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:

Neuroendocrine neoplasia of the gastrointestinal tract revisited: towards precision medicine

Abstract

Over the past 5 years, a number of notable research advances have been made in the field of neuroendocrine cancer, specifically with regard to neuroendocrine cancer of the gastrointestinal tract. The aim of this Review is to provide an update on current knowledge that has proven effective for the clinical management of patients with these tumours. For example, for the first time in the tubular gastrointestinal tract, well-differentiated high-grade (grade 3) tumours and mixed neuroendocrine–non-neuroendocrine neoplasms (MiNENs) are defined in the WHO classification. This novel classification enables efficient identification of the most aggressive well-differentiated neuroendocrine tumours and helps in defining the degree of aggressiveness of MiNENs. The Review also discusses updates to epidemiology, cell biology (including vesicle-specific components) and the as-yet-unresolved complex genetic background that varies according to site and differentiation status. The Review summarizes novel diagnostic instruments, including molecules associated with the secretory machinery, novel radiological approaches (including pattern recognition techniques), novel PET tracers and liquid biopsy combined with DNA or RNA assays. Surgery remains the treatment mainstay; however, peptide receptor radionuclide therapy with novel radioligands and new emerging medical therapies (including vaccination and immunotherapy) are evolving and being tested in clinical trials, which are summarized and critically reviewed here.

Key points

  • Neuroendocrine neoplasms (NENs) are made up of two separate groups, well-differentiated, termed neuroendocrine tumour (NET), and poorly differentiated, termed neuroendocrine carcinoma (NEC).

  • WHO 2019 classification of gut NENs grades NETs as grade 1 to grade 3 based on tumour morphology, tumour cell proliferation measured in terms of the Ki67 proliferation index and/or the mitotic count, whereas NECs are by definition grade 3.

  • The genetic landscape of gut NETs is poorly understood and mainly involves chromosomal alterations combined with epigenetic changes.

  • Novel diagnostic approaches include neuroendocrine tissue markers, liquid biopsy for multigene detection, radiomics with computer-assisted diagnosis for CT and MRI, PET and novel endoscopy approaches including operational video capsule endoscopy.

  • Novel therapies include tyrosine kinase inhibitors, immunotherapy, vaccination, surgical debulking and locoregional treatments.

  • Research effort should target the personalized management of patients with NEN exploring the relationship between genetic background, NEN development and tumour response to therapy.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: Neuroendocrine cell secretion properties, control and neuroendocrine neoplasia.
Fig. 2: The relationship between size of primary neuroendocrine neoplasia and metastasis.

Similar content being viewed by others

References

  1. Rindi, G. & Wiedenmann, B. Neuroendocrine neoplasms of the gut and pancreas: new insights. Nat. Rev. Endocrinol. 8, 54–64 (2012).

    Google Scholar 

  2. Kurman, R. J., Carcangiu, M. L., Herrington, C. S. & Young, R. H. (eds) WHO Classification of Tumours of Female Reproductive Organs 4th edn Vol. 5 (IARC Press, 2014).

  3. Travis, W. D., Brambilla, E., Burke, A. P., Marx, A. & Nicholson, A. G. (eds) WHO Classification of Tumours of the Lung, Pleura, Thymus and Heart 4th edn Vol. 7 (IARC Press, 2015).

  4. Moch, H., Humphrey, P. A., Ulbright, T. M. & Reuter, V. E. (eds) Pathology and Genetics of Tumours of the Urinary System and Male Genital Organs 4th edn Vol. 8 (IARC Press, 2016).

  5. Lloyd, R. V., Osamura, R., Klöppel, G. & Rosai, J. (eds) WHO Classification of Tumours of Endocrine Organs 4th edn Vol. 10 (IARC Press, 2017).

  6. El-Naggar, A. K., Chan, J. K. C., Grandis, J. R., Takata, T. & Slootweg, P. J. (eds) WHO Classification of Head and Neck Tumours Vol. 9 (IARC Press, 2017).

  7. Elder, D. E., Massi, D., Scolyer, R. A. & Willemze, R. (eds) WHO Classification of Skin Tumours. 4th edn, Vol. 11 (IARC Press, 2018).

  8. WHO Classification of Tumours Editorial Board. Digestive System Tumours 5th edn Vol. 1 (IARC Press, 2019).

  9. WHO Classification of Tumours Editorial Board. Breast Tumours 5th edn Vol. 2 (IARC Press, 2019).

  10. Rindi, G. et al. A common classification framework for neuroendocrine neoplasms: an International Agency for Research on Cancer (IARC) and World Health Organization (WHO) expert consensus proposal. Mod. Pathol. 31, 1770–1786 (2018).

    PubMed  PubMed Central  Google Scholar 

  11. Klimstra, D., Klöppel, G., La Rosa, S. & Rindi, G. in Digestive System Tumours Vol. 1 (ed. WHO Classification of Tumours Editorial Board) 16–19 (IARC Press, 2019).

  12. Frost, M., Lines, K. E. & Thakker, R. V. Current and emerging therapies for PNETs in patients with or without MEN1. Nat. Rev. Endocrinol. 14, 216–227 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Dasari, A. et al. Trends in the incidence, prevalence, and survival outcomes in patients with neuroendocrine tumors in the United States. JAMA Oncol. 3, 1335–1342 (2017).

    PubMed  PubMed Central  Google Scholar 

  14. Leoncini, E. et al. Increased incidence trend of low-grade and high-grade neuroendocrine neoplasms. Endocrine 58, 368–379 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Hallet, J. et al. Exploring the rising incidence of neuroendocrine tumors: a population-based analysis of epidemiology, metastatic presentation, and outcomes. Cancer 121, 589–597 (2015).

    PubMed  Google Scholar 

  16. Ellis, L., Shale, M. J. & Coleman, M. P. Carcinoid tumors of the gastrointestinal tract: trends in incidence in England since 1971. Am. J. Gastroenterol. 105, 2563–2569 (2010).

    PubMed  Google Scholar 

  17. Korse, C. M., Taal, B. G., van Velthuysen, M. L. & Visser, O. Incidence and survival of neuroendocrine tumours in the Netherlands according to histological grade: experience of two decades of cancer registry. Eur. J. Cancer 49, 1975–1983 (2013).

    PubMed  Google Scholar 

  18. Riihimaki, M., Hemminki, A., Sundquist, K., Sundquist, J. & Hemminki, K. The epidemiology of metastases in neuroendocrine tumors. Int. J. Cancer 139, 2679–2686 (2016).

    PubMed  Google Scholar 

  19. Leoncini, E., Carioli, G., La Vecchia, C., Boccia, S. & Rindi, G. Risk factors for neuroendocrine neoplasms: a systematic review and meta-analysis. Ann. Oncol. 27, 68–81 (2016).

    CAS  PubMed  Google Scholar 

  20. Rindi, G. et al. in WHO Classification of Tumours of the Digestive System (eds Bosman, F., Carneiro, F., Hruban, R. H. & Theise, N. D.) 10–12 (IARC Press, 2010).

  21. Klöppel, G. et al. in WHO Classification of Tumours of Endocrine Organs (eds Lloyd, R. V., Osamura, R. Y., Klöppel, G., & Rosai, J.) (IARC Press, 2017).

  22. Velayoudom-Cephise, F. L. et al. Are G3 ENETS neuroendocrine neoplasms heterogeneous? Endocr. Relat. Cancer 20, 649–657 (2013).

    PubMed  Google Scholar 

  23. Heetfeld, M. et al. Characteristics and treatment of patients with G3 gastroenteropancreatic neuroendocrine neoplasms. Endocr. Relat. Cancer 22, 657–664 (2015).

    CAS  PubMed  Google Scholar 

  24. Basturk, O. 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. 39, 683–690 (2015).

    PubMed  PubMed Central  Google Scholar 

  25. Hijioka, S. et al. Does the WHO 2010 classification of pancreatic neuroendocrine neoplasms accurately characterize pancreatic neuroendocrine carcinomas? J. Gastroenterol. 50, 564–572 (2015).

    CAS  PubMed  Google Scholar 

  26. Rindi, G. et al. Competitive testing of the WHO 2010 versus the WHO 2017 grading of pancreatic neuroendocrine neoplasms: data from a large international cohort study. Neuroendocrinology 107, 375–386 (2018).

    CAS  PubMed  Google Scholar 

  27. Sorbye, H. 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. 24, 152–160 (2013).

    CAS  PubMed  Google Scholar 

  28. Milione, M. et al. The clinicopathologic heterogeneity of grade 3 gastroenteropancreatic neuroendocrine neoplasms: morphological differentiation and proliferation identify different prognostic categories. Neuroendocrinology 104, 85–93 (2017).

    CAS  PubMed  Google Scholar 

  29. Milione, M. et al. Ki67 proliferative index of the neuroendocrine component drives MANEC prognosis. Endocr. Relat. Cancer 25, 583–593 (2018).

    CAS  PubMed  Google Scholar 

  30. Kidd, M., Modlin, I. & Oberg, K. Towards a new classification of gastroenteropancreatic neuroendocrine neoplasms. Nat. Rev. Clin. Oncol. 13, 691–705 (2016).

    CAS  PubMed  Google Scholar 

  31. Solcia E. C. C., Buffa R., Usellini L., Fiocca R. & Sessa F. in Physiology of the Gastrointestinal Tract (ed Johnson, L. R.) 401–420 (Raven Press, 1986).

  32. Rindi, G., Leiter, A. B., Kopin, A. S., Bordi, C. & Solcia, E. The “normal” endocrine cell of the gut: changing concepts and new evidences. Ann. NY Acad. Sci. 1014, 1–12 (2004).

    CAS  PubMed  Google Scholar 

  33. Solcia, E. et al. in New Concepts in Neoplasia as Applied to Diagnostic Pathology (eds Fenoglio-Preiser, C. M., Weinstein, R. S. & Kaufman, N.) 242–261 (Williams & Wilkins, 1986).

  34. DeLellis, R. A., Tischler, A. S. & Wolfe, H. J. Multidirectional differentiation in neuroendocrine neoplasms. J. Histochem. Cytochem. 32, 899–904 (1984).

    CAS  PubMed  Google Scholar 

  35. Hanahan, D. Heritable formation of pancreatic β-cell tumours in transgenic mice expressing recombinant insulin/simian virus 40 oncogenes. Nature 315, 115–122 (1985).

    CAS  PubMed  Google Scholar 

  36. Rindi, G. et al. Development of neuroendocrine tumors in the gastrointestinal tract of transgenic mice. Heterogeneity of hormone expression. Am. J. Pathol. 136, 1349–1363 (1990).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Lopez, M. J., Upchurch, B. H., Rindi, G. & Leiter, A. B. Studies in transgenic mice reveal potential relationships between secretin-producing cells and other endocrine cell types. J. Biol. Chem. 270, 885–891 (1995).

    CAS  PubMed  Google Scholar 

  38. Upchurch, B. H., Fung, B. P., Rindi, G., Ronco, A. & Leiter, A. B. Peptide YY expression is an early event in colonic endocrine cell differentiation: evidence from normal and transgenic mice. Development 122, 1157–1163 (1996).

    CAS  PubMed  Google Scholar 

  39. Gehart, H. et al. Identification of enteroendocrine regulators by real-time single-cell differentiation mapping. Cell 176, 1158–1173.e16 (2019).

    CAS  PubMed  Google Scholar 

  40. Rindi, G. et al. Targeted ablation of secretin-producing cells in transgenic mice reveals a common differentiation pathway with multiple enteroendocrine cell lineages in the small intestine. Development 126, 4149–4156 (1999).

    CAS  PubMed  Google Scholar 

  41. Schonhoff, S. E., Giel-Moloney, M. & Leiter, A. B. Minireview: development and differentiation of gut endocrine cells. Endocrinology 145, 2639–2644 (2004).

    CAS  PubMed  Google Scholar 

  42. Li, H. J., Ray, S. K., Singh, N. K., Johnston, B. & Leiter, A. B. Basic helix-loop-helix transcription factors and enteroendocrine cell differentiation. Diabetes Obes. Metab. 13 (Suppl. 1), 5–12 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Beucher, A. et al. The homeodomain-containing transcription factors Arx and Pax4 control enteroendocrine subtype specification in mice. PLoS ONE 7, e36449 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Clevers, H. & Batlle, E. SnapShot: the intestinal crypt. Cell 152, 1198–1198 (2013).

    CAS  PubMed  Google Scholar 

  45. Grun, D. et al. Single-cell messenger RNA sequencing reveals rare intestinal cell types. Nature 525, 251–255 (2015).

    PubMed  Google Scholar 

  46. Beumer, J. et al. Enteroendocrine cells switch hormone expression along the crypt-to-villus BMP signalling gradient. Nat. Cell Biol. 20, 909–916 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Jenny, M. et al. Neurogenin3 is differentially required for endocrine cell fate specification in the intestinal and gastric epithelium. EMBO J. 21, 6338–6347 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Lee, C. S., Perreault, N., Brestelli, J. E. & Kaestner, K. H. Neurogenin 3 is essential for the proper specification of gastric enteroendocrine cells and the maintenance of gastric epithelial cell identity. Genes. Dev. 16, 1488–1497 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Wiedenmann, B. & Huttner, W. B. Synaptophysin and chromogranins/secretogranins–widespread constituents of distinct types of neuroendocrine vesicles and new tools in tumor diagnosis. Virchows Arch. B Cell Pathol. 58, 95–121 (1989).

    CAS  Google Scholar 

  50. Hocker, M. et al. Molecular dissection of regulated secretory pathways in human gastric enterochromaffin-like cells: an immunohistochemical analysis. Histochem. Cell Biol. 112, 205–214 (1999).

    CAS  PubMed  Google Scholar 

  51. Burre, J. & Volknandt, W. The synaptic vesicle proteome. J. Neurochem. 101, 1448–1462 (2007).

    CAS  PubMed  Google Scholar 

  52. Glassmeier, G. et al. Electrophysiological properties of human carcinoid cells of the gut. Gastroenterology 113, 90–100 (1997).

    CAS  PubMed  Google Scholar 

  53. Wiedenmann, B., John, M., Ahnert-Hilger, G. & Riecken, E. O. Molecular and cell biological aspects of neuroendocrine tumors of the gastroenteropancreatic system. J. Mol. Med. 76, 637–647 (1998).

    CAS  PubMed  Google Scholar 

  54. Suckale, J. & Solimena, M. The insulin secretory granule as a signaling hub. Trends Endocrinol. Metab. 21, 599–609 (2010).

    CAS  PubMed  Google Scholar 

  55. Alvarez, Y. D. & Marengo, F. D. The immediately releasable vesicle pool: highly coupled secretion in chromaffin and other neuroendocrine cells. J. Neurochem. 116, 155–163 (2011).

    CAS  PubMed  Google Scholar 

  56. Park, D. et al. Molecular organization of Drosophila neuroendocrine cells by dimmed. Curr. Biol. 21, 1515–1524 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Nakanishi, N., Takeuchi, F. & Tsubaki, M. Histidine cycle mechanism for the concerted proton/electron transfer from ascorbate to the cytosolic haem b centre of cytochrome b561: a unique machinery for the biological transmembrane electron transfer. J. Biochem. 142, 553–560 (2007).

    CAS  PubMed  Google Scholar 

  58. Iliadi, K. G. et al. nemy encodes a cytochrome b561 that is required for Drosophila learning and memory. Proc. Natl Acad. Sci. USA 105, 19986–19991 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Malsam, J., Kreye, S. & Sollner, T. H. Membrane fusion: SNAREs and regulation. Cell Mol. Life Sci. 65, 2814–2832 (2008).

    CAS  PubMed  Google Scholar 

  60. McNew, J. A. et al. Compartmental specificity of cellular membrane fusion encoded in SNARE proteins. Nature 407, 153–159 (2000).

    CAS  PubMed  Google Scholar 

  61. Sollner, T. et al. SNAP receptors implicated in vesicle targeting and fusion. Nature 362, 318–324 (1993).

    CAS  PubMed  Google Scholar 

  62. Jahn, R. & Fasshauer, D. Molecular machines governing exocytosis of synaptic vesicles. Nature 490, 201–207 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Wang, V. E. et al. A case of metastatic atypical neuroendocrine tumor with ALK translocation and diffuse brain metastases. Oncologist 22, 768–773 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Weber, T. et al. SNAREpins: minimal machinery for membrane fusion. Cell 92, 759–772 (1998).

    CAS  PubMed  Google Scholar 

  65. Hay, J. C. SNARE complex structure and function. Exp. Cell Res. 271, 10–21 (2001).

    CAS  PubMed  Google Scholar 

  66. Redecker, P. Expression of synaptic vesicle trafficking proteins in the developing rat pineal gland. Cell Tissue Res. 301, 255–265 (2000).

    CAS  PubMed  Google Scholar 

  67. Ahnert-Hilger, G., Kutay, U., Chahoud, I., Rapoport, T. & Wiedenmann, B. Synaptobrevin is essential for secretion but not for the development of synaptic processes. Eur. J. Cell Biol. 70, 1–11 (1996).

    CAS  PubMed  Google Scholar 

  68. Hocker, M. & Wiedenmann, B. Therapeutic and diagnostic implications of the somatostatin system in gastroenteropancreatic neuroendocrine tumour disease. Ital. J. Gastroenterol. Hepatol. 31 (Suppl. 2), 139–142 (1999).

    Google Scholar 

  69. Farquhar, M. G. Multiple pathways of exocytosis, endocytosis, and membrane recycling: validation of a Golgi route. Fed. Proc. 42, 2407–2413 (1983).

    CAS  PubMed  Google Scholar 

  70. Bauer, R. A., Overlease, R. L., Lieber, J. L. & Angleson, J. K. Retention and stimulus-dependent recycling of dense core vesicle content in neuroendocrine cells. J. Cell Sci. 117, 2193–2202 (2004).

    CAS  PubMed  Google Scholar 

  71. Tanner, V. A., Ploug, T. & Tao-Cheng, J. H. Subcellular localization of SV2 and other secretory vesicle components in PC12 cells by an efficient method of preembedding EM immunocytochemistry for cell cultures. J. Histochem. Cytochem. 44, 1481–1488 (1996).

    CAS  PubMed  Google Scholar 

  72. Duncan, R. R. et al. Functional and spatial segregation of secretory vesicle pools according to vesicle age. Nature 422, 176–180 (2003).

    CAS  PubMed  Google Scholar 

  73. von Blankenfeld, G. et al. Expression of functional GABAA receptors in neuroendocrine gastropancreatic cells. Pflug. Arch. 430, 381–388 (1995).

    Google Scholar 

  74. Somasundaram, A. & Taraska, J. W. Local protein dynamics during microvesicle exocytosis in neuroendocrine cells. Mol. Biol. Cell 29, 1891–1903 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Meng, F. et al. Identification of the receptors for somatostatin (SST) and cortistatin (CST) in chickens and investigation of the roles of cSST28, cSST14, and cCST14 in inhibiting cGHRH1-27NH2-induced growth hormone secretion in cultured chicken pituitary cells. Mol. Cell Endocrinol. 384, 83–95 (2014).

    CAS  PubMed  Google Scholar 

  76. Ben-Shlomo, R. Invasiveness, chimerism and genetic diversity. Mol. Ecol. 26, 6502–6509 (2017).

    CAS  PubMed  Google Scholar 

  77. Buscail, L. et al. Stimulation of tyrosine phosphatase and inhibition of cell proliferation by somatostatin analogues: mediation by human somatostatin receptor subtypes SSTR1 and SSTR2. Proc. Natl Acad. Sci. USA 91, 2315–2319 (1994).

    CAS  PubMed  PubMed Central  Google Scholar 

  78. Todisco, A., Campbell, V., Dickinson, C. J., DelValle, J. & Yamada, T. Molecular basis for somatostatin action: inhibition of c-fos expression and AP-1 binding. Am. J. Physiol. 267, G245–G253 (1994).

    CAS  PubMed  Google Scholar 

  79. Barbieri, F. et al. Peptide receptor targeting in cancer: the somatostatin paradigm. Int. J. Pept. 2013, 926295 (2013).

    PubMed  PubMed Central  Google Scholar 

  80. Aoki, T. et al. Somatostatin analog inhibits the growth of insulinoma cells by p27-mediated G1 cell cycle arrest. Pancreas 43, 720–729 (2014).

    CAS  PubMed  Google Scholar 

  81. Rocheville, M. et al. Receptors for dopamine and somatostatin: formation of hetero-oligomers with enhanced functional activity. Science 288, 154–157 (2000).

    CAS  PubMed  Google Scholar 

  82. Rocheville, M. et al. Subtypes of the somatostatin receptor assemble as functional homo- and heterodimers. J. Biol. Chem. 275, 7862–7869 (2000).

    CAS  PubMed  Google Scholar 

  83. Patel, R. C. et al. Ligand binding to somatostatin receptors induces receptor-specific oligomer formation in live cells. Proc. Natl Acad. Sci. USA 99, 3294–3299 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Csaba, Z., Peineau, S. & Dournaud, P. Molecular mechanisms of somatostatin receptor trafficking. J. Mol. Endocrinol. 48, R1–R12 (2012).

    CAS  PubMed  Google Scholar 

  85. Schafer, M. K., Weihe, E. & Eiden, L. E. Localization and expression of VMAT2 aross mammalian species: a translational guide for its visualization and targeting in health and disease. Adv. Pharmacol. 68, 319–334 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  86. Zhu, J., Klein-Fedyshin, M. & Stevenson, J. M. Serotonin transporter gene polymorphisms and selective serotonin reuptake inhibitor tolerability: review of pharmacogenetic evidence. Pharmacotherapy 37, 1089–1104 (2017).

    PubMed  Google Scholar 

  87. Wimalasena, K. Vesicular monoamine transporters: structure-function, pharmacology, and medicinal chemistry. Med. Res. Rev. 31, 483–519 (2011).

    CAS  PubMed  Google Scholar 

  88. Schaefer, M. TRPs: modulation by drug-like compounds. Handb. Exp. Pharmacol. 223, 1077–1106 (2014).

    CAS  PubMed  Google Scholar 

  89. Zhang, S., Li, N., Zeng, W., Gao, N. & Yang, M. Cryo-EM structures of the mammalian endo-lysosomal TRPML1 channel elucidate the combined regulation mechanism. Protein Cell 8, 834–847 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Mergler, S. et al. Transient receptor potential channel TRPM8 agonists stimulate calcium influx and neurotensin secretion in neuroendocrine tumor cells. Neuroendocrinology 85, 81–92 (2007).

    CAS  PubMed  Google Scholar 

  91. Mergler, S. et al. Thermo-sensitive transient receptor potential vanilloid channel-1 regulates intracellular calcium and triggers chromogranin a secretion in pancreatic neuroendocrine BON-1 tumor cells. Cell Signal. 24, 233–246 (2012).

    CAS  PubMed  Google Scholar 

  92. Zhang, X., Hu, M., Yang, Y. & Xu, H. Organellar TRP channels. Nat. Struct. Mol. Biol. 25, 1009–1018 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  93. Pape, U. F. et al. Prognostic relevance of a novel TNM classification system for upper gastroenteropancreatic neuroendocrine tumors. Cancer 113, 256–265 (2008).

    PubMed  Google Scholar 

  94. La Rosa, S. et al. Histologic characterization and improved prognostic evaluation of 209 gastric neuroendocrine neoplasms. Hum. Pathol. 42, 1373–1384 (2011).

    PubMed  Google Scholar 

  95. Jann, H. et al. Neuroendocrine tumors of midgut and hindgut origin: tumor-node-metastasis classification determines clinical outcome. Cancer 117, 3332–3341 (2011).

    PubMed  Google Scholar 

  96. Norlen, O. et al. Long-term results of surgery for small intestinal neuroendocrine tumors at a tertiary referral center. World J. Surg. 36, 1419–1431 (2012).

    PubMed  Google Scholar 

  97. Dhall, D. et al. Ki-67 proliferative index predicts progression-free survival of patients with well-differentiated ileal neuroendocrine tumors. Hum. Pathol. 43, 489–495 (2012).

    CAS  PubMed  Google Scholar 

  98. Weinstock, B. et al. Clinical and prognostic features of rectal neuroendocrine tumors. Neuroendocrinology 98, 180–187 (2013).

    CAS  PubMed  Google Scholar 

  99. Vanoli, A. et al. Four neuroendocrine tumor types and neuroendocrine carcinoma of the duodenum: analysis of 203 cases. Neuroendocrinology 104, 112–125 (2017).

    CAS  PubMed  Google Scholar 

  100. Vanoli, A. et al. Prognostic evaluations tailored to specific gastric neuroendocrine neoplasms: analysis of 200 cases with extended follow-up. Neuroendocrinology 107, 114–126 (2018).

    CAS  PubMed  Google Scholar 

  101. Vanoli, A. et al. Neuroendocrine tumors (NETs) of the minor papilla/ampulla: analysis of 16 cases underlines homology with major ampulla NETs and differences from extra-ampullary duodenal NETs. Am. J. Surg. Pathol. 43, 725–736 (2019).

    PubMed  Google Scholar 

  102. Halperin, D. M. et al. Frequency of carcinoid syndrome at neuroendocrine tumour diagnosis: a population-based study. Lancet Oncol. 18, 525–534 (2017).

    PubMed  PubMed Central  Google Scholar 

  103. Ito, T., Igarashi, H. & Jensen, R. T. Zollinger-Ellison syndrome: recent advances and controversies. Curr. Opin. Gastroenterol. 29, 650–661 (2013).

    PubMed  PubMed Central  Google Scholar 

  104. Ito, T., Lee, L. & Jensen, R. T. Carcinoid-syndrome: recent advances, current status and controversies. Curr. Opin. Endocrinol. Diabetes Obes. 25, 22–35 (2018).

    PubMed  PubMed Central  Google Scholar 

  105. Garbrecht, N. et al. Somatostatin-producing neuroendocrine tumors of the duodenum and pancreas: incidence, types, biological behavior, association with inherited syndromes, and functional activity. Endocr. Relat. Cancer 15, 229–241 (2008).

    PubMed  Google Scholar 

  106. Donow, C. et al. Surgical pathology of gastrinoma. Site, size, multicentricity, association with multiple endocrine neoplasia type 1, and malignancy. Cancer 68, 1329–1334 (1991).

    CAS  PubMed  Google Scholar 

  107. Anlauf, M. et al. Sporadic versus hereditary gastrinomas of the duodenum and pancreas: distinct clinico-pathological and epidemiological features. World J. Gastroenterol. 12, 5440–5446 (2006).

    PubMed  PubMed Central  Google Scholar 

  108. Strosberg, J. Neuroendocrine tumours of the small intestine. Best. Pract. Res. Clin. Gastroenterol. 26, 755–773 (2012).

    CAS  PubMed  Google Scholar 

  109. Bosman, F., Carneiro, F., Hruban, R. H. & Theise, N. D. WHO Classification of Tumours of the Digestive System 4th edn Vol. 3 (IARC Press, 2010).

  110. Crona, J. & Skogseid, B. GEP- NETS UPDATE: genetics of neuroendocrine tumors. Eur. J. Endocrinol. 174, R275–R290 (2016).

    CAS  PubMed  Google Scholar 

  111. Debelenko, L. V. et al. The multiple endocrine neoplasia type I gene locus is involved in the pathogenesis of type II gastric carcinoids. Gastroenterology 113, 773–781 (1997).

    CAS  PubMed  Google Scholar 

  112. Zhuang, Z. et al. Somatic mutations of the MEN1 tumor suppressor gene in sporadic gastrinomas and insulinomas. Cancer Res. 57, 4682–4686 (1997).

    CAS  PubMed  Google Scholar 

  113. Anlauf, M. et al. Allelic deletion of the MEN1 gene in duodenal gastrin and somatostatin cell neoplasms and their precursor lesions. Gut 56, 637–644 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  114. Lee, H. Y. & Garber, P. E. Von Recklinghausen’s disease associated with pheochromocytoma and carcinoid tumor. Ohio State Med. J. 66, 583–586 (1970).

    CAS  PubMed  Google Scholar 

  115. Alshikho, M. J. et al. Zollinger-Ellison syndrome associated with von Recklinghausen disease: case report and literature review. Am. J. Case Rep. 17, 398–405 (2016).

    PubMed  PubMed Central  Google Scholar 

  116. Fujii, T. et al. MEN1 gene mutations in sporadic neuroendocrine tumors of foregut derivation. Pathol. Int. 49, 968–973 (1999).

    CAS  PubMed  Google Scholar 

  117. Banck, M. S. et al. The genomic landscape of small intestine neuroendocrine tumors. J. Clin. Invest. 123, 2502–2508 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  118. Scarpa, A. et al. Whole-genome landscape of pancreatic neuroendocrine tumours. Nature 543, 65–71 (2017).

    CAS  PubMed  Google Scholar 

  119. Francis, J. M. et al. Somatic mutation of CDKN1B in small intestine neuroendocrine tumors. Nat. Genet. 45, 1483–1486 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  120. Crona, J. et al. Somatic mutations and genetic heterogeneity at the CDKN1B locus in small intestinal neuroendocrine tumors. Ann. Surg. Oncol. 22 (Suppl. 3), 1428–1435 (2015).

    Google Scholar 

  121. Simbolo, M. et al. Mutational and copy number asset of primary sporadic neuroendocrine tumors of the small intestine. Virchows Arch. 473, 709–717 (2018).

    PubMed  PubMed Central  Google Scholar 

  122. Kulke, M. H. et al. High-resolution analysis of genetic alterations in small bowel carcinoid tumors reveals areas of recurrent amplification and loss. Genes Chromosomes Cancer 47, 591–603 (2008).

    CAS  PubMed  Google Scholar 

  123. Cunningham, J. L. et al. Common pathogenetic mechanism involving human chromosome 18 in familial and sporadic ileal carcinoid tumors. Genes Chromosomes Cancer 50, 82–94 (2011).

    CAS  PubMed  Google Scholar 

  124. de Mestier, L. et al. Familial small-intestine carcinoids: chromosomal alterations and germline inositol polyphosphate multikinase sequencing. Dig. Liver Dis. 49, 98–102 (2017).

    PubMed  Google Scholar 

  125. Barazeghi, E., Hellman, P., Westin, G. & Stalberg, P. PTPRM, a candidate tumor suppressor gene in small intestinal neuroendocrine tumors. Endocr. Connect. 8, 1126–1135 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  126. Sei, Y. et al. A hereditary form of small intestinal carcinoid associated with a germline mutation in inositol polyphosphate multikinase. Gastroenterology 149, 67–78 (2015).

    CAS  PubMed  Google Scholar 

  127. Dumanski, J. P. et al. A MUTYH germline mutation is associated with small intestinal neuroendocrine tumors. Endocr. Relat. Cancer 24, 427–443 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  128. Karpathakis, A. et al. Prognostic impact of novel molecular subtypes of small intestinal neuroendocrine tumor. Clin. Cancer Res. 22, 250–258 (2016).

    CAS  PubMed  Google Scholar 

  129. Li, S. C. et al. Global microRNA profiling of well-differentiated small intestinal neuroendocrine tumors. Mod. Pathol. 26, 685–696 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  130. Miller, H. C. et al. MicroRNAs associated with small bowel neuroendocrine tumours and their metastases. Endocr. Relat. Cancer 23, 711–726 (2016).

    CAS  PubMed  Google Scholar 

  131. Verdugo, A. D. et al. Global DNA methylation patterns through an array-based approach in small intestinal neuroendocrine tumors. Endocr. Relat. Cancer 21, L5–L7 (2014).

    CAS  PubMed  Google Scholar 

  132. Finnerty, B. M., Gray, K. D., Moore, M. D., Zarnegar, R. & Fahey, T. J. III Epigenetics of gastroenteropancreatic neuroendocrine tumors: a clinicopathologic perspective. World J. Gastrointest. Oncol. 9, 341–353 (2017).

    PubMed  PubMed Central  Google Scholar 

  133. Edfeldt, K. et al. Different gene expression profiles in metastasizing midgut carcinoid tumors. Endocr. Relat. Cancer 18, 479–489 (2011).

    CAS  PubMed  Google Scholar 

  134. Alvarez, M. J. et al. A precision oncology approach to the pharmacological targeting of mechanistic dependencies in neuroendocrine tumors. Nat. Genet. 50, 979–989 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  135. Priestley, P. et al. Pan-cancer whole-genome analyses of metastatic solid tumours. Nature 575, 210–216 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  136. Ilett, E. E. et al. Neuroendocrine carcinomas of the gastroenteropancreatic system: a comprehensive review. Diagnostics 5, 119–176 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  137. Girardi, D. M., Silva, A. C. B., Rego, J. F. M., Coudry, R. A. & Riechelmann, R. P. Unraveling molecular pathways of poorly differentiated neuroendocrine carcinomas of the gastroenteropancreatic system: a systematic review. Cancer Treat. Rev. 56, 28–35 (2017).

    CAS  PubMed  Google Scholar 

  138. Shamir, E. R. et al. Identification of high-risk human papillomavirus and Rb/E2F pathway genomic alterations in mutually exclusive subsets of colorectal neuroendocrine carcinoma. Mod. Pathol. 32, 290–305 (2019).

    CAS  PubMed  Google Scholar 

  139. Scardoni, M. et al. Mixed adenoneuroendocrine carcinomas of the gastrointestinal tract: targeted next-generation sequencing suggests a monoclonal origin of the two components. Neuroendocrinology 100, 310–316 (2014).

    CAS  PubMed  Google Scholar 

  140. Jesinghaus, M. et al. Colorectal mixed adenoneuroendocrine carcinomas and neuroendocrine carcinomas are genetically closely related to colorectal adenocarcinomas. Mod. Pathol. 30, 610–619 (2017).

    CAS  PubMed  Google Scholar 

  141. Woischke, C. et al. In-depth mutational analyses of colorectal neuroendocrine carcinomas with adenoma or adenocarcinoma components. Mod. Pathol. 30, 95–103 (2017).

    CAS  PubMed  Google Scholar 

  142. Rickman, D. S., Beltran, H., Demichelis, F. & Rubin, M. A. Biology and evolution of poorly differentiated neuroendocrine tumors. Nat. Med. 23, 1–10 (2017).

    CAS  PubMed  Google Scholar 

  143. Mafficini, A. & Scarpa, A. Genetics and epigenetics of gastroenteropancreatic neuroendocrine neoplasms. Endocr. Rev. 40, 506–536 (2019).

    PubMed  PubMed Central  Google Scholar 

  144. Tang, L. H. et al. Well-differentiated neuroendocrine tumors with a morphologically apparent high-grade component: a pathway distinct from poorly differentiated neuroendocrine carcinomas. Clin. Cancer Res. 22, 1011–1017 (2016).

    CAS  PubMed  Google Scholar 

  145. Chan, C. S. et al. ATRX, DAXX or MEN1 mutant pancreatic neuroendocrine tumors are a distinct alpha-cell signature subgroup. Nat. Commun. 9, 4158 (2018).

    PubMed  PubMed Central  Google Scholar 

  146. Grillo, F. et al. Grade increases in gastroenteropancreatic neuroendocrine tumor metastases compared to the primary tumor. Neuroendocrinology 103, 452–459 (2016).

    CAS  PubMed  Google Scholar 

  147. Walter, D. et al. Genetic heterogeneity of primary lesion and metastasis in small intestine neuroendocrine tumors. Sci. Rep. 8, 3811 (2018).

    PubMed  PubMed Central  Google Scholar 

  148. Rindi, G. & Solcia, E. Endocrine hyperplasia and dysplasia in the pathogenesis of gastrointestinal and pancreatic endocrine tumors. Gastroenterol. Clin. North. Am. 36, 851–865 (2007).

    PubMed  Google Scholar 

  149. Capuano, F. et al. Grade 3 neuroendocrine tumor (G3 NET) in a background of multiple serotonin cell neoplasms of the ileum associated with carcinoid syndrome and aggressive behavior. Endocr. Pathol. 29, 369–373 (2018).

    CAS  PubMed  Google Scholar 

  150. Amin, M. B. AJCC Cancer Staging Manual 8th edn (Springer, 2017).

  151. Laskaratos, F. M. et al. Neuroendocrine tumors and fibrosis: an unsolved mystery? Cancer 123, 4770–4790 (2017).

    PubMed  Google Scholar 

  152. Rosenbaum, J. N. et al. INSM1: a novel immunohistochemical and molecular marker for neuroendocrine and neuroepithelial neoplasms. Am. J. Clin. Pathol. 144, 579–591 (2015).

    CAS  PubMed  Google Scholar 

  153. Chen, G. et al. Exosomal PD-L1 contributes to immunosuppression and is associated with anti-PD-1 response. Nature 560, 382–386 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  154. Aerts, H. J. et al. Decoding tumour phenotype by noninvasive imaging using a quantitative radiomics approach. Nat. Commun. 5, 4006 (2014).

    CAS  PubMed  Google Scholar 

  155. Gillies, R. J., Kinahan, P. E. & Hricak, H. Radiomics: images are more than pictures, they are data. Radiology 278, 563–577 (2016).

    PubMed  Google Scholar 

  156. Hosny, A., Parmar, C., Quackenbush, J., Schwartz, L. H. & Aerts, H. Artificial intelligence in radiology. Nat. Rev. Cancer 18, 500–510 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  157. Thrall, J. H. et al. Artificial intelligence and machine learning in radiology: opportunities, challenges, pitfalls, and criteria for success. J. Am. Coll. Radiol. 15, 504–508 (2018).

    PubMed  Google Scholar 

  158. Lambin, P. et al. Radiomics: extracting more information from medical images using advanced feature analysis. Eur. J. Cancer 48, 441–446 (2012).

    PubMed  PubMed Central  Google Scholar 

  159. Kumar, V. et al. Radiomics: the process and the challenges. Magn. Reson. Imaging 30, 1234–1248 (2012).

    PubMed  PubMed Central  Google Scholar 

  160. Liu, Z. et al. The applications of radiomics in precision diagnosis and treatment of oncology: opportunities and challenges. Theranostics 9, 1303–1322 (2019).

    PubMed  PubMed Central  Google Scholar 

  161. Braman, N. et al. Association of peritumoral radiomics with tumor biology and pathologic response to preoperative targeted therapy for HER2 (ERBB2)-positive breast cancer. JAMA Netw. Open 2, e192561 (2019).

    PubMed  PubMed Central  Google Scholar 

  162. Kickingereder, P. et al. Automated quantitative tumour response assessment of MRI in neuro-oncology with artificial neural networks: a multicentre, retrospective study. Lancet Oncol. 20, 728–740 (2019).

    PubMed  Google Scholar 

  163. Masood, A. et al. Computer-assisted decision support system in pulmonary cancer detection and stage classification on CT images. J. Biomed. Inf. 79, 117–128 (2018).

    Google Scholar 

  164. Giesel, F. L. et al. Intraindividual Comparison of (18)F-PSMA-1007 and (18)F-DCFPyL PET/CT in the prospective evaluation of patients with newly diagnosed prostate carcinoma: a pilot study. J. Nucl. Med. 59, 1076–1080 (2018).

    CAS  PubMed  Google Scholar 

  165. Bure, L. et al. Can magnetic resonance spectroscopy differentiate malignant and benign causes of lymphadenopathy? An in-vitro approach. PLoS ONE 12, e0182169 (2017).

    PubMed  PubMed Central  Google Scholar 

  166. Dash, A., Knapp, F. F. & Pillai, M. R. Targeted radionuclide therapy–an overview. Curr. Radiopharm. 6, 152–180 (2013).

    CAS  PubMed  Google Scholar 

  167. Kotzerke, J., Runge, R., Braune, A. & Wunderlich, G. Different radionuclides in DOTA-EB-TATE effect different uptake in somatostatin receptor-positive HEK293 cells. J. Nucl. Med. 60, 436 (2019).

    PubMed  PubMed Central  Google Scholar 

  168. Gourni, E. et al. Radiochemistry and preclinical PET imaging of 68Ga-desferrioxamine radiotracers targeting prostate-specific membrane antigen. Mol. Imaging 16, 1536012117737010 (2017).

    PubMed  PubMed Central  Google Scholar 

  169. Nicolas, G. P., Morgenstern, A., Schottelius, M. & Fani, M. New developments in peptide receptor radionuclide therapy. J. Nucl. Med. 60, 167–171 (2019).

    CAS  Google Scholar 

  170. Baum, R. P. et al. Clinical evaluation of the radiolanthanide terbium-152: first-in-human PET/CT with 152Tb-DOTATOC. Dalton Trans. 46, 14638–14646 (2017).

    CAS  PubMed  Google Scholar 

  171. Hicks, R. J. et al. 64Cu-SARTATE PET imaging of patients with neuroendocrine tumors demonstrates high tumor uptake and retention, potentially allowing prospective dosimetry for peptide receptor radionuclide therapy. J. Nucl. Med. 60, 777–785 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  172. Ha, S. Perspectives in radiomics for personalized medicine and theranostics. Nucl. Med. Mol. Imaging 53, 164–166 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  173. Wild, D. et al. Comparison of somatostatin receptor agonist and antagonist for peptide receptor radionuclide therapy: a pilot study. J. Nucl. Med. 55, 1248–1252 (2014).

    CAS  PubMed  Google Scholar 

  174. Nikolas, M. A. & Momany, A. M. DRD4 variants moderate the impact of parental characteristics on child attention-deficit hyperactivity disorder: exploratory evidence from a multiplex family design. J. Abnorm. Child. Psychol. 45, 429–442 (2017).

    PubMed  PubMed Central  Google Scholar 

  175. Dalm, S. U. et al. Comparison of the therapeutic response to treatment with a 177Lu-labeled somatostatin receptor agonist and antagonist in preclinical models. J. Nucl. Med. 57, 260–265 (2016).

    CAS  PubMed  Google Scholar 

  176. Reubi, J. C. & Waser, B. Triple-peptide receptor targeting in vitro allows detection of all tested gut and bronchial NETs. J. Nucl. Med. 56, 613–615 (2015).

    CAS  PubMed  Google Scholar 

  177. Andrews, L. E., Chan, M. H. & Liu, R. S. Nano-lipospheres as acoustically active ultrasound contrast agents: evolving tumor imaging and therapy technique. Nanotechnology 30, 182001 (2019).

    CAS  PubMed  Google Scholar 

  178. Zullino, S., Argenziano, M., Stura, I., Guiot, C. & Cavalli, R. From micro- to nano-multifunctional theranostic platform: effective ultrasound imaging is not just a matter of scale. Mol. Imaging 17, 1536012118778216 (2018).

    PubMed  PubMed Central  Google Scholar 

  179. Li, S. Y. et al. Ultrasonic cavitation ameliorates antitumor efficacy of residual cancer after incomplete radiofrequency ablation in rabbit VX2 liver tumor model. Transl Oncol. 12, 1113–1121 (2019).

    PubMed  PubMed Central  Google Scholar 

  180. Bianchi, F. et al. Localization strategies for robotic endoscopic capsules: a review. Expert Rev. Med. Devices 16, 381–403 (2019).

    CAS  PubMed  Google Scholar 

  181. Ching, H. L. et al. Diagnostic yield of magnetically assisted capsule endoscopy versus gastroscopy in recurrent and refractory iron deficiency anemia. Endoscopy 51, 409–418 (2019).

    PubMed  Google Scholar 

  182. Keuchel, M. Will the steerable capsule replace upper endoscopy in the future? Endoscopy 51, 401–402 (2019).

    PubMed  Google Scholar 

  183. Park, S. B. et al. Advantage of endoscopic mucosal resection with a cap for rectal neuroendocrine tumors. World J. Gastroenterol. 21, 9387–9393 (2015).

    PubMed  PubMed Central  Google Scholar 

  184. Bertani, E. et al. Neuroendocrine neoplasms of rectum: a management update. Cancer Treat. Rev. 66, 45–55 (2018).

    PubMed  Google Scholar 

  185. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT03982264 (2019).

  186. Verbeek, W. H., Korse, C. M. & Tesselaar, M. E. GEP-NETs UPDATE: secreting gastro-enteropancreatic neuroendocrine tumours and biomarkers. Eur. J. Endocrinol. 174, R1–R7 (2016).

    CAS  PubMed  Google Scholar 

  187. Modlin, I. M. et al. Neuroendocrine tumor biomarkers: current status and perspectives. Neuroendocrinology 100, 265–277 (2014).

    CAS  PubMed  Google Scholar 

  188. Modlin, I. M. et al. The NETest: the clinical utility of multigene blood analysis in the diagnosis and management of neuroendocrine tumors. Endocrinol. Metab. Clin. North. Am. 47, 485–504 (2018).

    PubMed  PubMed Central  Google Scholar 

  189. Kidd, M., Drozdov, I. & Modlin, I. Blood and tissue neuroendocrine tumor gene cluster analysis correlate, define hallmarks and predict disease status. Endocr. Relat. Cancer 22, 561–575 (2015).

    CAS  PubMed  Google Scholar 

  190. Modlin, I. M., Kidd, M., Bodei, L., Drozdov, I. & Aslanian, H. The clinical utility of a novel blood-based multi-transcriptome assay for the diagnosis of neuroendocrine tumors of the gastrointestinal tract. Am. J. Gastroenterol. 110, 1223–1232 (2015).

    CAS  PubMed  Google Scholar 

  191. Pavel, M. et al. NET blood transcript analysis defines the crossing of the clinical rubicon: when stable disease becomes progressive. Neuroendocrinology 104, 170–182 (2017).

    CAS  PubMed  Google Scholar 

  192. van Treijen, M. J. C. et al. Blood transcript profiling for the detection of neuroendocrine tumors: results of a large independent validation study. Front. Endocrinol. 9, 740 (2018).

    Google Scholar 

  193. Hofland, J., Zandee, W. T. & de Herder, W. W. Role of biomarker tests for diagnosis of neuroendocrine tumours. Nat. Rev. Endocrinol. 14, 656–669 (2018).

    CAS  PubMed  Google Scholar 

  194. Rindi, G. & Wiedenmann, B. Neuroendocrine neoplasia goes molecular-time for a change. Nat. Rev. Clin. Oncol. 16, 149–150 (2019).

    PubMed  Google Scholar 

  195. Rizzo, F. M. & Meyer, T. Liquid biopsies for neuroendocrine tumors: circulating tumor cells, DNA, and microRNAs. Endocrinol. Metab. Clin. North. Am. 47, 471–483 (2018).

    PubMed  Google Scholar 

  196. Zatelli, M. C. et al. Circulating tumor cells and miRNAs as prognostic markers in neuroendocrine neoplasms. Endocr. Relat. Cancer 24, R223–R237 (2017).

    CAS  PubMed  Google Scholar 

  197. Herrera-Martinez, A. D. et al. Neuroendocrine neoplasms: current and potential diagnostic, predictive and prognostic markers. Endocr. Relat. Cancer 26, R157–R179 (2019).

    CAS  PubMed  Google Scholar 

  198. Carter, L. et al. Molecular analysis of circulating tumor cells identifies distinct copy-number profiles in patients with chemosensitive and chemorefractory small-cell lung cancer. Nat. Med. 23, 114–119 (2017).

    CAS  PubMed  Google Scholar 

  199. Su, Z. et al. Inferring the evolution and progression of small-cell lung cancer by single-cell sequencing of circulating tumor cells. Clin. Cancer Res. 25, 5049–5060 (2019).

    CAS  PubMed  Google Scholar 

  200. Mohamed, A. & Strosberg, J. R. Medical management of gastroenteropancreatic neuroendocrine tumors: current strategies and future advances. J. Nucl. Med. 60, 721–727 (2019).

    CAS  PubMed  Google Scholar 

  201. Hendifar, A. E., Dhall, D. & Strosberg, J. R. The evolving treatment algorithm for advanced neuroendocrine neoplasms: diversity and commonalities across tumor types. Oncologist 24, 54–61 (2019).

    PubMed  Google Scholar 

  202. O’Toole, D., Kianmanesh, R. & Caplin, M. ENETS 2016 consensus guidelines for the management of patients with digestive neuroendocrine tumors: an update. Neuroendocrinology 103, 117–118 (2016).

    PubMed  Google Scholar 

  203. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT03375320 (2020).

  204. Grande, E. 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. 26, 1987–1993 (2015).

    CAS  PubMed  Google Scholar 

  205. Wilky, B. A. A phase II trial of axitinib plus pembrolizumab for patients with advanced alveolar soft part sarcoma (ASPS) and other soft tissue sarcomas (STS). J. Clin. Oncol. 36, 11547–11547 (2018).

    Google Scholar 

  206. Shoji, H. et al. A phase II study of lenvatinib in patients with metastatic colorectal cancer refractory to standard chemotherapy: LEMON study (NCCH1503) [abstract]. J. Clin. Oncol. 37 (Suppl. 15), 3538 (2019).

    Google Scholar 

  207. Sorbye, H., Baudin, E. & Perren, A. The problem of high-grade gastroenteropancreatic neuroendocrine neoplasms: well-differentiated neuroendocrine tumors, neuroendocrine carcinomas, and beyond. Endocrinol. Metab. Clin. North. Am. 47, 683–698 (2018).

    PubMed  Google Scholar 

  208. Sorbye, H. et al. Unmet needs in high-grade gastroenteropancreatic neuroendocrine neoplasms (WHO G3). Neuroendocrinology 108, 54–62 (2019).

    CAS  PubMed  Google Scholar 

  209. Cives, M., Strosberg, J., Al Diffalha, S. & Coppola, D. Analysis of the immune landscape of small bowel neuroendocrine tumors. Endocr. Relat. Cancer 26, 119–130 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  210. Mehnert, J. M. et al. Pembrolizumab for patients with PD-L1–positive advanced carcinoid or pancreatic neuroendocrine tumors: results from the KEYNOTE-028 study [abstract 427O]. Ann. Oncol. 28 (Suppl. 5), v142 (2017).

    Google Scholar 

  211. Schmidt, D. & Wiedenmann, B. Extremely long survival under combined immunotherapy in a metastatic functional neuroendocrine neoplasia patient. Neuroendocrinology 106, 381–388 (2018).

    CAS  PubMed  Google Scholar 

  212. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT03591731 (2019).

  213. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02834013 (2020).

  214. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02923934 (2020).

  215. Yu, D. et al. Preclinical evaluation of AdVince, an oncolytic adenovirus adapted for treatment of liver metastases from neuroendocrine cancer. Neuroendocrinology 105, 54–66 (2017).

    CAS  PubMed  Google Scholar 

  216. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT03879694 (2020).

  217. Begum, N. et al. Neuroendocrine tumours of the GI tract–data from the German NET Registry [German]. Zentralbl Chir. 139, 276–283 (2014).

    PubMed  Google Scholar 

  218. Moertel, C. G. The other CHOP. J. Clin. Oncol. 5, 337–338 (1987).

    CAS  PubMed  Google Scholar 

  219. Chan, M. Y., Ma, K. W. & Chan, A. Surgical management of neuroendocrine tumor-associated liver metastases: a review. Gland. Surg. 7, 28–35 (2018).

    PubMed  PubMed Central  Google Scholar 

  220. Sham, J. G. et al. The impact of extent of liver resection among patients with neuroendocrine liver metastasis: an international multi-institutional study. J. Gastrointest. Surg. 23, 484–491 (2019).

    PubMed  Google Scholar 

  221. Pavel, M. et al. 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 103, 172–185 (2016).

    CAS  PubMed  Google Scholar 

  222. Saxena, A., Chua, T. C., Perera, M., Chu, F. & Morris, D. L. Surgical resection of hepatic metastases from neuroendocrine neoplasms: a systematic review. Surg. Oncol. 21, e131–e141 (2012).

    PubMed  Google Scholar 

  223. Sarmiento, J. M. & Que, F. G. Hepatic surgery for metastases from neuroendocrine tumors. Surg. Oncol. Clin. N. Am. 12, 231–242 (2003).

    PubMed  Google Scholar 

  224. Choi, J. H. et al. Outcomes after endoscopic ultrasound-guided ethanol-lipiodol ablation of small pancreatic neuroendocrine tumors. Dig. Endosc. 30, 652–658 (2018).

    PubMed  Google Scholar 

  225. Hibi, T. et al. Surgery for hepatic neuroendocrine tumors: a single institutional experience in Japan. Jpn. J. Clin. Oncol. 37, 102–107 (2007).

    PubMed  Google Scholar 

  226. de Mestier, L., Zappa, M., Hentic, O., Vilgrain, V. & Ruszniewski, P. Liver transarterial embolizations in metastatic neuroendocrine tumors. Rev. Endocr. Metab. Disord. 18, 459–471 (2017).

    PubMed  Google Scholar 

  227. Steinmuller, T. et al. Consensus guidelines for the management of patients with liver metastases from digestive (neuro)endocrine tumors: foregut, midgut, hindgut, and unknown primary. Neuroendocrinology 87, 47–62 (2008).

    PubMed  Google Scholar 

  228. Kennedy, A. et al. Role of hepatic intra-arterial therapies in metastatic neuroendocrine tumours (NET): guidelines from the NET-Liver-Metastases Consensus Conference. HPB 17, 29–37 (2015).

    PubMed  Google Scholar 

  229. Davar, J. et al. Diagnosing and managing carcinoid heart disease in patients with neuroendocrine tumors: an expert statement. J. Am. Coll. Cardiol. 69, 1288–1304 (2017).

    PubMed  Google Scholar 

  230. Bhattacharyya, S. et al. Circulating plasma and platelet 5-hydroxytryptamine in carcinoid heart disease: a pilot study. J. Heart Valve Dis. 22, 400–407 (2013).

    PubMed  Google Scholar 

  231. Buchanan-Hughes, A. et al. Carcinoid heart disease: prognostic value of 5-hydroxyindoleacetic acid levels and impact on survival – a systematic literature review. Neuroendocrinology https://doi.org/10.1159/000506744 (2020).

  232. Timmermans, P. Jr. et al. Exercise cardiac magnetic resonance imaging with pulmonary artery catheter monitoring in carcinoid heart disease: a shift towards early intervention? ESC Heart Fail. 5, 953–955 (2018).

    PubMed  PubMed Central  Google Scholar 

  233. Laule, M. et al. Staged catheter-based valve treatment of severe carcinoid heart disease. Neuroendocrinology 103, 259–262 (2016).

    CAS  PubMed  Google Scholar 

  234. Kong, G. & Hicks, R. J. Peptide receptor radiotherapy: current approaches and future directions. Curr. Treat. Options Oncol. 20, 77 (2019).

    PubMed  Google Scholar 

  235. Dorffel, Y., Swidsinski, A., Loening-Baucke, V., Wiedenmann, B. & Pavel, M. Common biostructure of the colonic microbiota in neuroendocrine tumors and Crohn’s disease and the effect of therapy. Inflamm. Bowel Dis. 18, 1663–1671 (2012).

    PubMed  Google Scholar 

  236. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT03211988 (2019).

  237. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT03411915 (2020).

  238. White, B. H. et al. Discovery of an SSTR2-targeting maytansinoid conjugate (PEN-221) with potent activity in vitro and in vivo. J. Med. Chem. 62, 2708–2719 (2019).

    CAS  PubMed  Google Scholar 

  239. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02936323 (2020).

  240. Ciardiello, F. et al. Delivering precision medicine in oncology today and in future–the promise and challenges of personalised cancer medicine: a position paper by the European Society for Medical Oncology (ESMO). Ann. Oncol. 25, 1673–1678 (2014).

    CAS  PubMed  Google Scholar 

  241. Klimstra, D. S., Kloppel, G., La Rosa, S. & Rindi, G. in WHO Classification of Tumours: Digestive System Tumours (ed Cree, I. A.) 7–10 (IARC, 2019).

  242. Rindi, G. & Inzani, F. Neuroendocrine neoplasm update: toward universal nomenclature. Endocr. Relat. Cancer 27, R211–R218 (2020).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors acknowledge the support of internal university grant (Università Cattolica line D.1 2017-R412500403) to G.R. and the WILL Foundation and generous donation in memoriam “Sören Piepgras” to B.W. We are grateful to C. Klersy, Clinical Epidemiology and Biometry, IRCCS Fondazione Policlinico San Matteo, Pavia, Italy, for generous and skilled statistical work on published databases and to M. Sigal, Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany, T. Penzkofer, Department of Radiology, Charité - Universitätsmedizin Berlin, Germany, L. Hammerich, Department of Medicine, Charité - Universitätsmedizin Berlin, Germany and R. Baum, THERANOSTICS Centre for Molecular Radiotherapy and Molecular Imaging, Zentralklinik Bad Berka, Germany for critical revision and helpful suggestions.

Review criteria

A search for original articles published between 1980 and 2019 focusing on neuroendocrine neoplasms was performed in MEDLINE and PubMed. The search terms used were “carcinoids”, “endocrine tumours”, “neuroendocrine tumours”, “neuroendocrine carcinoma” AND “gastrointestinal tract”, as well as AND “epidemiology”, “genetics”, “molecular biology”, “imaging”, “medical therapy”, “surgery” and “PRRT”. Also, a specific search was done on “gastrointestinal tract” AND “neuroendocrine cells” AND “SNARE proteins”, “secretion”, “signal transduction”, “development “, “organoid”, “single cell RNA”, “transcription factors” and “lineage specification”. All papers identified were English-language, full-text papers. We also searched the reference lists of identified articles for further papers.

Author information

Authors and Affiliations

Authors

Contributions

The authors contributed equally to all aspects of the article.

Corresponding author

Correspondence to Guido Rindi.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information

Nature Reviews Endocrinology thanks the anonymous reviewer(s) for their contribution to the peer review of this work.

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Rindi, G., Wiedenmann, B. Neuroendocrine neoplasia of the gastrointestinal tract revisited: towards precision medicine. Nat Rev Endocrinol 16, 590–607 (2020). https://doi.org/10.1038/s41574-020-0391-3

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41574-020-0391-3

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