Therapy of metastatic pancreatic neuroendocrine tumors (pNETs): recent insights and advances
详细信息    查看全文
  • 作者:Tetsuhide Ito (1)
    Hisato Igarashi (1)
    Robert T. Jensen (2) robertj@bdg10.niddk.nih.gov
  • 关键词:Neuroendocrine tumor – ; Pancreatic endocrine tumor – ; Liver metastases – ; Gastrinoma – ; Insulinoma – ; Surgery – ; Chemotherapy – ; Streptozotocin – ; Everolimus – ; Sunitinib – ; Somatostatin – ; Octreotide – ; Lanreotide – ; Hepatic transarterial embolization – ; Liver transplantation – ; Peptide receptor radionuclide therapy – ; Somatostatin receptor – ; SIRT – ; Chemoembolization – ; Unresectable liver metastases
  • 刊名:Journal of Gastroenterology
  • 出版年:2012
  • 出版时间:September 2012
  • 年:2012
  • 卷:47
  • 期:9
  • 页码:941-960
  • 全文大小:474.2 KB
  • 参考文献:1. Modlin IM, Oberg K, Chung DC, et al. Gastroenteropancreatic neuroendocrine tumours. Lancet Oncol. 2008;9:61–72.
    2. Jiao Y, Shi C, Edil BH, et al. DAXX/ATRX, MEN1, and mTOR pathway genes are frequently altered in pancreatic neuroendocrine tumors. Science. 2011;331:1199–203.
    3. Metz DC, Jensen RT. Gastrointestinal neuroendocrine tumors: pancreatic endocrine tumors. Gastroenterology. 2008;135:1469–92.
    4. de Wilde RF, Edil BH, Hruban RH, Maitra A. Well-differentiated pancreatic neuroendocrine tumors: from genetics to therapy. Nat Rev Gastroenterol Hepatol. 2012;9:199–208.
    5. Capurso G, Festa S, Valente R, et al. Molecular pathology and genetics of pancreatic endocrine tumours. J Mol Endocrinol. 2012;49:R37–50.
    6. Oberg K. Neuroendocrine tumors of the digestive tract: impact of new classifications and new agents on therapeutic approaches. Curr Opin Oncol. 2012;24:433–40.
    7. Oberg K. Pancreatic endocrine tumors. Semin Oncol. 2010;37:594–618.
    8. 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;104:764–77.
    9. Klimstra DS, Modlin IR, Coppola D, Lloyd RV, Suster S. The pathologic classification of neuroendocrine tumors: a review of nomenclature, grading, and staging systems. Pancreas. 2010;39:707–12.
    10. Klimstra DS, Modlin IR, Adsay NV, et al. Pathology reporting of neuroendocrine tumors: application of the Delphic consensus process to the development of a minimum pathology data set. Am J Surg Pathol. 2010;34:300–13.
    11. Pape UF, Jann H, Muller-Nordhorn J, et al. Prognostic relevance of a novel TNM classification system for upper gastroenteropancreatic neuroendocrine tumors. Cancer. 2008;113:256–65.
    12. Kloppel G. Tumour biology and histopathology of neuroendocrine tumours. Best Pract Res Clin Endocrinol Metab. 2007;21:15–31.
    13. Bettini R, Boninsegna L, Mantovani W, et al. Prognostic factors at diagnosis and value of WHO classification in a mono-institutional series of 180 non-functioning pancreatic endocrine tumours. Ann Oncol. 2008;19:903–8.
    14. Sundin A, Vullierme MP, Kaltsas G, Plockinger U. ENETS guidelines for the standards of care in patients with neuroendocrine tumours: radiological examinations in patients with neuroendocrine tumours. Neuroendocrinology. 2009;90:167–83.
    15. Oberg K. Gallium-68 somatostatin receptor PET/CT: Is it time to replace (111)Indium DTPA octreotide for patients with neuroendocrine tumors? Endocrine. 2012;42:3–4.
    16. Jensen RT, Delle Fave G. Promising advances in the treatment of malignant pancreatic endocrine tumors. N Engl J Med. 2011;364:564–5.
    17. Yao JC, Shah MH, Ito T, et al. Everolimus for advanced pancreatic neuroendocrine tumors. N Engl J Med. 2011;364:514–23.
    18. Raymond E, Dahan L, Raoul JL, et al. Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. N Engl J Med. 2011;364:501–13.
    19. Pavel M. Translation of molecular pathways into clinical trials of neuroendocrine tumors. Neuroendocrinology. 2012 [Epub ahead of print].
    20. Halperin DM, Kulke MH. Management of pancreatic neuroendocrine tumors. Gastroenterol Clin North Am. 2012;41:119–31.
    21. Harring TR, Nguyen NT, Goss JA, O’Mahony CA. Treatment of liver metastases in patients with neuroendocrine tumors: a comprehensive review. Int J Hepatol. 2011;2011:154541.
    22. van Vliet EI, Teunissen JJ, Kam BL, de Jong M, Krenning EP, Kwekkeboom DJ. Treatment of gastroenteropancreatic neuroendocrine tumors with peptide receptor radionuclide therapy. Neuroendocrinology. 2012 [Epub ahead of print].
    23. Kwekkeboom DJ, Kam BL, Van Essen M, et al. Somatostatin-receptor-based imaging and therapy of gastroenteropancreatic neuroendocrine tumors. Endocr Relat Cancer. 2010;17:R53–73.
    24. Kwekkeboom DJ, Krenning EP, Scheidhauer K, et al. ENETS consensus guidelines for the standards of care in neuroendocrine tumors: somatostatin receptor imaging with (111) In-pentetreotide. Neuroendocrinology. 2009;90:184–9.
    25. Jensen RT, Cadiot G, Brandi ML, et al. ENETS consensus guidelines for the management of patients with digestive neuroendocrine neoplasms: functional pancreatic endocrine tumor syndromes. Neuroendocrinology. 2012;95:98–119.
    26. Kulke MH, Anthony LB, Bushnell DL, et al. NANETS treatment guidelines: well-differentiated neuroendocrine tumors of the stomach and pancreas. Pancreas. 2010;39:735–52.
    27. Oberg K, Jelic S. Neuroendocrine gastroenteropancreatic tumors: ESMO clinical recommendations for diagnosis, treatment and follow-up. Ann Oncol. 2009;20 Suppl 4:147–9.
    28. Janson ET, Sorbye H, Welin S, et al. Nordic guidelines 2010 for diagnosis and treatment of gastroenteropancreatic neuroendocrine tumours. Acta Oncol. 2010;49:740–56.
    29. Pavel M, Baudin E, Couvelard A, et al. ENETS consensus guidelines for the management of patients with liver and other distant metastases from neuroendocrine neoplasms of foregut, midgut, hindgut, and unknown primary. Neuroendocrinology. 2012;95:157–76.
    30. Steinmuller T, Kianmanesh R, Falconi M, et al. Consensus guidelines for the management of patients with liver metastases from digestive (neuro) endocrine tumors: foregut, midgut, hindgut, and unknown primary. Neuroendocrinology. 2008;87:47–62.
    31. Pavel ME, Hainsworth JD, Baudin E, et al. Everolimus plus octreotide long-acting repeatable for the treatment of advanced neuroendocrine tumours associated with carcinoid syndrome (RADIANT-2): a randomised, placebo-controlled, phase 3 study. Lancet. 2011;378:2005–12.
    32. Kos-Kudla B, O’Toole D, Falconi M, et al. ENETS consensus guidelines for the management of bone and lung metastases from neuroendocrine tumors. Neuroendocrinology. 2010;91:341–50.
    33. Kianmanesh R, Ruszniewski P, Rindi G, et al. ENETS consensus guidelines for the management of peritoneal carcinomatosis from neuroendocrine tumors. Neuroendocrinology. 2010;91:333–40.
    34. Krampitz GW, Norton JA, Poultsides GA, Visser B, Sun L, Jensen RT. Lymph nodes and survival in duodenal and pancreatic neuroendocrine tumors. Arch. Surg. 2012;(in press) (abstr).
    35. Kulke MH, Bendell J, Kvols L, Picus J, Pommier R, Yao J. Evolving diagnostic and treatment strategies for pancreatic neuroendocrine tumors. J Hematol Oncol. 2011;4:29–37.
    36. Kimura W, Kuroda A, Morioka Y. Clinical pathology of endocrine tumors of the pancreas. Analysis of autopsy cases. Dig Dis Sci. 1991;36:933–42.
    37. Ito T, Sasano H, Tanaka M, et al. Epidemiological study of gastroenteropancreatic neuroendocrine tumors in Japan. J Gastroenterol. 2010;45:234–43.
    38. Yao JC, Hassan M, Phan A, et al. One hundred years after “carcinoid”: epidemiology of and prognostic factors for neuroendocrine tumors in 35,825 cases in the United States. J Clin Oncol. 2008;26:3063–72.
    39. Haynes AB, Deshpande V, Ingkakul T, et al. Implications of incidentally discovered, nonfunctioning pancreatic endocrine tumors: short-term and long-term patient outcomes. Arch Surg. 2011;146:534–8.
    40. Gullo L, Migliori M, Falconi M, et al. Nonfunctioning pancreatic endocrine tumors: a multicenter clinical study. Am J Gastroenterol. 2003;98:2435–9.
    41. Jensen RT. Endocrine neoplasms of the pancreas. In: Yamada T, Alpers DH, Kalloo AN, Kaplowitz N, Owyang C, editors. Textbook of gastroenterology. Oxford, England: Wiley-Blackwell; 2009. p. 1875–920.
    42. Halfdanarson TR, Rabe KG, Rubin J, Petersen GM. Pancreatic neuroendocrine tumors (PNETs): incidence, prognosis and recent trend toward improved survival. Ann Oncol. 2008;10:1727–33.
    43. Zerbi A, Falconi M, Rindi G, et al. Clinicopathological features of pancreatic endocrine tumors: a prospective multicenter study in Italy of 297 sporadic cases. Am J Gastroenterol. 2010;105:1421–9.
    44. Lepage C, Rachet B, Coleman MP. Survival from malignant digestive endocrine tumors in England and Wales: a population-based study. Gastroenterology. 2007;132:899–904.
    45. Yao JC, Eisner MP, Leary C, et al. Population-based study of islet cell carcinoma. Ann Surg Oncol. 2007;14:3492–500.
    46. Jensen RT. Natural history of digestive endocrine tumors. In: Mignon M, Colombel JF, editors. Recent advances in pathophysiology and management of inflammatory bowel diseases and digestive endocrine tumors. Paris, France: John Libbey Eurotext Publishing Co.; 1999. p. 192–219.
    47. Yu F, Venzon DJ, Serrano J, et al. Prospective study of the clinical course, prognostic factors and survival in patients with longstanding Zollinger-Ellison syndrome. J Clin Oncol. 1999;17:615–30.
    48. Weber HC, Venzon DJ, Lin JT, et al. Determinants of metastatic rate and survival in patients with Zollinger-Ellison syndrome: a prospective long-term study. Gastroenterology. 1995;108:1637–49.
    49. Sutliff VE, Doppman JL, Gibril F, et al. Growth of newly diagnosed, untreated metastatic gastrinomas and predictors of growth patterns. J Clin Oncol. 1997;15:2420–31.
    50. Durante C, Boukheris H, Dromain C, et al. Prognostic factors influencing survival from metastatic (stage IV) gastroenteropancreatic well-differentiated endocrine carcinoma. Endocr Relat Cancer. 2009;16:585–97.
    51. Panzuto F, Nasoni S, Falconi M, et al. Prognostic factors and survival in endocrine tumor patients:comparison between gastrointestinal and pancreatic localization. Endocr Relat Cancer. 2005;12:1083–92.
    52. Sarmiento JM, Que FG. Hepatic surgery for metastases from neuroendocrine tumors. Surg Oncol Clin North Am. 2003;12:231–42.
    53. Norton JA, Warren RS, Kelly MG, Zurek MB, Jensen RT. Aggressive surgery for metastatic liver neuroendocrine tumors. Surgery. 2003;134:1057–65.
    54. Solorzano CC, Lee JE, Pisters PW, et al. Nonfunctioning islet cell carcinoma of the pancreas: survival results in a contemporary series of 163 patients. Surgery. 2001;130:1078–85.
    55. Helle KB. Regulatory peptides from chromogranin A and secretogranin II. Cell Mol Neurobiol. 2010;30:1145–6.
    56. Niederle MB, Hackl M, Kaserer K, Niederle B. Gastroenteropancreatic neuroendocrine tumours: the current incidence and staging based on the WHO and European Neuroendocrine Tumour Society classification: an analysis based on prospectively collected parameters. Endocr Relat Cancer. 2010;17:909–18.
    57. Bosman FT, Carneiro F, Hruban RH, Theise ND. WHO World Health Organization classification of tumors and genetics of the digestive system. Lyon, France: IARC press; 2010.
    58. Rindi G, Kloppel G, Alhman H, et al. TNM staging of foregut (neuro)endocrine tumors: a consensus proposal including a grading system. Virchows Arch. 2006;449:395–401.
    59. Edge SB, Byrd DR, Compton CC, Fritz AG, Greene FL, Trotti A. AJCC Cancer Staging Manual. New York: Springer; 2010.
    60. 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;29:2372–7.
    61. Pomianowska E, Gladhaug IP, Grzyb K, et al. Survival following resection of pancreatic endocrine tumors: importance of R-status and the WHO and TNM classification systems. Scand J Gastroenterol. 2010;45:971–9.
    62. Scarpa A, Mantovani W, Capelli P, et al. Pancreatic endocrine tumors: improved TNM staging and histopathological grading permit a clinically efficient prognostic stratification of patients. Mod Pathol. 2010;23:824–33.
    63. Kloppel G. Classification and pathology of gastroenteropancreatic neuroendocrine neoplasms. Endocr Relat Cancer. 2011;18 Suppl 1:S1–16.
    64. Ekeblad S, Skogseid B, Dunder K, Oberg K, Eriksson B. Prognostic factors and survival in 324 patients with pancreatic endocrine tumor treated at a single institution. Clin Cancer Res. 2008;14:7798–803.
    65. Tsuchiya A, Koizumi M, Ohtani H. World Health Organization Classification (2004)-based reevaluation of 95 nonfunctioning “malignant” pancreatic endocrine tumors reported in Japan. Surg Today. 2009;39:500–9.
    66. Hentic O, Couvelard A, Rebours V, et al. Ki-67 index, tumor differentiation, and extent of liver involvement are independent prognostic factors in patients with liver metastases of digestive endocrine carcinomas. Endocr Relat Cancer. 2011;18:51–9.
    67. Krausz Y, Freedman N, Rubinstein R, et al. (68)Ga-DOTA-NOC PET/CT imaging of neuroendocrine tumors: comparison with (111)In-DTPA-Octreotide (OctreoScan(R)). Mol Imaging Biol. 2011;13:583–93.
    68. Nilsson O, Van Cutsem E, Delle Fave G, et al. Poorly differentiated carcinomas of the foregut (gastric, duodenal and pancreatic). Neuroendocrinology. 2006;84:212–5.
    69. Kloppel G, Couvelard A, Perren A, et al. ENETS guidelines for the standards of care in patients with neuroendocrine tumors: towards a standardized approach to the diagnosis of gastroenteropancreatic neuroendocrine tumors and their prognostic stratification. Neuroendocrinology. 2009;90:166.
    70. Krudy AG, Doppman JL, Jensen RT, et al. Localization of islet cell tumors by dynamic CT: comparison with plain CT, arteriography, sonography and venous sampling. Am J Roentgenol. 1984;143:585–9.
    71. Gibril F, Reynolds JC, Doppman JL, et al. Somatostatin receptor scintigraphy: its sensitivity compared with that of other imaging methods in detecting primary and metastatic gastrinomas: a prospective study. Ann Intern Med. 1996;125:26–34.
    72. Gibril F, Jensen RT. Diagnostic uses of radiolabelled somatostatin-receptor analogues in gastroenteropancreatic endocrine tumors. Dig Liver Dis. 2004;36:S106–20.
    73. Ruf J, Heuck F, Schiefer J, et al. Impact of Multiphase 68Ga-DOTATOC-PET/CT on therapy management in patients with neuroendocrine tumors. Neuroendocrinology. 2010;91:101–9.
    74. Buchmann I, Henze M, Engelbrecht S, et al. Comparison of 68Ga-DOTATOC PET and 111In-DTPAOC (Octreoscan) SPECT in patients with neuroendocrine tumours. Eur J Nucl Med Mol Imaging. 2007;34:1617–26.
    75. Putzer D, Gabriel M, Henninger B, et al. Bone metastases in patients with neuroendocrine tumor: 68Ga-DOTA-Tyr3-octreotide PET in comparison to CT and bone scintigraphy. J Nucl Med. 2009;50:1214–21.
    76. Termanini B, Gibril F, Reynolds JC, et al. Value of somatostatin receptor scintigraphy: a prospective study in gastrinoma of its effect on clinical management. Gastroenterology. 1997;112:335–47.
    77. Schillaci O, Spanu A, Scopinaro F, et al. Somatostatin receptor scintigraphy with 111In-pentetreotide in non-functioning gastroenteropancreatic neuroendocrine tumors. Int J Oncol. 2003;23:1687–95.
    78. Gibril F, Doppman JL, Reynolds JC, et al. Bone metastases in patients with gastrinomas: a prospective study of bone scanning, somatostatin receptor scanning, and MRI in their detection, their frequency, location and effect of their detection on management. J Clin Oncol. 1998;16:1040–53.
    79. Imamura M. Recent standardization of treatment strategy for pancreatic neuroendocrine tumors. World J Gastroenterol. 2010;16:4519–25.
    80. Doppman JL, Miller DL, Chang R, et al. Gastrinomas: localization by means of selective intraarterial injection of secretin. Radiology. 1990;174:25–9.
    81. Gibril F, Doppman JL, Chang R, Weber HC, Termanini B, Jensen RT. Metastatic gastrinomas: localization with selective arterial injection of secretin. Radiology. 1996;198:77–84.
    82. Morganstein DL, Lewis DH, Jackson J, et al. The role of arterial stimulation and simultaneous venous sampling in addition to cross-sectional imaging for localisation of biochemically proven insulinoma. Eur Radiol. 2009;19:2467–73.
    83. Strosberg JR, Coppola D, Klimstra DS, et al. The NANETS consensus guidelines for the diagnosis and management of poorly differentiated (high-grade) extrapulmonary neuroendocrine carcinomas. Pancreas. 2010;39:799–800.
    84. Iwasa S, Morizane C, Okusaka T, et al. Cisplatin and etoposide as first-line chemotherapy for poorly differentiated neuroendocrine carcinoma of the hepatobiliary tract and pancreas. Jpn J Clin Oncol. 2010;40:313–8.
    85. Osefo N, Ito T, Jensen RT. Gastric acid hypersecretory states: recent insights and advances. Curr Gastroenterol Rep. 2009;11:433–41.
    86. Jensen RT, Niederle B, Mitry E, et al. Gastrinoma (duodenal and pancreatic). Neuroendocrinology. 2006;84:173–82.
    87. de Herder WW, van SE, Kwekkeboom D, Feelders RA. New therapeutic options for metastatic malignant insulinomas. Clin Endocrinol (Oxf). 2011;75:277–84.
    88. Vezzosi D, Bennet A, Rochaix P, et al. Octreotide in insulinoma patients: efficacy on hypoglycemia, relationships with Octreoscan scintigraphy and immunostaining with anti-sst2A and anti-sst5 antibodies. Eur J Endocrinol. 2005;152:757–67.
    89. Vezzosi D, Bennet A, Courbon F, Caron P. Short- and long-term somatostatin analogue treatment in patients with hypoglycaemia related to endogenous hyperinsulinism. Clin Endocrinol (Oxf). 2008;68:904–11.
    90. Vleggaar FP, Bij de Vaate EA, Valk GD, Leguit RJ, Siersema PD. Endoscopic ultrasound-guided ethanol ablation of a symptomatic sporadic insulinoma. Endoscopy. 2011;43 Suppl 2 UCTN:E328–9.
    91. Levy MJ, Thompson GB, Topazian MD, Callstrom MR, Grant CS, Vella A. US-guided ethanol ablation of insulinomas: a new treatment option. Gastrointest Endosc. 2012;75:200–6.
    92. Jurgensen C, Schuppan D, Neser F, Ernstberger J, Junghans U, Stolzel U. EUS-guided alcohol ablation of an insulinoma. Gastrointest Endosc. 2006;63:1059–62.
    93. Kvols LK, Turaga KK, Strosberg J, Choi J. Role of interventional radiology in the treatment of patients with neuroendocrine metastases in the liver. J Natl Compr Canc Netw. 2009;7:765–72.
    94. Fiebrich HB, Siemerink EJ, Brouwers AH, et al. Everolimus induces rapid plasma glucose normalization in insulinoma patients by effects on tumor as well as normal tissues. Oncologist. 2011;16:783–7.
    95. Kulke MH, Bergsland EK, Yao JC. Glycemic control in patients with insulinoma treated with everolimus. N Engl J Med. 2009;360:195–7.
    96. Ong GS, Henley DE, Hurley D, Turner JH, Claringbold PG, Fegan PG. Therapies for the medical management of persistent hypoglycaemia in two cases of inoperable malignant insulinoma. Eur J Endocrinol. 2010;162:1001–8.
    97. Chandra P, Yarandi SS, Khazai N, Jacobs S, Umpierrez GE. Management of intractable hypoglycemia with Yttirum-90 radioembolization in a patient with malignant insulinoma. Am J Med Sci. 2010;340:414–7.
    98. Falconi M, Bartsch DK, Eriksson B, et al. ENETS consensus guidelines for the management of patients with digestive neuroendocrine neoplasms of the digestive system: well-differentiated pancreatic non-functioning tumors. Neuroendocrinology. 2012;95:120–34.
    99. Falconi M, Bettini R, Boninsegna L, Crippa S, Butturini G, Pederzoli P. Surgical strategy in the treatment of pancreatic neuroendocrine tumors. JOP. 2006;7:150–6.
    100. Carty SE, Jensen RT, Norton JA. Prospective study of aggressive resection of metastatic pancreatic endocrine tumors. Surgery. 1992;112:1024–31.
    101. Falconi M, Bassi C, Bonora A, et al. Role of chemoembolization in synchronous liver metastases from pancreatic endocrine tumours. Dig Surg. 1999;16:32–8.
    102. Chamberlain RS, Canes D, Brown KT, et al. Hepatic neuroendocrine metastases: does intervention alter outcomes? J Am Coll Surg. 2000;190:432–45.
    103. Bettini R, Mantovani W, Boninsegna L, et al. Primary tumour resection in metastatic nonfunctioning pancreatic endocrine carcinomas. Dig Liver Dis. 2009;41:49–55.
    104. Norton JA, Harris EJ, Chen Y, et al. Pancreatic endocrine tumors with major vascular abutment, involvement, or encasement and indication for resection. Arch Surg. 2011;146:724–32.
    105. Tsuchikawa T, Kondo S, Hirano S, et al. Distal pancreatectomy and portal vein resection without vascular reconstruction for endocrine tumors with massive intraportal growth: report of a case. Hepatogastroenterology. 2011;58:1029–31.
    106. Hellman P, Andersson M, Rastad J, et al. Surgical strategy for large or malignant endocrine pancreatic tumors. World J Surg. 2000;24:1353–60.
    107. Handa M, Nakada T, Kajitsuka S, Hirose M, Sato Y. Portosystemic A-V fistula and portal hypertension associated with islet-cell tumor of the pancreas. Nippon Geka Gakkai Zasshi. 1985;86:953–8.
    108. Kawakami H, Kuwatani M, Hirano S, et al. Pancreatic endocrine tumors with intraductal growth into the main pancreatic duct and tumor thrombus within the portal vein: a case report and review of the literature. Intern Med. 2007;46:273–7.
    109. Ochiai T, Masuda T, Nishizawa M, et al. Curative resection of a huge malignant pancreatic endocrine tumor by pancreatoduodenectomy with portal and superior mesenteric vein resection and reconstruction using the right ovarian vein: report of a case. Surg Today. 2011;41:1260–5.
    110. Okuno M, Sakaguchi S, Nagayama M, et al. Nonfunctioning islet cell carcinoma presenting bleeding gastric varices and splenomegaly. Jpn J Surg. 1984;14:244–7.
    111. Yamaguchi T, Takahashi H, Kagawa R, et al. Nonfunctioning pancreatic endocrine tumor presenting with hemorrhage from isolated gastric varices. Am Surg. 2005;71:1027–30.
    112. Pascher A, Klupp J, Neuhaus P. Transplantation in the management of metastatic endocrine tumours. Best Pract Res Clin Gastroenterol. 2005;19:637–48.
    113. Olausson M, Friman S, Herlenius G, et al. Orthotopic liver or multivisceral transplantation as treatment of metastatic neuroendocrine tumors. Liver Transpl. 2007;13:327–33.
    114. Le Treut YP, Gregoire E, Belghiti J, et al. Predictors of long-term survival after liver transplantation for metastatic endocrine tumors: an 85-case French multicentric report. Am J Transplant. 2008;8:1205–13.
    115. Gregoire E, Le Treut YP. Liver transplantation for primary or secondary endocrine tumors. Transpl Int. 2010;23:704–11.
    116. Murthy R, Kamat P, Nunez R, et al. Yttrium-90 microsphere radioembolotherapy of hepatic metastatic neuroendocrine carcinomas after hepatic arterial embolization. J Vasc Interv Radiol. 2008;19:145–51.
    117. Vogl TJ, Naguib NN, Zangos S, Eichler K, Hedayati A, Nour-Eldin NE. Liver metastases of neuroendocrine carcinomas: interventional treatment via transarterial embolization, chemoembolization and thermal ablation. Eur J Radiol. 2009;72:517–28.
    118. Eriksson J, Stalberg P, Nilsson A, et al. Surgery and radiofrequency ablation for treatment of liver metastases from midgut and foregut carcinoids and endocrine pancreatic tumors. World J Surg. 2008;32:930–8.
    119. Basuroy R, Srirajaskanthan R, Ramage JK. A multimodal approach to the management of neuroendocrine tumour liver metastases. Int J Hepatol. 2012;2012:819193.
    120. O’Toole D, Ruszniewski P. Chemoembolization and other ablative therapies for liver metastases of gastrointestinal endocrine tumours. Best Pract Res Clin Gastroenterol. 2005;19:585–94.
    121. Elias D, Goere D, Leroux G, et al. Combined liver surgery and RFA for patients with gastroenteropancreatic endocrine tumors presenting with more than 15 metastases to the liver. Eur J Surg Oncol. 2009;35:1092–7.
    122. Karabulut K, Akyildiz HY, Lance C, et al. Multimodality treatment of neuroendocrine liver metastases. Surgery. 2011;150:316–25.
    123. Akyildiz HY, Mitchell J, Milas M, Siperstein A, Berber E. Laparoscopic radiofrequency thermal ablation of neuroendocrine hepatic metastases: long-term follow-up. Surgery. 2010;148:1288–93.
    124. Mazzaglia PJ, Berber E, Milas M, Siperstein AE. Laparoscopic radiofrequency ablation of neuroendocrine liver metastases: a 10-year experience evaluating predictors of survival. Surgery. 2007;142:10–9.
    125. Toumpanakis C, Meyer T, Caplin ME. Cytotoxic treatment including embolization/chemoembolization for neuroendocrine tumours. Best Pract Res Clin Endocrinol Metab. 2007;21:131–44.
    126. Gu P, Wu J, Newman E, Muggia F. Treatment of liver metastases in patients with neuroendocrine tumors of gastroesophageal and pancreatic origin. Int J Hepatol. 2012;2012:131659.
    127. Reddy SK, Clary BM. Neuroendocrine liver metastases. Surg Clin North Am. 2010;90:853–61.
    128. Nazario J, Gupta S. Transarterial liver-directed therapies of neuroendocrine hepatic metastases. Semin Oncol. 2010;37:118–26.
    129. Osborne DA, Zervos EE, Strosberg J, et al. Improved outcome with cytoreduction versus embolization for symptomatic hepatic metastases of carcinoid and neuroendocrine tumors. Ann Surg Oncol. 2006;13:572–81.
    130. Srirajaskanthan R, Toumpanakis C, Meyer T, Caplin ME. Review article: future therapies for management of metastatic gastroenteropancreatic neuroendocrine tumours. Aliment Pharmacol Ther. 2009;29:1143–54.
    131. Lewis MA, Jaramillo S, Roberts L, Fleming CJ, Rubin J, Grothey A. Hepatic artery embolization for neuroendocrine tumors: postprocedural management and complications. Oncologist. 2012;17:725–31.
    132. Deleporte A, Flamen P, Hendlisz A. State of the art: radiolabeled microspheres treatment for liver malignancies. Expert Opin Pharmacother. 2010;11:579–86.
    133. Kennedy AS, Salem R. Radioembolization (yttrium-90 microspheres) for primary and metastatic hepatic malignancies. Cancer J. 2010;16:163–75.
    134. Memon K, Lewandowski RJ, Mulcahy MF, et al. Radioembolization for neuroendocrine liver metastases: safety, imaging, and long-term outcomes. Int J Radiat Oncol Biol Phys. 2012;83:887–94.
    135. Vyleta M, Coldwell D. Radioembolization in the treatment of neuroendocrine tumor metastases to the liver. Int J Hepatol. 2011;2011:785315.
    136. Kennedy A, Coldwell D, Sangro B, Wasan H, Salem R. Integrating radioembolization into the treatment paradigm for metastatic neuroendocrine tumors in the liver. Am J Clin Oncol. 2012;35:293–301.
    137. Paprottka PM, Hoffmann RT, Haug A, et al. Radioembolization of symptomatic, unresectable neuroendocrine hepatic metastases using yttrium-90 microspheres. Cardiovasc Intervent Radiol. 2012;35:334–42.
    138. Lacin S, Oz I, Ozkan E, Kucuk O, Bilgic S. Intra-arterial treatment with 90yttrium microspheres in treatment-refractory and unresectable liver metastases of neuroendocrine tumors and the use of 111in-octreotide scintigraphy in the evaluation of treatment response. Cancer Biother Radiopharm. 2011;26:631–7.
    139. Shaheen M, Hassanain M, Aljiffry M, et al. Predictors of response to radio-embolization (TheraSphere(R)) treatment of neuroendocrine liver metastasis. HPB (Oxford). 2012;14:60–6.
    140. King J, Quinn R, Glenn DM, et al. Radioembolization with selective internal radiation microspheres for neuroendocrine liver metastases. Cancer. 2008;113:921–9.
    141. Whitney R, Valek V, Fages JF, et al. Transarterial chemoembolization and selective internal radiation for the treatment of patients with metastatic neuroendocrine tumors: a comparison of efficacy and cost. Oncologist. 2011;16:594–601.
    142. Kennedy AS, Dezarn WA, McNeillie P, et al. Radioembolization for unresectable neuroendocrine hepatic metastases using resin 90Y-microspheres: early results in 148 patients. Am J Clin Oncol. 2008;31:271–9.
    143. Saxena A, Chua TC, Bester L, Kokandi A, Morris DL. Factors predicting response and survival after yttrium-90 radioembolization of unresectable neuroendocrine tumor liver metastases: a critical appraisal of 48 cases. Ann Surg. 2010;251:910–6.
    144. Rhee TK, Lewandowski RJ, Liu DM, et al. 90Y Radioembolization for metastatic neuroendocrine liver tumors: preliminary results from a multi-institutional experience. Ann Surg. 2008;247:1029–35.
    145. Lee E, Leon Pachter H, Sarpel U. Hepatic arterial embolization for the treatment of metastatic neuroendocrine tumors. Int. J Hepatol. 2012;2012:471203.
    146. Riccardi F, Rizzo M, Festino L, et al. Therapy innovation for the treatment of pancreatic neuroendocrine tumors. Expert Opin Ther Targets. 2012;16 Suppl 2:S91–102.
    147. Eriksson B, Annibale B, Bajetta E, et al. ENETS consensus guidelines for the standards of care in neuroendocrine tumors: chemotherapy in patients with neuroendocrine tumors. Neuroendocrinology. 2009;90:214–9.
    148. Maire F, Hammel P, Kianmanesh R, et al. Is adjuvant therapy with streptozotocin and 5-fluorouracil useful after resection of liver metastases from digestive endocrine tumors? Surgery. 2009;145:69–75.
    149. Hoffmann KM, Gibril F, Entsuah LK, Serrano J, Jensen RT. Patients with multiple endocrine neoplasia type 1 with gastrinomas have an increased risk of severe esophageal disease including stricture and the premalignant condition, Barrett’s esophagus. J Clin Endocrinol Metab. 2006;91:204–12.
    150. Kouvaraki MA, Ajani JA, Hoff P, et al. Fluorouracil, doxorubicin, and streptozocin in the treatment of patients with locally advanced and metastatic pancreatic endocrine carcinomas. J Clin Oncol. 2004;22:4762–71.
    151. Strosberg JR, Fine RL, Choi J, et al. First-line chemotherapy with capecitabine and temozolomide in patients with metastatic pancreatic endocrine carcinomas. Cancer. 2011;117:268–75.
    152. Maire F, Hammel P, Faivre S, et al. Temozolomide: a safe and effective treatment for malignant digestive endocrine tumors. Neuroendocrinology. 2009;90:67–72.
    153. Ekeblad S, Sundin A, Janson ET, et al. Temozolomide as monotherapy is effective in treatment of advanced malignant neuroendocrine tumors. Clin Cancer Res. 2007;13:2986–91.
    154. 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;15:338–45.
    155. Sideris L, Dube P, Rinke A. Antitumor effects of somatostatin analogs in neuroendocrine tumors. Oncologist. 2012;17:747–55.
    156. Appetecchia M, Baldelli R. Somatostatin analogues in the treatment of gastroenteropancreatic neuroendocrine tumours, current aspects and new perspectives. J Exp Clin Cancer Res. 2010;29:19–31.
    157. Plockinger U, Wiedenmann B. Biotherapy. Best Pract Res Clin Endocrinol Metab. 2007;21:145–62.
    158. Strosberg J, Kvols L. Antiproliferative effect of somatostatin analogs in gastroenteropancreatic neuroendocrine tumors. World J Gastroenterol. 2010;16:2963–70.
    159. Panzuto F, Di Francesco V, Iannicelli E, et al. Long-term clinical outcome of somatostatin analogues for treatment of progressive, metastatic, well-differentiated entero-pancreatic endocrine carcinoma. Ann Oncol. 2006;17:461–6.
    160. Rinke A, Muller HH, Schade-Brittinger C, et al. Placebo-controlled, double-blind, prospective, randomized study on the effect of octreotide LAR in the control of tumor growth in patients with metastatic neuroendocrine midgut tumors: a report from the PROMID Study Group. J Clin Oncol. 2009;27:4656–63.
    161. Blumberg J, Liyanage N, Caplin M, UK and Ireland NET/ENET Society. The Clarinet study-assessing the effect of Lanreotide Autogel on tumor progression-free survival in patients with non-functioning gastroenteropancreatic neuroendocrine tumors (GEP-NETs). NANETS-2011-Neuroendocrine Tumor Symposium (abstr C4).
    162. Oberg K, Eriksson B. Endocrine tumours of the pancreas. Best Pract Res Clin Gastroenterol. 2005;19:753–81.
    163. Butturini G, Bettini R, Missiaglia E, et al. Predictive factors of efficacy of the somatostatin analogue octreotide as first line therapy for advanced pancreatic endocrine carcinoma. Endocr Relat Cancer. 2006;13:1213–21.
    164. Yamaguchi M, Yamada Y, Hosokawa Y, et al. Long-term suppressive effect of octreotide on progression of metastatic gastrinoma with multiple endocrine neoplasia type 1: seven-year follow up. Intern Med. 2010;49:1557–63.
    165. Granberg D, Jacobsson H, Oberg K, Gustavsson J, Lehtihet M. Regression of a large malignant gastrinoma on treatment with Sandostatin LAR: a case report. Digestion. 2008;77:92–5.
    166. Guillermet-Guibert J, Lahlou H, Pyronnet S, Bousquet C, Susini C. Somatostatin receptors as tools for diagnosis and therapy: Molecular aspects. Best Pract Res Clin Gastroenterol. 2005;19:535–51.
    167. Oberg K, Ferone D, Kaltsas G, Knigge UP, Taal B, Plockinger U. ENETS consensus guidelines for the standards of care in neuroendocrine tumors: biotherapy. Neuroendocrinology. 2009;90:209–13.
    168. The NCCN clinical practice guidelines in oncology for neuroendocrine tumors version 1.2012. Version1.2012. 2012. online. go to www.nccn.org. Ref Type: Computer Program
    169. Miljkovic MD, Girotra M, Abraham RR, Erlich RB. Novel medical therapies of recurrent and metastatic gastroenteropancreatic neuroendocrine tumors. Dig Dis Sci. 2012;57:9–18.
    170. Yao JC. Molecular targeted therapy for carcinoid and islet-cell carcinoma. Best Pract Res Clin Endocrinol Metab. 2007;21:163–72.
    171. Phan AT. Metastatic pancreatic neuroendocrine tumors (pNET): placing current findings into perspective. Cancer Treat Rev. 2012 [Epub ahead of print].
    172. Capurso G, Fazio N, Festa S, Panzuto F, de Braud F, Delle Fave G. Molecular target therapy for gastroenteropancreatic endocrine tumours: biological rationale and clinical perspectives. Crit Rev Oncol Hematol. 2009;72:110–24.
    173. Fazio N, Cinieri S, Lorizzo K, et al. Biological targeted therapies in patients with advanced enteropancreatic neuroendocrine carcinomas. Cancer Treat Rev. 2010;36 Suppl 3:S87–94.
    174. Missiaglia E, Dalai I, Barbi S, et al. Pancreatic endocrine tumors: expression profiling evidences a role for AKT-mTOR pathway. J Clin Oncol. 2010;28:245–55.
    175. Varas M, Gornals J, Ponseti JM, et al. Pancreatic endocrine tumors or apudomas. Rev Esp Enferm Dig. 2011;103:184–90.
    176. Yao JC, Phan AT, Chang DZ, et al. Efficacy of RAD001 (everolimus) and octreotide LAR in advanced low- to intermediate-grade neuroendocrine tumors: results of a phase II study. J Clin Oncol. 2008;26:4311–8.
    177. Ito T, Okusaka T, Ikeda M et al. Everolimus versus placebo in Japanese patients with advanced pancreatic neuroendocrine tumors (pNETs); Japanese subgroup analysis of RADIANT-3. J. Clin. Oncol. 2011;29:289 (abstr).
    178. Raymond E, Hobday T, Castellano D, Reidy-Lagunes D, Garcia-Carbonero R, Carrato A. Therapy innovations: tyrosine kinase inhibitors for the treatment of pancreatic neuroendocrine tumors. Cancer Metastasis Rev. 2011;30 Suppl 1:19–26.
    179. Faivre S, Sablin MP, Dreyer C, Raymond E. Novel anticancer agents in clinical trials for well-differentiated neuroendocrine tumors. Endocrinol Metab Clin North Am. 2010;39:811–26.
    180. Furukawa M, Raffeld M, Mateo C, et al. Increased expression of insulin-like growth factor I (IGF-1) and/or its receptor (IGF-1R) in gastrinomas is associated with low curability, increased growth and development of metastases. Cancer Res. 2005;11:3233–42.
    181. Peghini PL, Iwamoto M, Raffeld M, et al. Overexpression of epidermal growth factor and hepatocyte growth factor receptors in a proportion of gastrinomas correlates with aggressive growth and lower curability. Clin Cancer Res. 2002;8:2273–85.
    182. Pavel ME, Wiedenmann B. Novel therapeutic agents for the treatment of gastroenteropancreatic neuroendocrine tumors. Horm Metab Res. 2011;43:844–53.
    183. Capdevila J, Salazar R. Molecular targeted therapies in the treatment of gastroenteropancreatic neuroendocrine tumors. Target Oncol. 2009;4:287–96.
    184. Fjallskog ML, Lejonklou MH, Oberg KE, Eriksson BK, Janson ET. Expression of molecular targets for tyrosine kinase receptor antagonists in malignant endocrine pancreatic tumors. Clin Cancer Res. 2003;9:1469–73.
    185. Kulke MH, Lenz HJ, Meropol NJ, et al. Activity of sunitinib in patients with advanced neuroendocrine tumors. J Clin Oncol. 2008;26:3403–10.
    186. Okusaka T, Ito T, Nishida T, et al. Phase 11 study of sunitinib (SU) in Japanese patients with unresectable or metastatic, well-differentiated pancreatic neuroendocrine tumors (NET). J. Clin. Oncol. 2012;30:381 (abstr).
    187. Virgolini I, Traub T, Novotny C, et al. Experience with indium-111 and yttrium-90-labeled somatostatin analogs. Curr Pharm Des. 2002;8:1781–807.
    188. Oberg K. Somatostatin analog octreotide LAR in gastro-entero-pancreatic tumors. Expert Rev Anticancer Ther. 2009;9:557–66.
    189. Van Essen M, Krenning EP, Kam BL, de Jong M, Valkema R, Kwekkeboom DJ. Peptide-receptor radionuclide therapy for endocrine tumors. Nat Rev Endocrinol. 2009;5:382–93.
    190. Kwekkeboom DJ, de Herder WW, Kam BL, et al. Treatment with the radiolabeled somatostatin analog [177 Lu-DOTA 0, Tyr3]octreotate: toxicity, efficacy, and survival. J Clin Oncol. 2008;26:2124–30.
    191. Kwekkeboom DJ, Teunissen JJ, Bakker WH, et al. Radiolabeled somatostatin analog [177Lu-DOTA0, Tyr3]octreotate in patients with endocrine gastroenteropancreatic tumors. J Clin Oncol. 2005;23:2754–62.
    192. Forrer F, Valkema R, Kwekkeboom DJ, de Jong M, Krenning EP. Neuroendocrine tumors. Peptide receptor radionuclide therapy. Best Pract Res Clin Endocrinol Metab. 2007;21:111–29.
    193. Kwekkeboom DJ, de Herder WW, Krenning EP. Somatostatin receptor-targeted radionuclide therapy in patients with gastroenteropancreatic neuroendocrine tumors. Endocrinol Metab Clin North Am. 2011;40:173–85.
    194. Oberg KE, Reubi JC, Kwekkeboom DJ, Krenning EP. Role of somatostatins in gastroenteropancreatic neuroendocrine tumor development and therapy. Gastroenterology. 2010;139:742–53.
    195. Dong M, Phan AT, Yao JC. New strategies for advanced neuroendocrine tumors in the era of targeted therapy. Clin Cancer Res. 2012;18:1830–6.
    196. Duran I, Kortmansky J, Singh D, et al. A phase II clinical and pharmacodynamic study of temsirolimus in advanced neuroendocrine carcinomas. Br J Cancer. 2006;95:1148–54.
    197. Yao JC, Phan A, Hoff PM, et al. 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. 2008;26:1316–23.
    198. Chan JA, Kulke MH. New treatment options for patients with advanced neuroendocrine tumors. Curr Treat Options Oncol. 2011;12:136–48.
    199. Kulke MH, Siu LL, Tepper JE, et al. Future directions in the treatment of neuroendocrine tumors: consensus report of the national cancer institute neuroendocrine tumor clinical trials planning meeting. J Clin Oncol. 2011;29:934–43.
    200. Kulke MH, Stuart K, Enzinger PC, et al. Phase II study of temozolomide and thalidomide in patients with metastatic neuroendocrine tumors. J Clin Oncol. 2006;24:401–6.
    201. Raut CP, Kulke MH. Targeted therapy in advanced well-differentiated neuroendocrine tumors. Oncologist. 2011;16:286–95.
    202. Eriksson B. New drugs in neuroendocrine tumors: rising of new therapeutic philosophies? Curr Opin Oncol. 2010;22:381–6.
    203. Schmid HA, Schoeffter P. Functional activity of the multiligand analog SOM230 at human recombinant somatostatin receptor subtypes supports its usefulness in neuroendocrine tumors. Neuroendocrinology. 2004;80 Suppl 1:47–50.
    204. Schmitt AM, Anlauf M, Rousson V, et al. WHO 2004 criteria and CK19 are reliable prognostic markers in pancreatic endocrine tumors. Am J Surg Pathol. 2007;31:1677–82.
    205. Ruf J, Schiefer J, Furth C, et al. 68Ga-DOTATOC PET/CT of neuroendocrine tumors: spotlight on the CT phases of a triple-phase protocol. J Nucl Med. 2011;52:697–704.
    206. Hainsworth JD, Spigel DR, Litchy S, Greco FA. Phase II trial of paclitaxel, carboplatin, and etoposide in advanced poorly differentiated neuroendocrine carcinoma: a Minnie Pearl Cancer Research Network Study. J Clin Oncol. 2006;24:3548–54.
    207. Olsen IH, Langer SW, Jepsen I, et al. First-line treatment of patients with disseminated poorly differentiated neuroendocrine carcinomas with carboplatin, etoposide, and vincristine: a single institution experience. Acta Oncol. 2012;51:97–100.
    208. Clancy TE, Sengupta TP, Paulus J, Ahmed F, Duh MS, Kulke MH. Alkaline phosphatase predicts survival in patients with metastatic neuroendocrine tumors. Dig Dis Sci. 2006;51:877–84.
    209. Lo CY, Van Heerden JA, Thompson GB, Grant CS, Soreide JA, Harmsen WS. Islet cell carcinoma of the pancreas. World J Surg. 1996;20:878–84.
    210. Janson ET, Holmberg L, Stridsberg M, et al. Carcinoid tumors: analysis of prognostic factors and survival in 301 patients from a referral center. Ann Oncol. 1997;8:685–90.
    211. Madeira I, Terris B, Voss M, et al. Prognostic factors in patients with endocrine tumours of the duodenopancreatic area. Gut. 1998;43:422–7.
    212. Maton PN, Gardner JD, Jensen RT. Cushing’s syndrome in patients with Zollinger-Ellison syndrome. N Engl J Med. 1986;315:1–5.
    213. Arnold R, Wilke A, Rinke A, et al. Plasma chromogranin a as marker for survival in patients with metastatic endocrine gastroenteropancreatic tumors. Clin Gastroenterol Hepatol. 2008;6:820–7.
    214. Arnold R, Rinke A, Klose KJ, et al. Octreotide versus octreotide plus interferon-alpha in endocrine gastroenteropancreatic tumors: a randomized trial. Clin Gastroenterol Hepatol. 2005;3:761–71.
    215. Strosberg J, Gardner N, Kvols L. Survival and prognostic factor analysis of 146 metastatic neuroendocrine tumors of the mid-gut. Neuroendocrinology. 2009;89:471–6.
  • 作者单位:1. Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582 Japan2. Digestive Diseases Branch, NIDDK, NIH, Building 10, Room 9C-103, Bethesda, MD 20892, USA
  • ISSN:1435-5922
文摘
Neuroendocrine tumors (NETs) [carcinoids, pancreatic neuroendocrine tumors (pNETs)] are becoming an increasing clinical problem because not only are they increasing in frequency, but they can frequently present with advanced disease that requires diagnostic and treatment approaches different from those used in the neoplasms that most physicians are used to seeing and treating. In the past few years there have been numerous advances in all aspects of NETs including: an understanding of their unique pathogenesis; specific classification systems developed which have prognostic value; novel methods of tumor localization developed; and novel treatment approaches described. In patients with advanced metastatic disease these include the use of newer chemotherapeutic approaches, an increased understanding of the role of surgery and cytoreductive methods, the development of methods for targeted delivery of cytotoxic agents, and the development of targeted medical therapies (everolimus, sunitinib) based on an increased understanding of the disease biology. Although pNETs and gastrointestinal NETs share many features, recent studies show they differ in pathogenesis and in many aspects of diagnosis and treatment, including their responsiveness to different therapies. Because of limited space, this review will be limited to the advances made in the management and treatment of patients with advanced metastatic pNETs over the past 5 years.

© 2004-2018 中国地质图书馆版权所有 京ICP备05064691号 京公网安备11010802017129号

地址:北京市海淀区学院路29号 邮编:100083

电话:办公室:(+86 10)66554848;文献借阅、咨询服务、科技查新:66554700