Skip to main content
Log in

Cyto-histology in NET: what is necessary today and what is the future?

  • Published:
Reviews in Endocrine and Metabolic Disorders Aims and scope Submit manuscript

Abstract

The carcinoid as originally described is part of the relatively large family of neuroendocrine neoplasia found in almost every organ. Historical reasons back their current definitions. Neuroendocrine cancer is most frequently observed in the lung and the digestive tract. In the lung is defined as carcinoid (typical and atypical) for well differentiated, low to intermediate grade, and small cell and large cell neuroendocrine carcinoma for poorly differentiated, high grade. In the digestive system are respectively defined as neuroendocrine tumor (NET) and neuroendocrine carcinoma (NEC) of small and large cell types. Grading and staging are developed for their clinical classification by the World Health Organization (WHO) and the American Joint Committee on Cancer (AJCC). In both anatomical sites the morphological features are overlapping, with bland histology for carcinoid and NET, and aggressive features with extensive necrosis, severe atypia and abundant, atypical mitoses for high grade cancer types. Such features are also essential diagnostic clues in cytological preparations. The confirmation of the neuroendocrine signature by immunohistochemistry is mandatory for the diagnosis; a minimum panel comprising chromogranin A and synaptophysin is recommended in the digestive system. In addition, the application of grading requires the mitotic count and or spotty necrosis assessment for lung, or the mitotic count and the Ki67 assessment in the digestive system.

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

Access this article

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

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3

Similar content being viewed by others

References

  1. Oberndorfer S. Karzinoide Tumoren des Dünndarms. Frankf Z Pathol Int. 1907;1:425–32.

    Google Scholar 

  2. Kloppel G. Oberndorfer and his successors: from carcinoid to neuroendocrine carcinoma. Endocr Pathol. 2007;18(3):141–4. https://doi.org/10.1007/s12022-007-0021-9.

    Article  PubMed  Google Scholar 

  3. Inzani F, Rindi G. Classification of neuroendocrine neoplasms. In: Pacak K, Taieb D, editors. Radionuclide imaging and therapy for endocrine tumors. Cham: Humana Press; 2017. p. 1–13.

    Google Scholar 

  4. Rindi G, Wiedenmann B. Neuroendocrine neoplasms of the gut and pancreas: new insights. Nat Rev Endocrinol. 2012;8(1):54–64. https://doi.org/10.1038/nrendo.2011.120.

    Article  Google Scholar 

  5. Adams MS, Bronner-Fraser M. Review: the role of neural crest cells in the endocrine system. Endocr Pathol. 2009;20(2):92–100. https://doi.org/10.1007/s12022-009-9070-6.

    Article  CAS  PubMed  Google Scholar 

  6. Pearse AG. The diffuse neuroendocrine system and the apud concept: related "endocrine" peptides in brain, intestine, pituitary, placenta, and anuran cutaneous glands. La Medicina Biologica. 1977;55(3):115–25.

    CAS  Google Scholar 

  7. DeLellis RA, Lloyd RV, Heitz PU, Eng C. Pathology and genetics of Tumours of endocrine organs. 3rd ed. World Health Organization classification of Tumours. Lyon: IARC Press; 2004.

    Google Scholar 

  8. Barnes L, Eveson JW, Reichart P, Sidransky D. Pathology and genetics of Tumours of head and neck. World Health Organization classification of Tumours. Lyon: IARC Press; 2005.

    Google Scholar 

  9. LeBoit PE, Burg G, Weedon D, Sarasain A. Pathology and genetics of skin Tumours. 3rd ed. World Health Organization classification of Tumours. Lyon: IARC Press; 2006.

    Google Scholar 

  10. Bosman F, Carneiro F, Hruban RH, Theise ND. Pathology and genetics of Tumours of the digestive system. 4th ed. World Health Organization classification of Tumours. Lyon: IARC Press; 2010.

    Google Scholar 

  11. Lakhani SR, Ellis IO, Schnitt SJ, Tan PH, van de Vijver MJ. Pathology and genetics of Tumours of the breast. 4th ed. World Health Organization classification of Tumours. Lyon: IARC Press; 2012.

    Google Scholar 

  12. Travis WD, Brambilla E, Burke AP, Marx A, Nicholson AG. Pathology and genetics of Tumours of the lung, pleura, thymus and heart. 4th ed. World Health Organization classification of Tumours. Lyon: IARC Press; 2015.

    Google Scholar 

  13. Moch H, Humphrey PA, Ulbright TM, Reuter VE. Pathology and genetics of Tumours of the urinary system and male genital organs. 4th ed. World Health Organization classification of Tumours. Lyon: IARC Press; 2016.

    Google Scholar 

  14. Leoncini E, Boffetta P, Shafir M, Aleksovska K, Boccia S, Rindi G. Increased incidence trend of low-grade and high-grade neuroendocrine neoplasms. Endocrine. 2017; https://doi.org/10.1007/s12020-017-1273-x.

  15. Dasari A, Shen C, Halperin D, Zhao B, Zhou S, Xu Y, et al. Trends in the incidence, prevalence, and survival outcomes in patients with neuroendocrine tumors in the United States. JAMA Oncol. 2017; https://doi.org/10.1001/jamaoncol.2017.0589.

  16. Amin MB. AJCC cancer staging manual. VIII ed. New York: Springer-Verlag; 2017.

    Book  Google Scholar 

  17. Lloyd RV, Osamura R, Kloppel G, Rosai J. WHO classification of Tumours of endocrine organs. 4th ed. WHO Classification of Tumours. Lyon: IARC Press; 2017.

    Google Scholar 

  18. Travis WD, Linnoila RI, Tsokos MG, Hitchcock CL, Cutler GB Jr, Nieman L, et al. Neuroendocrine tumors of the lung with proposed criteria for large-cell neuroendocrine carcinoma. An ultrastructural, immunohistochemical, and flow cytometric study of 35 cases. Am J Surg Pathol. 1991;15(6):529–53.

    Article  CAS  PubMed  Google Scholar 

  19. Righi L, Volante M, Rapa I, Tavaglione V, Inzani F, Pelosi G, et al. Mammalian target of rapamycin signaling activation patterns in neuroendocrine tumors of the lung. Endocr Relat Cancer. 2010;17(4):977–87. https://doi.org/10.1677/ERC-10-0157.

    Article  CAS  PubMed  Google Scholar 

  20. Rindi G, Klersy C, Inzani F, Fellegara G, Ampollini L, Ardizzoni A, et al. Grading the neuroendocrine tumors of the lung: an evidence-based proposal. Endocr Relat Cancer. 2014;21(1):1–16. https://doi.org/10.1530/ERC-13-0246.

    Article  CAS  PubMed  Google Scholar 

  21. Chansky K, Detterbeck FC, Nicholson AG, Rusch VW, Vallieres E, Groome P, et al. The IASLC lung cancer staging project: external validation of the revision of the TNM stage groupings in the eighth edition of the TNM classification of lung cancer. J Thorac Oncol. 2017; https://doi.org/10.1016/j.jtho.2017.04.011.

  22. Pape UF, Jann H, Muller-Nordhorn J, Bockelbrink A, Berndt U, Willich SN, et al. Prognostic relevance of a novel TNM classification system for upper gastroenteropancreatic neuroendocrine tumors. Cancer. 2008;113(2):256–65. https://doi.org/10.1002/cncr.23549.

    Article  PubMed  Google Scholar 

  23. La Rosa S, Inzani F, Vanoli A, Klersy C, Dainese L, Rindi G, et al. Histologic characterization and improved prognostic evaluation of 209 gastric neuroendocrine neoplasms. Hum Pathol. 2011;42(10):1373–84. https://doi.org/10.1016/j.humpath.2011.01.018.

    Article  PubMed  Google Scholar 

  24. Jann H, Roll S, Couvelard A, Hentic O, Pavel M, Muller-Nordhorn J, et al. Neuroendocrine tumors of midgut and hindgut origin: tumor-node-metastasis classification determines clinical outcome. Cancer. 2011;117(15):3332–41. https://doi.org/10.1002/cncr.25855.

    Article  PubMed  Google Scholar 

  25. Norlen O, Stalberg P, Oberg K, Eriksson J, Hedberg J, Hessman O, et al. Long-term results of surgery for small intestinal neuroendocrine tumors at a tertiary referral center. World J Surg. 2012;36(6):1419–31. https://doi.org/10.1007/s00268-011-1296-z.

    Article  PubMed  Google Scholar 

  26. Rindi G, Falconi M, Klersy C, Albarello L, Boninsegna L, Buchler MW, et al. TNM staging of neoplasms of the endocrine pancreas: results from a large international cohort study. J Natl Cancer Inst. 2012;104(10):764–77. https://doi.org/10.1093/jnci/djs208.

    Article  CAS  PubMed  Google Scholar 

  27. Ciaccio M. Sur une nouvelle espèce cellulaire dans les glandes de Lieberkuhn. CR Seances Soc Biol Fil (Paris). 1906;60:76–7.

    Google Scholar 

  28. Gosset A, Masson P. Tumeurs endocrines de l’appendice. Presse Med. 1914;25:237–40.

    Google Scholar 

  29. Masson P. Carcinoids (Argentaffin-Cell Tumors) and Nerve Hyperplasia of the Appendicular Mucosa. Am J Pathol. 1928;4(3):181–212 19.

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Thorson A, Biorck G, Bjorkman G, Waldenstrom J. Malignant carcinoid of the small intestine with metastases to the liver, valvular disease of the right side of the heart (pulmonary stenosis and tricuspid regurgitation without septal defects), peripheral vasomotor symptoms, bronchoconstriction, and an unusual type of cyanosis; a clinical and pathologic syndrome. Am Heart J. 1954;47(5):795–817.

    Article  CAS  PubMed  Google Scholar 

  31. de Herder WW, Rehfeld JF, Kidd M, Modlin IM. A short history of neuroendocrine tumours and their peptide hormones. Best Pract Res Clin Endocrinol Metab. 2016;30(1):3–17. https://doi.org/10.1016/j.beem.2015.10.004.

    Article  PubMed  Google Scholar 

  32. Williams ED, Azzopardi JG. Tumours of the lung and the carcinoid syndrome. Thorax. 1960;15:30–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Caplin ME, Baudin E, Ferolla P, Filosso P, Garcia-Yuste M, Lim E, et al. Pulmonary neuroendocrine (carcinoid) tumors: European neuroendocrine tumor society expert consensus and recommendations for best practice for typical and atypical pulmonary carcinoids. Ann Oncol. 2015;26(8):1604–20. https://doi.org/10.1093/annonc/mdv041.

    Article  CAS  PubMed  Google Scholar 

  34. Rindi G, Kloppel G, Alhman H, Caplin M, Couvelard A, de Herder WW, et al. TNM staging of foregut (neuro)endocrine tumors: a consensus proposal including a grading system. Virchows Arch. 2006;449(4):395–401. https://doi.org/10.1007/s00428-006-0250-1.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Rindi G, Kloppel G, Couvelard A, Komminoth P, Korner M, Lopes JM, et al. TNM staging of midgut and hindgut (neuro) endocrine tumors: a consensus proposal including a grading system. Virchows Arch. 2007;451(4):757–62. https://doi.org/10.1007/s00428-007-0452-1.

    Article  CAS  PubMed  Google Scholar 

  36. Weynand B, Borbath I, Bernard V, Sempoux C, Gigot JF, Hubert C, et al. Pancreatic neuroendocrine tumour grading on endoscopic ultrasound-guided fine needle aspiration: high reproducibility and inter-observer agreement of the Ki-67 labelling index. Cytopathology. 2014;25(6):389–95. https://doi.org/10.1111/cyt.12111.

    CAS  PubMed  Google Scholar 

  37. Biancosino C, Kruger M, Vollmer E, Welker L. Intraoperative fine needle aspirations - diagnosis and typing of lung cancer in small biopsies: challenges and limitations. Diagn Pathol. 2016;11(1):59. https://doi.org/10.1186/s13000-016-0510-6.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Larghi A, Capurso G, Carnuccio A, Ricci R, Alfieri S, Galasso D, et al. Ki-67 grading of nonfunctioning pancreatic neuroendocrine tumors on histologic samples obtained by EUS-guided fine-needle tissue acquisition: a prospective study. Gastrointest Endosc. 2012;76(3):570–7. https://doi.org/10.1016/j.gie.2012.04.477.

    Article  PubMed  Google Scholar 

  39. Soga J, Tazawa K. Pathologic analysis of carcinoids; histologic reevaluation of 62 cases. Cancer. 1971;28:990–8.

    Article  CAS  PubMed  Google Scholar 

  40. Sorbye H, Welin S, Langer SW, Vestermark LW, Holt N, Osterlund P, et al. Predictive and prognostic factors for treatment and survival in 305 patients with advanced gastrointestinal neuroendocrine carcinoma (WHO G3): the NORDIC NEC study. Annals of oncology : official journal of the European Society for Medical Oncology / ESMO. 2012; https://doi.org/10.1093/annonc/mds276.

  41. Velayoudom-Cephise FL, Duvillard P, Foucan L, Hadoux J, Chougnet CN, Leboulleux S, et al. Are G3 ENETS neuroendocrine neoplasms heterogeneous? Endocr Relat Cancer. 2013;20(5):649–57. https://doi.org/10.1530/ERC-13-0027.

    Article  PubMed  Google Scholar 

  42. Heetfeld M, Chougnet CN, Olsen IH, Rinke A, Borbath I, Crespo G, et al. Characteristics and treatment of patients with G3 gastroenteropancreatic neuroendocrine neoplasms. Endocr Relat Cancer. 2015;22(4):657–64. https://doi.org/10.1530/ERC-15-0119.

    Article  CAS  PubMed  Google Scholar 

  43. Basturk O, Yang Z, Tang LH, Hruban RH, Adsay V, McCall CM, 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):683–90. https://doi.org/10.1097/PAS.0000000000000408.

    Article  PubMed  PubMed Central  Google Scholar 

  44. Milione M, Maisonneuve P, Spada F, Pellegrinelli A, Spaggiari P, Albarello L, 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. https://doi.org/10.1159/000445165.

    Article  CAS  PubMed  Google Scholar 

  45. Tang LH, Untch BR, Reidy DL, O'Reilly E, Dhall D, Jih L, et al. Well-differentiated neuroendocrine tumors with a morphologically apparent high-grade component: a pathway distinct from poorly differentiated neuroendocrine carcinomas. Clin Cancer Res. 2016;22(4):1011–7. https://doi.org/10.1158/1078-0432.CCR-15-0548.

    Article  CAS  PubMed  Google Scholar 

  46. Tang LH, Basturk O, Sue JJ, Klimstra DS. A practical approach to the classification of WHO grade 3 (G3) well-differentiated neuroendocrine tumor (WD-NET) and poorly differentiated neuroendocrine carcinoma (PD-NEC) of the pancreas. Am J Surg Pathol. 2016;40(9):1192–202. https://doi.org/10.1097/PAS.0000000000000662.

    Article  PubMed  PubMed Central  Google Scholar 

  47. Rindi G, Leiter AB, Kopin AS, Bordi C, Solcia E. The "normal" endocrine cell of the gut: changing concepts and new evidences. Ann N Y Acad Sci. 2004;1014:1–12.

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  49. Pelosi G, Rodriguez J, Viale G, Rosai J. Typical and atypical pulmonary carcinoid tumor overdiagnosed as small-cell carcinoma on biopsy specimens: a major pitfall in the management of lung cancer patients. Am J Surg Pathol. 2005;29(2):179–87.

    Article  PubMed  Google Scholar 

  50. Koo J, Mertens RB, Mirocha JM, Wang HL, Dhall D. Value of islet 1 and PAX8 in identifying metastatic neuroendocrine tumors of pancreatic origin. Mod Pathol. 2012;25(6):893–901. https://doi.org/10.1038/modpathol.2012.34.

    Article  CAS  PubMed  Google Scholar 

  51. McCluggage WG, Oliva E, Connolly LE, McBride HA, Young RH. An immunohistochemical analysis of ovarian small cell carcinoma of hypercalcemic type. Int J Gynecol Pathol. 2004;23(4):330–6.

    Article  CAS  PubMed  Google Scholar 

  52. McCluggage WG, Kennedy K, Busam KJ. An immunohistochemical study of cervical neuroendocrine carcinomas: neoplasms that are commonly TTF1 positive and which may express CK20 and P63. Am J Surg Pathol. 2010;34(4):525–32. https://doi.org/10.1097/PAS.0b013e3181d1d457.

    Article  PubMed  Google Scholar 

  53. Korner M, Waser B, Schonbrunn A, Perren A, Reubi JC. Somatostatin receptor subtype 2A immunohistochemistry using a new monoclonal antibody selects tumors suitable for in vivo somatostatin receptor targeting. Am J Surg Pathol. 2012;36(2):242–52. https://doi.org/10.1097/PAS.0b013e31823d07f3.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Fischer T, Doll C, Jacobs S, Kolodziej A, Stumm R, Schulz S. Reassessment of sst2 somatostatin receptor expression in human normal and neoplastic tissues using the novel rabbit monoclonal antibody UMB-1. J Clin Endocrinol Metab. 2008;93(11):4519–24. https://doi.org/10.1210/jc.2008-1063.

    Article  CAS  PubMed  Google Scholar 

  55. Lupp A, Hunder A, Petrich A, Nagel F, Doll C, Schulz S. Reassessment of sst(5) somatostatin receptor expression in normal and neoplastic human tissues using the novel rabbit monoclonal antibody UMB-4. Neuroendocrinology. 2011;94(3):255–64. https://doi.org/10.1159/000329876.

    Article  CAS  PubMed  Google Scholar 

  56. Vanoli A, La Rosa S, Klersy C, Grillo F, Albarello L, Inzani F, et al. Four neuroendocrine tumor types and neuroendocrine carcinoma of the duodenum: analysis of 203 cases. Neuroendocrinology. 2017;104(2):112–25. https://doi.org/10.1159/000444803.

    Article  CAS  PubMed  Google Scholar 

  57. Basturk O, Tang L, Hruban RH, Adsay V, Yang Z, Krasinskas AM, et al. Poorly differentiated neuroendocrine carcinomas of the pancreas: a clinicopathologic analysis of 44 cases. Am J Surg Pathol. 2014;38(4):437–47. https://doi.org/10.1097/PAS.0000000000000169.

    Article  PubMed  PubMed Central  Google Scholar 

  58. O'Toole D, Kianmanesh R, Caplin M. ENETS 2016 consensus guidelines for the Management of Patients with digestive neuroendocrine tumors: an update. Neuroendocrinology. 2016;103(2):117–8. https://doi.org/10.1159/000443169.

    Article  PubMed  Google Scholar 

  59. Oberg K, Hellman P, Ferolla P, Papotti M, Group EGW. Neuroendocrine bronchial and thymic tumors: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2012;23(Suppl 7):vii120–3. doi:https://doi.org/10.1093/annonc/mds267.

  60. Oberg K, Knigge U, Kwekkeboom D, Perren A, Group EGW. Neuroendocrine gastro-entero-pancreatic tumors: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2012;23(Suppl 7):vii124–30. doi:https://doi.org/10.1093/annonc/mds295.

  61. Garcia-Carbonero R, Sorbye H, Baudin E, Raymond E, Wiedenmann B, Niederle B, et al. ENETS consensus guidelines for high-grade Gastroenteropancreatic neuroendocrine tumors and neuroendocrine carcinomas. Neuroendocrinology. 2016;103(2):186–94. https://doi.org/10.1159/000443172.

    Article  CAS  PubMed  Google Scholar 

  62. Vinik AI, Woltering EA, Warner RR, Caplin M, O'Dorisio TM, Wiseman GA, et al. NANETS consensus guidelines for the diagnosis of neuroendocrine tumor. Pancreas. 2010;39(6):713–34. https://doi.org/10.1097/MPA.0b013e3181ebaffd.

    Article  PubMed  Google Scholar 

  63. Neuroendocrine Tumors - NCCN Guidelines. https://www.nccn.org/professionals/physician_gls/f_guidelines.asp.

  64. Rindi G, Petrone G, Inzani F. 25 years of neuroendocrine neoplasms of the gastrointestinal tract. Endocr Pathol. 2014;25(1):59–64. https://doi.org/10.1007/s12022-013-9292-5.

    Article  CAS  PubMed  Google Scholar 

  65. Rinke A, Muller HH, Schade-Brittinger C, Klose KJ, Barth P, Wied M, 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(28):4656–63. https://doi.org/10.1200/JCO.2009.22.8510.

    Article  CAS  PubMed  Google Scholar 

  66. Yao JC, Shah MH, Tetsuhide I, Lombard Bohas C, Wolin EM, Van Cutsem E, et al. Everolimus for advanced pancreatic neuroendocrine tumors. N Engl J Med. 2011;364(6):514–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Raymond E, Dahan L, Raoul JL, Bang YJ, Borbath I, Lombard-Bohas C, et al. Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. N Engl J Med. 2011;364(6):501–13. https://doi.org/10.1056/NEJMoa1003825.

    Article  CAS  PubMed  Google Scholar 

  68. Caplin ME, Pavel M, Cwikla JB, Phan AT, Raderer M, Sedlackova E, et al. Lanreotide in metastatic enteropancreatic neuroendocrine tumors. N Engl J Med. 2014;371(3):224–33. https://doi.org/10.1056/NEJMoa1316158.

    Article  PubMed  Google Scholar 

  69. Strosberg J, El-Haddad G, Wolin E, Hendifar A, Yao J, Chasen B, et al. Phase 3 trial of 177Lu-Dotatate for midgut neuroendocrine tumors. N Engl J Med. 2017;376(2):125–35. https://doi.org/10.1056/NEJMoa1607427.

    Article  CAS  PubMed  Google Scholar 

  70. Jiao Y, Shi C, Edil BH, de Wilde RF, Klimstra DS, Maitra A, et al. DAXX/ATRX, MEN1, and mTOR pathway genes are frequently altered in pancreatic neuroendocrine tumors. Science. 2011;331(6021):1199–203. https://doi.org/10.1126/science.1200609.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Banck MS, Kanwar R, Kulkarni AA, Boora GK, Metge F, Kipp BR, et al. The genomic landscape of small intestine neuroendocrine tumors. J Clin Invest. 2013;123(6):2502–8. https://doi.org/10.1172/JCI67963.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Francis JM, Kiezun A, Ramos AH, Serra S, Pedamallu CS, Qian ZR, et al. Somatic mutation of CDKN1B in small intestine neuroendocrine tumors. Nat Genet. 2013;45(12):1483–6. https://doi.org/10.1038/ng.2821.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Scarpa A, Chang DK, Nones K, Corbo V, Patch AM, Bailey P, et al. Whole-genome landscape of pancreatic neuroendocrine tumours. Nature. 2017;543(7643):65–71. https://doi.org/10.1038/nature21063.

    Article  CAS  PubMed  Google Scholar 

  74. Righi L, Volante M, Rapa I, Vatrano S, Pelosi G, Papotti M. Therapeutic biomarkers in lung neuroendocrine neoplasia. Endocr Pathol. 2014;25(4):371–7. https://doi.org/10.1007/s12022-014-9335-6.

    Article  CAS  PubMed  Google Scholar 

  75. Oksuz MO, Winter L, Pfannenberg C, Reischl G, Mussig K, Bares R, et al. Peptide receptor radionuclide therapy of neuroendocrine tumors with (90)Y-DOTATOC: is treatment response predictable by pre-therapeutic uptake of (68)Ga-DOTATOC? Diagn Interv Imaging. 2014;95(3):289–300. https://doi.org/10.1016/j.diii.2013.07.006.

    Article  CAS  PubMed  Google Scholar 

  76. Zatelli MC, Fanciulli G, Malandrino P, Ramundo V, Faggiano A, Colao A, et al. Predictive factors of response to mTOR inhibitors in neuroendocrine tumours. Endocr Relat Cancer. 2016;23(3):R173–83. https://doi.org/10.1530/ERC-15-0413.

    Article  CAS  PubMed  Google Scholar 

Download references

Funding

In part supported by internal university grants (Università Cattolica line D.1 2014–70,201,266) and by the Associazione Italiana Ricerca sul Cancro - AIRC IG 2013 14,696 to GR. The funders had no role in the study design and data analysis.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Guido Rindi.

Ethics declarations

Conflict of interest

GR declares that he has received speaker’s fee by Novartis Pharma and Ipsen Pharma. All remaining authors have declared no conflicts of interest.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Inzani, F., Petrone, G., Fadda, G. et al. Cyto-histology in NET: what is necessary today and what is the future?. Rev Endocr Metab Disord 18, 381–391 (2017). https://doi.org/10.1007/s11154-017-9428-x

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11154-017-9428-x

Keywords

Navigation