy of MR for pancreatic neuroendocrine tumor and imaging findings according to the tumor grade. Abdom Imaging. 2013; 38(5): 1106–1114.
  107. Krenning EP, Kwekkeboom DJ, Bakker WH, et al. Somatostatin receptor scintigraphy with [111In-DTPA-d-Phe1]- and [123I-Tyr3]-octreotide: the Rotterdam experience with more than 1000 patients. European Journal of Nuclear Medicine. 1993; 20(8): 716–731.
  108. Hubalewska-Dydejczyk A, Fröss-Baron K, Mikołajczak R, et al. 99mTc-EDDA/HYNIC-octreotate scintigraphy, an efficient method for the detection and staging of carcinoid tumours: results of 3 years' experience. Eur J Nucl Med Mol Imaging. 2006; 33(10): 1123–1133.
  109. Kunikowska J, Słodkowski M, Koperski Ł, et al. Radioguided surgery in patient with pancreatic neuroendocrine tumour followed by PET/CT scan as a new approach of complete resection evaluation--case report. Nucl Med Rev Cent East Eur. 2014; 17(2): 110–114.
  110. Reubi JC, Waser B. Concomitant expression of several peptide receptors in neuroendocrine tumours: molecular basis for in vivo multireceptor tumour targeting. Eur J Nucl Med Mol Imaging. 2003; 30(5): 781–793.
  111. Kunikowska J, Królicki L, Pawlak D, et al. Semiquantitative analysis and characterization of physiological biodistribution of (68)Ga-DOTA-TATE PET/CT. Clin Nucl Med. 2012; 37(11): 1052–1057.
  112. Sharma P, Arora S, Dhull VS, et al. Evaluation of (68)Ga-DOTANOC PET/CT imaging in a large exclusive population of pancreatic neuroendocrine tumors. Abdom Imaging. 2015; 40(2): 299–309.
  113. Treglia G, Castaldi P, Rindi G, et al. Diagnostic performance of Gallium-68 somatostatin receptor PET and PET/CT in patients with thoracic and gastroenteropancreatic neuroendocrine tumours: a meta-analysis. Endocrine. 2012; 42(1): 80–87.
  114. Sharma P, Arora S, Mukherjee A, et al. Predictive value of 68Ga-DOTANOC PET/CT in patients with suspicion of neuroendocrine tumors: is its routine use justified? Clin Nucl Med. 2014; 39(1): 37–43.
  115. Rufini V, Baum RP, Castaldi P, et al. Role of PET/CT in the functional imaging of endocrine pancreatic tumors. Abdom Imaging. 2012; 37(6): 1004–1020.
  116. Schmid-Tannwald C, Schmid-Tannwald CM, Morelli JN, et al. Comparison of abdominal MRI with diffusion-weighted imaging to 68Ga-DOTATATE PET/CT in detection of neuroendocrine tumors of the pancreas. Eur J Nucl Med Mol Imaging. 2013; 40(6): 897–907.
  117. Etchebehere EC, de Oliveira Santos A, Gumz B, et al. 68Ga-DOTATATE PET/CT, 99mTc-HYNIC-octreotide SPECT/CT, and whole-body MR imaging in detection of neuroendocrine tumors: a prospective trial. J Nucl Med. 2014; 55(10): 1598–1604.
  118. Wild D, Bomanji JB, Benkert P, et al. Comparison of 68Ga-DOTANOC and 68Ga-DOTATATE PET/CT within patients with gastroenteropancreatic neuroendocrine tumors. J Nucl Med. 2013; 54(3): 364–372.
  119. Ilhan H, Fendler WP, Cyran CC, et al. Impact of (68)Ga-DOTATATE PET/CT on the surgical management of primary neuroendocrine tumors of the pancreas or ileum. Ann Surg Oncol. 2015; 22(1): 164–171.
  120. Haug A, Auernhammer CJ, Wängler B, et al. Intraindividual comparison of 68Ga-DOTA-TATE and 18F-DOPA PET in patients with well-differentiated metastatic neuroendocrine tumours. Eur J Nucl Med Mol Imaging. 2009; 36(5): 765–770.
  121. Ambrosini V, Morigi JJ, Nanni C, et al. Current status of PET imaging of neuroendocrine tumours ([18F]FDOPA, [68Ga]tracers, [11C]/[18F]-HTP). Q J Nucl Med Mol Imaging. 2015; 59(1): 58–69.
  122. Binderup T, Knigge U, Loft A, et al. 18F-fluorodeoxyglucose positron emission tomography predicts survival of patients with neuroendocrine tumors. Clin Cancer Res. 2010; 16(3): 978–985.
  123. Severi S, Nanni O, Bodei L, et al. Role of 18FDG PET/CT in patients treated with 177Lu-DOTATATE for advanced differentiated neuroendocrine tumours. Eur J Nucl Med Mol Imaging. 2013; 40(6): 881–888.
  124. Sowa-Staszczak A, Trofimiuk-Müldner M, Stefańska A, et al. 99mTc Labeled Glucagon-Like Peptide-1-Analogue (99mTc-GLP1) Scintigraphy in the Management of Patients with Occult Insulinoma. PLoS One. 2016; 11(8): e0160714.
  125. Sowa-Staszczak A, Pach D, Mikołajczak R, et al. Glucagon-like peptide-1 receptor imaging with [Lys40(Ahx-HYNIC- 99mTc/EDDA)NH2]-exendin-4 for the detection of insulinoma. Eur J Nucl Med Mol Imaging. 2013; 40(4): 524–531.
  126. Wild D, Christ E, Caplin ME, et al. Glucagon-like peptide-1 versus somatostatin receptor targeting reveals 2 distinct forms of malignant insulinomas. J Nucl Med. 2011; 52(7): 1073–1078.
  127. Benjegård SA, Forssell-Aronsson E, Wängberg B, et al. Intraoperative tumour detection using 111In-DTPA-D-Phe1-octreotide and a scintillation detector. Eur J Nucl Med. 2001; 28(10): 1456–1462.
  128. Hubalewska-Dydejczyk A, Kulig J, Szybinski P, et al. Radio-guided surgery with the use of [99mTc-EDDA/HYNIC]octreotate in intra-operative detection of neuroendocrine tumours of the gastrointestinal tract. Eur J Nucl Med Mol Imaging. 2007; 34(10): 1545–1555.
  129. Sharma P, Arora S, Karunanithi S, et al. Somatostatin receptor based PET/CT imaging with 68Ga-DOTA-Nal3-octreotide for localization of clinically and biochemically suspected insulinoma. Q J Nucl Med Mol Imaging. 2016; 60(1): 69–76.
  130. Jackson JE. Angiography and arterial stimulation venous sampling in the localization of pancreatic neuroendocrine tumours. Best Pract Res Clin Endocrinol Metab. 2005; 19(2): 229–239.
  131. Morera J, Guillaume A, Courtheoux P, et al. Preoperative localization of an insulinoma: selective arterial calcium stimulation test performance. J Endocrinol Invest. 2016; 39(4): 455–463.
  132. Moon JHo, Kim EKy, Khang AhR, et al. An insulinoma with an aberrant feeder from the splenic artery detected by super-selective arterial calcium stimulation with venous sampling. Korean J Intern Med. 2015; 30(1): 118–121.
  133. Jensen RT, Niederle B, Mitry E, et al. Frascati Consensus Conference, European Neuroendocrine Tumor Society. Gastrinoma (duodenal and pancreatic). Neuroendocrinology. 2006; 84(3): 173–182.
  134. Falconi M, Plockinger U, Kwekkeboom DJ, et al. Frascati Consensus Conference, European Neuroendocrine Tumor Society. Well-differentiated pancreatic nonfunctioning tumors/carcinoma. Neuroendocrinology. 2006; 84(3): 196–211.
  135. Grozinsky-Glasberg S, Barak D, Fraenkel M, et al. Peptide receptor radioligand therapy is an effective treatment for the long-term stabilization of malignant gastrinomas. Cancer. 2011; 117(7): 1377–1385.
  136. Sharma J, Duque M, Saif MW. Emerging therapies and latest development in the treatment of unresectable pancreatic neuroendocrine tumors: an update for clinicians. Therap Adv Gastroenterol. 2013; 6(6): 474–490.
  137. Lee LC, Grant CS, Salomao DR, et al. Small, nonfunctioning, asymptomatic pancreatic neuroendocrine tumors (PNETs): role for nonoperative management. Surgery. 2012; 152(6): 965–974.
  138. Watzka FM, Laumen C, Fottner C, et al. Resection strategies for neuroendocrine pancreatic neoplasms. Langenbecks Arch Surg. 2013; 398(3): 431–440.
  139. Al-Kurd A, Chapchay K, Grozinsky-Glasberg S, et al. Laparoscopic resection of pancreatic neuroendocrine tumors. World J Gastroenterol. 2014; 20(17): 4908–4916.
  140. Haugvik SP, Labori KJ, Edwin B, et al. Surgical treatment of sporadic pancreatic neuroendocrine tumors: a state of the art review. ScientificWorldJournal. 2012; 2012: 357475.
  141. Parekh JR, Wang SC, Bergsland EK, et al. Lymph node sampling rates and predictors of nodal metastasis in pancreatic neuroendocrine tumor resections: the UCSF experience with 149 patients. Pancreas. 2012; 41(6): 840–844.
  142. Ramage JK, Ahmed A, Ardill J, et al. UK and Ireland Neuroendocrine Tumour Society. Guidelines for the management of gastroenteropancreatic neuroendocrine (including carcinoid) tumours (NETs). Gut. 2012; 61(1): 6–32.
  143. Shrikhande SV, Sirohi B, Goel M, et al. Pancreatic neuroendocrine tumors. Indian J Gastroenterol. 2013; 32(1): 3–17.
  144. Kulke MH, Anthony LB, Bushnell DL, et al. North American Neuroendocrine Tumor Society (NANETS). NANETS treatment guidelines: well-differentiated neuroendocrine tumors of the stomach and pancreas. Pancreas. 2010; 39(6): 735–752.
  145. Norton JA, Fraker DL, Alexander HR, et al. Value of surgery in patients with negative imaging and sporadic Zollinger-Ellison syndrome. Ann Surg. 2012; 256(3): 509–517.
  146. Bartsch DK, Waldmann J, Fendrich V, et al. Impact of lymphadenectomy on survival after surgery for sporadic gastrinoma. Br J Surg. 2012; 99(9): 1234–1240.
  147. Krampitz GW, Norton JA, Poultsides GA, et al. Lymph nodes and survival in pancreatic neuroendocrine tumors. Arch Surg. 2012; 147(9): 820–827.
  148. Birnbaum DJ, Turrini O, Vigano L, et al. Surgical management of advanced pancreatic neuroendocrine tumors: short-term and long-term results from an international multi-institutional study. Ann Surg Oncol. 2015; 22(3): 1000–1007.
  149. Haugvik SP, Labori KJ, Waage A, et al. Pancreatic surgery with vascular reconstruction in patients with locally advanced pancreatic neuroendocrine tumors. J Gastrointest Surg. 2013; 17(7): 1224–1232.
  150. Krampitz GW, Norton JA. Current management of the Zollinger-Ellison syndrome. Adv Surg. 2013; 47: 59–79.
  151. Su AP, Ke NW, Zhang Yi, et al. Is laparoscopic approach for pancreatic insulinomas safe? Results of a systematic review and meta-analysis. J Surg Res. 2014; 186(1): 126–134.
  152. Mehrabi A, Fischer L, Hafezi M, et al. A systematic review of localization, surgical treatment options, and outcome of insulinoma. Pancreas. 2014; 43(5): 675–686.
  153. Levy MJ, Thompson GB, Topazian MD, et al. US-guided ethanol ablation of insulinomas: a new treatment option. Gastrointest Endosc. 2012; 75(1): 200–206.
  154. Zhao YP, Zhan HX, Zhang TP, et al. Surgical management of patients with insulinomas: Result of 292 cases in a single institution. J Surg Oncol. 2011; 103(2): 169–174.
  155. Gaujoux S, Partelli S, Maire F, et al. Observational study of natural history of small sporadic nonfunctioning pancreatic neuroendocrine tumors. J Clin Endocrinol Metab. 2013; 98(12): 4784–4789.
  156. Massironi S, Rossi RE, Zilli A, et al. A wait-and-watch approach to small pancreatic neuroendocrine tumors: prognosis and survival. Oncotarget. 2016; 7(14): 18978–18983.
  157. Gratian L, Pura J, Dinan M, et al. Impact of extent of surgery on survival in patients with small nonfunctional pancreatic neuroendocrine tumors in the United States. Ann Surg Oncol. 2014; 21(11): 3515–3521.
  158. Crippa S, Partelli S, Zamboni G, et al. Incidental diagnosis as prognostic factor in different tumor stages of nonfunctioning pancreatic endocrine tumors. Surgery. 2014; 155(1): 145–153.
  159. Hashim YM, Trinkaus KM, Linehan DC, et al. Regional lymphadenectomy is indicated in the surgical treatment of pancreatic neuroendocrine tumors (PNETs). Ann Surg. 2014; 259(2): 197–203.
  160. Regenet N, Carrere N, Boulanger G, et al. Is the 2-cm size cutoff relevant for small nonfunctioning pancreatic neuroendocrine tumors: A French multicenter study. Surgery. 2016; 159(3): 901–907.
  161. Cherenfant J, Stocker SJ, Gage MK, et al. Predicting aggressive behavior in nonfunctioning pancreatic neuroendocrine tumors. Surgery. 2013; 154(4): 785–91; discussion 791.
  162. Kuo EJ, Salem RR. Population-level analysis of pancreatic neuroendocrine tumors 2 cm or less in size. Ann Surg Oncol. 2013; 20(9): 2815–2821.
  163. Lombardi M, De Lio N, Funel N, et al. Prognostic factors for pancreatic neuroendocrine neoplasms (pNET) and the risk of small non-functioning pNET. J Endocrinol Invest. 2015; 38(6): 605–613.
  164. Keutgen XM, Nilubol N, Glanville J, et al. Resection of primary tumor site is associated with prolonged survival in metastatic nonfunctioning pancreatic neuroendocrine tumors. Surgery. 2016; 159(1): 311–318.
  165. Milan SA, Yeo CJ. Neuroendocrine tumors of the pancreas. Curr Opin Oncol. 2012; 24(1): 46–55.
  166. Akyildiz HY, Mitchell J, Milas M, et al. Laparoscopic radiofrequency thermal ablation of neuroendocrine hepatic metastases: long-term follow-up. Surgery. 2010; 148(6): 1288–93; discussion 1293.
  167. Watzka FM, Fottner C, Miederer M, et al. Surgical therapy of neuroendocrine neoplasm with hepatic metastasis: patient selection and prognosis. Langenbecks Arch Surg. 2015; 400(3): 349–358.
  168. Bacchetti S, Pasqual EM, Bertozzi S, et al. Curative versus palliative surgical resection of liver metastases in patients with neuroendocrine tumors: a meta-analysis of observational studies. Gland Surg. 2014; 3(4): 243–251.
  169. Pavel M, Baudin E, Couvelard A, et al. Barcelona Consensus Conference participants. ENETS Consensus Guidelines for the management of patients with liver and other distant metastases from neuroendocrine neoplasms of foregut, midgut, hindgut, and unknown primary. Neuroendocrinology. 2012; 95(2): 157–176.
  170. Gaujoux S, Gonen M, Tang L, et al. Synchronous resection of primary and liver metastases for neuroendocrine tumors. Ann Surg Oncol. 2012; 19(13): 4270–4277.
  171. Hüttner FJ, Schneider L, Tarantino I, et al. Palliative resection of the primary tumor in 442 metastasized neuroendocrine tumors of the pancreas: a population-based, propensity score-matched survival analysis. Langenbecks Arch Surg. 2015; 400(6): 715–723.
  172. Kianmanesh R, Ruszniewski P, Rindi G, et al. Palma de Mallorca Consensus Conference Participants. ENETS consensus guidelines for the management of peritoneal carcinomatosis from neuroendocrine tumors. Neuroendocrinology. 2010; 91(4): 333–340.
  173. de Mestier L, Lardière-Deguelte S, Brixi H, et al. Updating the surgical management of peritoneal carcinomatosis in patients with neuroendocrine tumors. Neuroendocrinology. 2015; 101(2): 105–111.
  174. Cidon EU. New therapeutic approaches to metastatic gastroenteropancreatic neuroendocrine tumors: A glimpse into the future. World J Gastrointest Oncol. 2017; 9(1): 4–20.
  175. Wang YZ, Chauhan A, Rau J, et al. Neuroendocrine tumors (NETs) of unknown primary: is early surgical exploration and aggressive debulking justifiable? Chin Clin Oncol. 2016; 5(1): 4.
  176. Sorbye H, Welin S, Langer SW, 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 Oncol. 2013; 24(1): 152–160.
  177. Garcia-Carbonero R, Sorbye H, Baudin E, et al. Vienna Consensus Conference participants. ENETS Consensus Guidelines for High-Grade Gastroenteropancreatic Neuroendocrine Tumors and Neuroendocrine Carcinomas. Neuroendocrinology. 2016; 103(2): 186–194.
  178. Du S, Ni J, Weng L, et al. Aggressive Locoregional Treatment Improves the Outcome of Liver Metastases from Grade 3 Gastroenteropancreatic Neuroendocrine Tumors. Medicine (Baltimore). 2015; 94(34): e1429.
  179. Haugvik SP, Kaemmerer D, Gaujoux S, et al. Pathology and Surgical Treatment of High-Grade Pancreatic Neuroendocrine Carcinoma: an Evolving Landscape. Curr Oncol Rep. 2016; 18(5): 28.
  180. Harada N, Wiersema MJ, Wiersema LM, et al. Endosonography-guided celiac plexus neurolysis. Gastrointest Endosc. 1996; 44(6): 656–662.
  181. Jürgensen C, Schuppan D, Neser F, et al. EUS-guided alcohol ablation of an insulinoma. Gastrointest Endosc. 2006; 63(7): 1059–1062.
  182. Deprez PH, Claessens A, Borbath I, et al. Successful endoscopic ultrasound-guided ethanol ablation of a sporadic insulinoma. Acta Gastroenterol Belg. 2008; 71(3): 333–337.
  183. Muscatiello N, Salcuni A, Macarini L, et al. Treatment of a pancreatic endocrine tumor by ethanol injection (PEI) guided by endoscopic ultrasound. Endoscopy. 2008; 40 Suppl 2: E83–E259.
  184. Andrysiak-Mamos E, Starzynska T, Zochowska E, et al. Therapeutic difficulties in elderly patients with insulinoma. Endocrine Abstracts. 2013.
  185. Pai M, Senturk H, Lakhtakia S, et al. Endoscopic ultrasound guided radiofrequency ablation (EUS-RFA) for cystic neoplasms and neuroendocrine tumours of the pancreas. Gastrointest Endosc 2013; 77: AB143-AB144.
  186. Lakhtakia S, Ramchandani M, Galasso D, et al. EUS-guided radiofrequency ablation for management of pancreatic insulinoma by using a novel needle electrode (with videos). Gastrointest Endosc. 2016; 83(1): 234–239.
  187. Modlin IM, Pavel M, Kidd M, et al. Review article: somatostatin analogues in the treatment of gastroenteropancreatic neuroendocrine (carcinoid) tumours. Aliment Pharmacol Ther. 2010; 31(2): 169–188.
  188. Strosberg JR, Benson AlB, Huynh L, et al. Clinical benefits of above-standard dose of octreotide LAR in patients with neuroendocrine tumors for control of carcinoid syndrome symptoms: a multicenter retrospective chart review study. Oncologist. 2014; 19(9): 930–936.
  189. Al-Efraij K, Aljama MA, Kennecke HF. Association of dose escalation of octreotide long-acting release on clinical symptoms and tumor markers and response among patients with neuroendocrine tumors. Cancer Med. 2015; 4(6): 864–870.
  190. Caplin M, Ruszniewski P, Pavel M, et al. A randomized, double-blind, placebo-Controlled study of Lanreotide Antiproliferative Response in patients with gastroenteropancreatic NeuroEndocrine Tumours (CLARINET). EJC 2013; 49 (supl. 3): S3.
  191. Rinke A, Müller HH, Schade-Brittinger C, et al. PROMID Study Group. Placebo-controlled, double-blind, prospective, randomized study on the effect of octreotide LAR in the control of tumor growth in patients with metastatic neuroendocrine midgut tumors: a report from the PROMID Study Group. J Clin Oncol. 2009; 27(28): 4656–4663.
  192. Martín-Richard M, Massutí B, Pineda E, et al. TTD (Tumores del Tracto Digestivo) Study Group. Antiproliferative effects of lanreotide autogel in patients with progressive, well-differentiated neuroendocrine tumours: a Spanish, multicentre, open-label, single arm phase II study. BMC Cancer. 2013; 13: 427.
  193. Caplin ME, Pavel M, Ćwikła JB, et al. CLARINET Investigators. Lanreotide in metastatic enteropancreatic neuroendocrine tumors. N Engl J Med. 2014; 371(3): 224–233.
  194. Shen C, Shih YCT, Xu Y, et al. Octreotide long-acting repeatable use among elderly patients with carcinoid syndrome and survival outcomes: a population-based analysis. Cancer. 2014; 120(13): 2039–2049.
  195. Pavel M, O'Toole D, Costa F, et al. Vienna Consensus Conference participants. ENETS Consensus Guidelines Update for the Management of Distant Metastatic Disease of Intestinal, Pancreatic, Bronchial Neuroendocrine Neoplasms (NEN) and NEN of Unknown Primary Site. Neuroendocrinology. 2016; 103(2): 172–185.
  196. Rindi G, Falconi M, Klersy C, et al. TNM staging of neoplasms of the endocrine pancreas: results from a large international cohort study. J Natl Cancer Inst. 2012; 104(10): 764–777.
  197. Jann H, Denecke T, Koch M, et al. Impact of octreotide long-acting release on tumour growth control as a first-line treatment in neuroendocrine tumours of pancreatic origin. Neuroendocrinology. 2013; 98(2): 137–143.
  198. Yao JC, Lombard-Bohas C, Baudin E, et al. Daily oral everolimus activity in patients with metastatic pancreatic neuroendocrine tumors after failure of cytotoxic chemotherapy: a phase II trial. J Clin Oncol. 2010; 28(1): 69–76.
  199. Rosiek V, Kunikowska J, Kos-Kudła B. A non-functioning pancreatic neuroendocrine tumour: a case report. Endokrynol Pol. 2012; 63(1): 59–64.
  200. Ito T, Igarashi H, Jensen RT. Pancreatic neuroendocrine tumors: clinical features, diagnosis and medical treatment: Advances. Best Pract Res Clin Gastroenterol 2012;26:737-753.
  201. Ito T, Igarashi H, Jensen RT. Therapy of metastatic pancreatic neuroendocrine tumors (pNETs): recent insights and advances. J Gastroenterol. 2012; 47(9): 941–960.
  202. Tabarin A, Goichot B. French Endocrine Society. Treatment: symptomatic treatment of hypoglycaemia. Ann Endocrinol (Paris). 2013; 74(3): 196–199.
  203. Baudin E, Caron P, Lombard-Bohas C, et al. Société française d’endocrinologie, Groupe d’étude des tumeurs endocrines. Malignant insulinoma: recommendations for characterisation and treatment. Ann Endocrinol (Paris). 2013; 74(5-6): 523–533.
  204. Ferrer-García JC, Iranzo González-Cruz V, Navas-DeSolís S, et al. Management of malignant insulinoma. Clin Transl Oncol. 2013; 15(9): 725–731.
  205. Jawiarczyk A, Bolanowski M, Syrycka J, et al. Effective therapy of insulinoma by using long-acting somatostatin analogue. A case report and literature review. Exp Clin Endocrinol Diabetes. 2012; 120(2): 68–72.
  206. Oberg KE, Reubi JC, Kwekkeboom DJ, et al. Role of somatostatins in gastroenteropancreatic neuroendocrine tumor development and therapy. Gastroenterology. 2010; 139(3): 742–53, 753.e1.
  207. Rosiek V, Kos-Kudła B. Guidelines for the management of pancreatic neuroendocrine tumors. Gastroenterol Prakt 2010; 2: 28–34.
  208. Bernard V, Lombard-Bohas C, Taquet MC, et al. French Group of Endocrine Tumors. Efficacy of everolimus in patients with metastatic insulinoma and refractory hypoglycemia. Eur J Endocrinol. 2013; 168(5): 665–674.
  209. Srirajaskanthan R, McStay M, Toumpanakis C, et al. Parathyroid hormone-related peptide-secreting pancreatic neuroendocrine tumours: case series and literature review. Neuroendocrinology. 2009; 89(1): 48–55.
  210. Yao JC, Shah MH, Ito T, et al. RAD001 in Advanced Neuroendocrine Tumors, Third Trial (RADIANT-3) Study Group. Everolimus for advanced pancreatic neuroendocrine tumors. N Engl J Med. 2011; 364(6): 514–523.
  211. Raymond E, Dahan L, Raoul JL, et al. Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. N Engl J Med. 2011; 364(6): 501–513.
  212. Lombard-Bohas C, Yao JC, Hobday T, et al. Impact of prior chemotherapy use on the efficacy of everolimus in patients with advanced pancreatic neuroendocrine tumors: a subgroup analysis of the phase III RADIANT-3 trial. Pancreas. 2015; 44(2): 181–189.
  213. Bernard V, Lombard-Bohas C, Taquet MC, et al. French Group of Endocrine Tumors. Efficacy of everolimus in patients with metastatic insulinoma and refractory hypoglycemia. Eur J Endocrinol. 2013; 168(5): 665–674.
  214. Kulke MH, Bergsland EK, Yao JC. Glycemic control in patients with insulinoma treated with everolimus. N Engl J Med. 2009; 360(2): 195–197.
  215. Pavel ME, Hainsworth JD, Baudin E, et al. RADIANT-2 Study Group. Everolimus plus octreotide long-acting repeatable for the treatment of advanced neuroendocrine tumours associated with carcinoid syndrome (RADIANT-2): a randomised, placebo-controlled, phase 3 study. Lancet. 2011; 378(9808): 2005–2012.
  216. Yao JC, Fazio N, Singh S, et al. RAD001 in Advanced Neuroendocrine Tumours, Fourth Trial (RADIANT-4) Study Group. Everolimus for the treatment of advanced, non-functional neuroendocrine tumours of the lung or gastrointestinal tract (RADIANT-4): a randomised, placebo-controlled, phase 3 study. Lancet. 2016; 387(10022): 968–977.
  217. Chen J, Wang C, Han J, et al. Therapeutic effect of sunitinib malate and its influence on blood glucose concentrations in a patient with metastatic insulinoma. Expert Rev Anticancer Ther. 2013; 13(6): 737–743.
  218. Kulke et al. A Randomized Open-label Phase II Study of Everolimus Alone or in Combination with Pasireotide LAR in Advanced, Progressive Pancreatic Neuroendocrine Tumors (pNET): COOPERATE-2 Trial; presented at the 12th Annual ENETS Conference for the Diagnosis and Treatment of Neuroendocrine Tumor Disease, March 11-13, 2015 Barcelona, Spain
  219. Panzuto F, Rinzivillo M, Fazio N, et al. Real-World Study of Everolimus in Advanced Progressive Neuroendocrine Tumors. The Oncologist. 2015;20(5):570.
  220. Alonso-Gordoa T, Díez JJ, Molina J, et al. An Overview on the Sequential Treatment of Pancreatic Neuroendocrine Tumors (pNETs). Rare Cancers Ther. 2015; 3: 13–33.
  221. Grande E. Sequential treatment in disseminated well- and intermediate-differentiated pancreatic neuroendocrine tumors: Common sense or low rationale? World J Clin Oncol. 2016; 7(2): 149–154.
  222. Hobday TJ, Yin J, Pettinger A, et al. Multicenter prospective phase II trial of bevacizumab (bev) for progressive pancreatic neuroendocrine tumor (PNET). J Clin Oncol. 2015; 33 (Suppl.): abstr 4096.
  223. Castellano D, Capdevila J, Sastre J, et al. Sorafenib and bevacizumab combination targeted therapy in advanced neuroendocrine tumour: a phase II study of Spanish Neuroendocrine Tumour Group (GETNE0801). Eur J Cancer. 2013; 49(18): 3780–3787.
  224. Grande E, Capdevila J, Castellano D, et al. Pazopanib in pretreated advanced neuroendocrine tumors: a phase II, open-label trial of the Spanish Task Force Group for Neuroendocrine Tumors (GETNE). Ann Oncol. 2015; 26(9): 1987–1993.
  225. Okusaka T, Ueno H, Morizane C, et al. Cytotoxic chemotherapy for pancreatic neuroendocrine tumors. J Hepatobiliary Pancreat Sci. 2015; 22(8): 628–633.
  226. Strosberg JR, Fine RL, Choi J, et al. First-line chemotherapy with capecitabine and temozolomide in patients with metastatic pancreatic endocrine carcinomas. Cancer. 2011; 117(2): 268–275.
  227. Moertel CG, Hanley JA, Johnson LA. Streptozocin alone compared with streptozocin plus fluorouracil in the treatment of advanced islet-cell carcinoma. N Engl J Med. 1980; 303(21): 1189–1194.
  228. Dilz LM, Denecke T, Steffen IG, et al. Streptozocin/5-fluorouracil chemotherapy is associated with durable response in patients with advanced pancreatic neuroendocrine tumours. Eur J Cancer. 2015; 51(10): 1253–1262.
  229. Kulke MH, Hornick JL, Frauenhoffer C, et al. O6-methylguanine DNA methyltransferase deficiency and response to temozolomide-based therapy in patients with neuroendocrine tumors. Clin Cancer Res. 2009; 15(1): 338–345.
  230. Cives M, Ghayouri M, Morse B, et al. Analysis of potential response predictors to capecitabine/temozolomide in metastatic pancreatic neuroendocrine tumors. Endocr Relat Cancer. 2016; 23(9): 759–767.
  231. Cros J, Hentic O, Rebours V, et al. MGMT expression predicts response to temozolomide in pancreatic neuroendocrine tumors. Endocr Relat Cancer. 2016; 23(8): 625–633.
  232. Cassier PA, Walter T, Eymard B, et al. Gemcitabine and oxaliplatin combination chemotherapy for metastatic well-differentiated neuroendocrine carcinomas: a single-center experience. Cancer. 2009; 115(15): 3392–3399.
  233. Spada F, Antonuzzo L, Marconcini R, et al. Oxaliplatin-Based Chemotherapy in Advanced Neuroendocrine Tumors: Clinical Outcomes and Preliminary Correlation with Biological Factors. Neuroendocrinology. 2016; 103(6): 806–814.
  234. Sorbye H, Welin S, Langer SW, 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 Oncol. 2013; 24(1): 152–160.
  235. Welin S, Sorbye H, Sebjornsen S, et al. Clinical effect of temozolomide-based chemotherapy in poorly differentiated endocrine carcinoma after progression on first-line chemotherapy. Cancer. 2011; 117(20): 4617–4622.
  236. Bajetta E, Catena L, Procopio G, et al. Are capecitabine and oxaliplatin (XELOX) suitable treatments for progressing low-grade and high-grade neuroendocrine tumours? Cancer Chemother Pharmacol. 2007; 59(5): 637–642.
  237. Hadoux J, Malka D, Planchard D, et al. Post-first-line FOLFOX chemotherapy for grade 3 neuroendocrine carcinoma. Endocr Relat Cancer. 2015; 22(3): 289–298.
  238. Hentic O, Hammel P, Couvelard A, et al. FOLFIRI regimen: an effective second-line chemotherapy after failure of etoposide-platinum combination in patients with neuroendocrine carcinomas grade 3. Endocr Relat Cancer. 2012; 19(6): 751–757.
  239. De Jong M, Bakker WH, Breeman WA, et al. Pre-clinical comparison of [DTPA0] octreotide, [DTPA0,Tyr3] octreotide and [DOTA0,Tyr3] octreotide as carriers for somatostatin receptor-targeted scintigraphy and radionuclide therapy. Int J Cancer. 1998; 75(3): 406–411.
  240. Krenning EP, Kwekkeboom DJ, Valkema R, et al. Peptide receptor radionuclide therapy. Ann N Y Acad Sci. 2004; 1014: 234–245.
  241. Bodei L, Mueller-Brand J, Baum RP, et al. The joint IAEA, EANM, and SNMMI practical guidance on peptide receptor radionuclide therapy (PRRNT) in neuroendocrine tumours. Eur J Nucl Med Mol Imaging. 2013; 40(5): 800–816.
  242. Villard L, Romer A, Marincek N, et al. Cohort study of somatostatin-based radiopeptide therapy with [(90)Y-DOTA]-TOC versus [(90)Y-DOTA]-TOC plus [(177)Lu-DOTA]-TOC in neuroendocrine cancers. J Clin Oncol. 2012; 30(10): 1100–1106.
  243. Kaltsas G, Caplin M, Davies P, et al. all other Antibes Consensus Conference participants, DUST Investigators, German Mouse Clinic Consortium. Effectiveness and side-effects of peptide receptor radionuclide therapy for neuroendocrine neoplasms in Germany: A multi-institutional registry study with prospective follow-up. Eur J Cancer. 2016; 58(6): 41–51.
  244. Ezziddin S, Khalaf F, Vanezi M, et al. Outcome of peptide receptor radionuclide therapy with 177Lu-octreotate in advanced grade 1/2 pancreatic neuroendocrine tumours. Eur J Nucl Med Mol Imaging. 2014; 41(5): 925–933.
  245. Sansovini M, Severi S, Ianniello A, et al. Long-term follow-up and role of FDG PET in advanced pancreatic neuroendocrine patients treated with (177)Lu-D OTATATE. Eur J Nucl Med Mol Imaging. 2017; 44(3): 490–499.
  246. Sowa-Staszczak A, Pach D, Chrzan R, et al. Peptide receptor radionuclide therapy as a potential tool for neoadjuvant therapy in patients with inoperable neuroendocrine tumours (NETs). Eur J Nucl Med Mol Imaging. 2011; 38(9): 1669–1674.
  247. Stoeltzing O, Loss M, Huber E, et al. Staged surgery with neoadjuvant 90Y-DOTATOC therapy for down-sizing synchronous bilobular hepatic metastases from a neuroendocrine pancreatic tumor. Langenbecks Arch Surg. 2010; 395(2): 185–192.
  248. van Schaik E, van Vliet EI, Feelders RA, et al. Improved control of severe hypoglycemia in patients with malignant insulinomas by peptide receptor radionuclide therapy. J Clin Endocrinol Metab. 2011; 96(11): 3381–3389.
  249. de Herder WW, van Schaik E, Kwekkeboom D, et al. New therapeutic options for metastatic malignant insulinomas. Clin Endocrinol (Oxf). 2011; 75(3): 277–284.
  250. Pach D, Sowa-Staszczak A, Kunikowska J, et al. Repeated cycles of peptide receptor radionuclide therapy (PRRT)--results and side-effects of the radioisotope 90Y-DOTA TATE, 177Lu-DOTA TATE or 90Y/177Lu-DOTA TATE therapy in patients with disseminated NET. Radiother Oncol. 2012; 102(1): 45–50.
  251. Paprottka PM, Hoffmann RT, Haug A, et al. Radioembolization of symptomatic, unresectable neuroendocrine hepatic metastases using yttrium-90 microspheres. Cardiovasc Intervent Radiol. 2012; 35(2): 334–342.
  252. OCEBM Levels of Evidence Working Group*. “The Oxford 2011 Levels of Evidence”. Oxford Centre for Evidence-Based Medicine. http://www.cebm.net/ index.aspx?o=5653.

Regulations

Important: This website uses cookies. More >>

The cookies allow us to identify your computer and find out details about your last visit. They remembering whether you've visited the site before, so that you remain logged in - or to help us work out how many new website visitors we get each month. Most internet browsers accept cookies automatically, but you can change the settings of your browser to erase cookies or prevent automatic acceptance if you prefer.

Via MedicaWydawcą jest  VM Media Group sp. z o.o., Grupa Via Medica, ul. Świętokrzyska 73, 80–180 Gdańsk

tel.:+48 58 320 94 94, faks:+48 58 320 94 60, e-mail:  viamedica@viamedica.pl