Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Treatment of advanced disease in patients with well-differentiated neuroendocrine tumors

Abstract

Well-differentiated neuroendocrine tumors (NETs), which are also referred to as well-differentiated endocrine carcinoma according to WHO terminology, are usually slow-growing cancers, even when they exhibit gross local invasion and/or metastases. The survival of patients with metastatic NET is often measured in years to decades. Once NET progresses or becomes symptomatic the patient's prognosis is poor. An important challenge for clinicians is to distinguish at an early stage those patients who will die with the disease, from those who will succumb because of it, so that the appropriate level of care can be administered. Reliable genomic predictors could provide substantial advancements in prognosis and, possibly, treatment; however, such markers are currently unavailable. Early literature on the treatment of NETs is confounded by a lack of formal objective response criteria. Somatostatin analogs can control symptoms and can stabilize some slow-growing tumors, but rarely result in tumor regression. Surgery is curative in only a minority of patients, and systemic chemotherapy is minimally effective. Advances in the understanding of tumor biology have led to the identification of important cellular processes involved in the pathogenesis of NETs, and agents that target these processes have now entered clinical trials. We will discuss the data on therapies currently used to treat well-differentiated NETs, and the strategies being used in clinical trials.

Key Points

  • Patients with well-differentiated neuroendocrine tumors (NETs), the most common of which are carcinoid tumors and islet-cell carcinomas, typically present with metastatic disease

  • Metastatic disease can be followed expectantly as it is often slow-growing and does not require intervention for many months to years

  • Once the NET progresses or is symptomatic despite somatostatin-analog treatment, therapeutic options are extremely limited and no therapy is currently considered standard

  • Tumor heterogeneity, the varying degree of aggressiveness, and the lack of standard regimens and guidelines for NET treatment pose a substantial clinical challenge

  • Well-differentiated NETs exhibit diverse clinical outcomes, which can sometimes be stratified by certain histopathological features including cytological grade degree, angioinvasion, proliferative index (as assessed by mitotic index and Ki67), tumor size, presence of metastases, and functional activity

  • An understanding of tumor signaling mechanisms has led to the development of promising agents that could target clinically important pathways, but an urgent need remains for genetic markers that identify the clinical phenotypes

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Grading of neuroendocrine tumors (NETs).
Figure 2: Typical presentation of an ileal carcinoid tumor.
Figure 3: A patient with a metastatic nonfunctional carcinoid tumor, Eastern Cooperative Oncology Group performance status 0, with bulky liver disease, which illustrates the tropism of NETs for the liver.

Similar content being viewed by others

References

  1. Moertel CG et al. (1991) Treatment of neuroendocrine carcinomas with combined etoposide and cisplatin. Evidence of major therapeutic activity in the anaplastic variants of these neoplasms. Cancer 68: 227–232

    Article  CAS  Google Scholar 

  2. Modlin IM et al. (2006) The functional characterization of normal and neoplastic human enterochromaffin cells. J Clin Endocrinol Metab 91: 2340–2348

    Article  CAS  Google Scholar 

  3. Rindi G et al. (2007) Prognostic factors in gastrointestinal endocrine tumors. Endocr Pathol 18: 145–149

    Article  Google Scholar 

  4. Ferrone CR et al. (2007) Determining prognosis in patients with pancreatic endocrine neoplasms: can the WHO classification system be simplified? J Clin Oncol 25: 5609–5615

    Article  Google Scholar 

  5. Van Eeden S et al. (2002) Classification of low-grade neuroendocrine tumors of midgut and unknown origin. Hum Pathol 33: 1126–1132

    Article  Google Scholar 

  6. Rindi G et al. (2007) TNM staging of midgut and hindgut (neuro) endocrine tumors: a consensus proposal including a grading system. Virchows Arch 451: 757–762

    Article  CAS  Google Scholar 

  7. Schurr P et al. (2007) Aggressive surgery improves long-term survival in neuroendocrine pancreatic tumors: an institutional experience. Ann Surg 245: 273–281

    Article  Google Scholar 

  8. Touzios JG et al. (2005) Neuroendocrine hepatic metastases: does aggressive management improve survival? Ann Surg 241: 776–783

    Article  Google Scholar 

  9. Sarmiento JM et al. (2003) Surgical treatment of neuroendocrine metastases to the liver: a plea for resection to increase survival. J Am Coll Surg 197: 29–37

    Article  Google Scholar 

  10. Chamberlain RS et al. (2000) Hepatic neuroendocrine metastases: does intervention affect outcome? J Am Coll Surg 190: 432–445

    Article  CAS  Google Scholar 

  11. Ruszniewski P et al. (1993) Hepatic arterial chemoembolization in patients with liver metastases of endocrine tumors. A prospective phase II study in 24 patients. Cancer 71: 2624–2630

    Article  CAS  Google Scholar 

  12. Eriksson BK et al. (1998) Liver embolizations of patients with malignant neuroendocrine gastrointestinal tumors. Cancer 83: 2293–2301

    Article  CAS  Google Scholar 

  13. Gupta S et al. (2003) Hepatic artery embolization and chemoembolization for treatment of patients with metastatic carcinoid tumors: the MD Anderson experience. Cancer 9: 261–267

    Article  Google Scholar 

  14. Moertel C et al. (1994) The management of patients with advanced carcinoid tumors and islet cell carcinomas. Ann Intern Med 120: 302–309

    Article  CAS  Google Scholar 

  15. Bloomston M et al. (2007) Hepatic artery chemoembolization in 122 patients with metastatic carcinoid tumor: lessons learned. J Gastrointest Surg 11: 264–271

    Article  Google Scholar 

  16. Lamberts SW et al. (1990) Somatostatin-receptor imaging in the localization of endocrine tumors. N Engl J Med 323: 1246–1249

    Article  CAS  Google Scholar 

  17. Cwikla JB et al. (2007) Diagnostic imaging approach to gastro-entero-pancreatic carcinomas of neuroendocrine origin—single NET center experience in Poland. Neuro Endocrinol Lett 28: 789–800

    PubMed  Google Scholar 

  18. Dromain C et al. (2005) Detection of liver metastases from endocrine tumors: a prospective comparison of somatostatin receptor scintigraphy, computed tomography, and magnetic resonance imaging. J Clin Oncol 23: 70–78

    Article  Google Scholar 

  19. Kumbasar B et al. (2004) Imaging of neuroendocrine tumors: accuracy of helical CT versus SRS. Abdom Imaging 29: 696–702

    Article  CAS  Google Scholar 

  20. Asnacios A et al. (2008) Indium-111–pentetreotide scintigraphy and somatostatin receptor subtype 2 expression: new prognostic factors for malignant well-differentiated endocrine tumors. J Clin Oncol 26: 963–970

    Article  Google Scholar 

  21. Reichlin S (1983) Somatostatin. N Engl J Med 309: 1495–1501

    Article  CAS  Google Scholar 

  22. Reubi JC et al. (1990) Detection of somatostatin receptors in surgical and percutaneous needle biopsy samples of carcinoids and islet cell carcinomas. Cancer Res 50: 5969–5977

    CAS  PubMed  Google Scholar 

  23. Kvols LK et al. (1986) Treatment of the malignant carcinoid syndrome. Evaluation of a long-acting somatostatin analogue. N Engl J Med 315: 663–666

    Article  CAS  Google Scholar 

  24. Di Bartolomeo M et al. (1996) Clinical efficacy of octreotide in the treatment of metastatic neuroendocrine tumors. A study by the Italian Trials in Medical Oncology Group. Cancer 77: 402–408

    Article  CAS  Google Scholar 

  25. Ruszniewski P et al. (1996) Treatment of the carcinoid syndrome with the longacting somatostatin analogue lanreotide: a prospective study in 39 patients. Gut 39: 279–283

    Article  CAS  Google Scholar 

  26. Faiss S et al. (1996) Drug therapy in metastatic neuroendocrine tumors of the gastroenteropancreatic system. Recent Results Cancer Res 142: 193–207

    Article  CAS  Google Scholar 

  27. Welin SV et al. (2004) High-dose treatment with a long-acting somatostatin analogue in patients with advanced midgut carcinoid tumours. Eur J Endocrinol 151: 107–112

    Article  CAS  Google Scholar 

  28. Arnold R et al. (1993) Gastroenteropancreatic endocrine tumours: effect of Sandostatin on tumour growth. The German Sandostatin Study Group. Digestion 54: 72–75

    Article  Google Scholar 

  29. Saltz L et al. (1993) Octreotide as an antineoplastic agent in the treatment of functional and nonfunctional neuroendocrine tumors. Cancer 72: 244–248

    Article  CAS  Google Scholar 

  30. Ducreux M et al. (2000) The antitumoral effect of the long-acting somatostatin analog lanreotide in neuroendocrine tumors. Am J Gastroenterol 95: 3276–3281

    Article  CAS  Google Scholar 

  31. Panzuto F et al. (2006) Long-term clinical outcome of somatostatin analogues for treatment of progressive, metastatic well-differentiated entero-pancreatic endocrine carcinoma. Ann Oncol 17: 461–466

    Article  CAS  Google Scholar 

  32. Moertel CG et al. (1989) Therapy of metastatic carcinoid tumor and the malignant carcinoid syndrome with recombinant leukocyte A interferon. J Clin Oncol 7: 865–888

    Article  CAS  Google Scholar 

  33. Saltz L et al. (1994) A phase II trial of α-interferon and 5-fluorouracil in patients with advanced carcinoid and islet cell tumors. Cancer 74: 958–961

    Article  CAS  Google Scholar 

  34. Plöckinger U and Wiedenmann B (2007) Neuroendocrine tumors. Biotherapy. Best Pract Res Clin Endocrinol Metab 21: 145–162

    Article  Google Scholar 

  35. Faiss S et al. (2003) Prospective, randomized multicenter trial on the antiproliferative effect of lanreotide, interferon alfa, and their combination for therapy of metastatic neuroendocrine gastroenteropancreatic tumors—the International Lanreotide and Interferon Alfa Study Group. J Clin Oncol 21: 2689–2696

    Article  CAS  Google Scholar 

  36. Välimäki M et al. (1991) Is the treatment of metastatic carcinoid tumor with interferon not as successful as suggested? Cancer 67: 547–549

    Article  Google Scholar 

  37. Vavra JJ et al. (1960) Streptozotocin, a new antibacterial antibiotic. Antibiot Annu 1: 230–235

    Google Scholar 

  38. Murray-Lyon IM et al. (1968) Treatment of multiple-hormone-producing malignant islet-cell tumor with streptozocin. Lancet 2: 895–898

    Article  CAS  Google Scholar 

  39. Schein PS et al. (1974) Clinical antitumour acitivity and toxicity of streptozotocin (NSC-85998). Cancer 34: 993–1000

    Article  CAS  Google Scholar 

  40. Schein P et al. (1973) Streptozocin for malignant insulinomas and carcinoid tumor. Report of eight cases and review of the literature. Arch Intern Med 132: 555–561

    Article  CAS  Google Scholar 

  41. Moertel CG et al. (1980) Streptozocin alone compared with streptozocin plus fluorouracil in the treatment of advanced islet-cell carcinoma. N Engl J Med 303: 1189–1194

    Article  CAS  Google Scholar 

  42. Moertel CG et al. (1992) Streptozocin–doxorubicin, streptozocin–fluorouracil, or chlorozotocin in the treatment of advanced islet-cell carcinoma. N Engl J Med 326: 519–523

    Article  CAS  Google Scholar 

  43. Cheng PN and Saltz LB (1999) Failure to confirm major objective antitumor activity for streptozocin and doxorubicin in the treatment of patients with advanced islet cell carcinoma. Cancer 86: 944–948

    Article  CAS  Google Scholar 

  44. Rivera E and Ajani JA (1998) Doxorubicin, streptozocin, and 5-fluorouracil chemotherapy for patients with metastatic islet-cell carcinoma. Am J Clin Oncol 21: 36–38

    Article  CAS  Google Scholar 

  45. Kouvaraki MA et al. (2004) Fluorouracil, doxorubicin, and streptozocin in the treatment of patients with locally advanced and metastatic pancreatic endocrine carcinomas. J Clin Oncol 22: 4762–4771

    Article  CAS  Google Scholar 

  46. Gonzalez MA et al. (2003) Treatment of neuroendocrine tumours with infusional 5-fluorouracil, folinic acid and streptozocin. Br J Cancer 89: 455–456

    Article  CAS  Google Scholar 

  47. Moertel CG and Hanley JA (1979) Combination chemotherapy trials in metastatic carcinoid tumor and the malignant carcinoid syndrome. Cancer Clin Trials 2: 327–334

    CAS  PubMed  Google Scholar 

  48. Engstrom PF et al. (1984) Streptozocin plus fluorouracil versus doxorubicin therapy for metastatic carcinoid tumor. J Clin Oncol 2: 1255–1259

    Article  CAS  Google Scholar 

  49. Bukowski RM et al. (1987) A phase II trial of combination chemotherapy in patients with metastatic carcinoid tumors. A Southwest Oncology Group Study. Cancer 60: 2891–2895

    Article  CAS  Google Scholar 

  50. Sun W et al. (2005) Phase II/III study of doxorubicin with fluorouracil compared with streptozocin with fluorouracil or dacarbazine in the treatment of advanced carcinoid tumors: Eastern Cooperative Oncology Group Study E1281. J Clin Oncol 23: 4897–4904

    Article  CAS  Google Scholar 

  51. Ramanathan RK et al. (2001) Phase II trial of dacarbazine (DTIC) in advanced pancreatic islet cell carcinoma. Study of the Eastern Cooperative Oncology Group–E6282. Ann Oncol 12: 1139–1143

    Article  CAS  Google Scholar 

  52. Bukowski RM et al. (1994) Phase II trial of dimethyltriazenoimidazole carboxamide in patients with metastatic carcinoid. A Southwest Oncology Group Study. Cancer 73: 1505–1508

    Article  CAS  Google Scholar 

  53. Kulke MH et al. (2006) Phase II study of temozolomide and thalidomide in patients with metastatic neuroendocrine tumors. J Clin Oncol 24: 401–406

    Article  CAS  Google Scholar 

  54. Varker KA et al. (2008) Phase II study of thalidomide in patients with metastatic carcinoid and islet cell tumors. Cancer Chemother Pharmacol 61: 661–668

    Article  CAS  Google Scholar 

  55. Ekeblad S et al. (2007) Temozolomide as monotherapy is effective in treatment of advanced malignant neuroendocrine tumors. Clin Cancer Res 13: 2986–2991

    Article  CAS  Google Scholar 

  56. Kulke MH et al. (2007) Prediction of response to temozolomide (TMZ)-based therapy by loss of MGMT expression in patients with advanced neuroendocrine tumors (NET) [abstract #4505]. ASCO Meeting Abstracts 25: 4505

    Google Scholar 

  57. Waldherr C et al. (2001) The clinical value of [90Y-DOTA]-D-Phe1-Tyr3-octreotide (90Y-DOTATOC) in the treatment of neuroendocrine tumours: a clinical phase II study. Ann Oncol 12: 941–945

    Article  CAS  Google Scholar 

  58. Waldherr C et al. (2002) Tumor response and clinical benefit in neuroendocrine tumors after 7.4 GBq 90Y-DOTATOC. J Nucl Med 43: 610–616

    CAS  PubMed  Google Scholar 

  59. Virgolini I et al. (2002) 111In- and 90Y-DOTA-lanreotide: results and implications of the MAURITIUS trial. J Nucl Med 32: 148–155

    Google Scholar 

  60. Valkema R et al. (2006) Survival and response after peptide receptor radionuclide therapy with [90Y-DOTA0,Tyr3]octreotide in patients with advanced gastroenteropancreatic neuroendocrine tumors. Semin Nucl Med 36: 147–156

    Article  Google Scholar 

  61. Kwekkeboom DJ et al. (2008) Treatment with the radiolabeled somatostatin analog [177 Lu-DOTA0,Tyr3]octreotate: toxicity, efficacy, and survival. J Clin Oncol 26: 2124–2130

    Article  CAS  Google Scholar 

  62. Kwekkeboom DJ et al. (2005) Radiolabeled somatostatin analog [177Lu-DOTA0,Tyr3]octreotate in patients with gastroenteropancreatic tumors. J Clin Oncol 23: 2754–2762

    Article  CAS  Google Scholar 

  63. Duran I et al. (2006) A phase II clinical and pharmacodynamic study trial of temsirolimus in advanced neuroendocrine tumors. Br J Cancer 95: 1148–1154

    Article  CAS  Google Scholar 

  64. Yao JC et al. (2007) Phase II study of RAD001 (everolimus) and depot octreotide (sandostatin LAR) in advanced low grade neuroendocrine carcinoma (LGNET) [abstract #4503]. ASCO Meeting Abstracts 25: 4503

    Google Scholar 

  65. Terris B et al. (1998) Expression of vascular endothelial growth factor in digestive tumours. Histopathology 32: 133–138

    Article  CAS  Google Scholar 

  66. Phan AT et al. (2006) Association of VEGF expression with poor prognosis among patients with low-grade neuroendocrine carcinoma [abstract #4091]. ASCO Meeting Abstracts 24: 4091

    Google Scholar 

  67. Yao JC et al. (2005) Improved progression free survival (PFS), and rapid, sustained decrease in tumor perfusion among patients with advanced carcinoid treated with bevacizumab [abstract #4007]. 2005 ASCO Meeting Abstracts 23: 4007

    Google Scholar 

  68. Hurwitz H et al. (2004) Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N Engl J Med 350: 2335–2342

    Article  CAS  Google Scholar 

  69. Sandler A et al. (2006) Paclitaxel-carboplatin alone or with bevacizumab for non-small cell lung cancer. N Engl J Med 355: 2542–2550

    Article  CAS  Google Scholar 

  70. Miller KD et al. (2005) Randomized phase III trial of capecitabine compared with bevacizumab plus capecitabine in patients with previously treated metastatic breast cancer. J Clin Oncol 23: 792–799

    Article  CAS  Google Scholar 

  71. Miller K et al. (2007) Paclitaxel plus bevacizumab versus paclitaxel alone for metastatic breast cancer. N Engl J Med 357: 2666–2676

    Article  CAS  Google Scholar 

  72. Kulke MH et al. (2006) A phase II study of temozolomide and bevacizumab in patients with advanced neuroendocrine tumors [abstract #4044]. ASCO Meeting Abstracts 24: 4044

    Google Scholar 

  73. Kulke M et al. (2005) Results of a phase II study with sunitinib malate (SU11248) in patients (pts) with advanced neuroendocrine tumours (NETs) [abstract #718]. Eur J Cancer Suppl 3: 204

    Google Scholar 

  74. Nilsson O et al. (1992) Presence of IGF-1 in human midgut carcinoid tumours—an autocrine regulator of carcinoid tumour growth? Int J Cancer 51: 195–203

    Article  CAS  Google Scholar 

  75. Adams TE et al. (2000) Structure and function of the type 1 insulin-like growth factor receptor. Cell Mol Life Sci 57: 1050–1093

    Article  CAS  Google Scholar 

  76. Wang Y and Sun Y (2002) Insulin-like growth factor receptor-1 as an anti-cancer target: blocking transformation and inducing apoptosis. Curr Cancer Drug Targets 2: 191–207

    Article  CAS  Google Scholar 

  77. Pollak MN et al. (2004) Insulin-like growth factors and neoplasia. Nat Rev Cancer 4: 505–518

    Article  CAS  Google Scholar 

  78. Ouban A et al. (2003) Expression and distribution of insulin-like growth factor-1 receptor in human carcinomas. Hum Pathol 34: 803–808

    Article  CAS  Google Scholar 

  79. García-Echeverría C et al. (2004) In vivo antitumor activity of NVP-AEW541—a novel, potent, and selective inhibitor of the IGF-IR kinase. Cancer Cell 5: 231–239

    Article  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Diane L Reidy.

Ethics declarations

Competing interests

Dr Leonard B Saltz has declared he is a consultant and receives grant/research support from Merck and Roche. D Reidy has declared she receives grant/research support from Merck. L Tang has declared no competing interests.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Reidy, D., Tang, L. & Saltz, L. Treatment of advanced disease in patients with well-differentiated neuroendocrine tumors. Nat Rev Clin Oncol 6, 143–152 (2009). https://doi.org/10.1038/ncponc1326

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncponc1326

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing