Выход
Вход/Login
 
E-mail
Пароль/Password
Забыли пароль?
Введите E-mail и жмите тут. Пароль будет выслан на указанный адрес
Войти (LogIn)

 

Если вы первый раз здесь, то зарегистрируйтесь

Регистрация/Sign Up
Полное имя (Ф И О)/Full name
E-mail
Телефон/Phone
Зарегистрироваться,
на ваш E-mail будет выслан временный пароль

Нажимая кнопку Зарегистрироваться, вы соглашаетесь с Правилами сайта и Политикой Конфиденциальности http://vidar.ru/rules.asp

 

Медицинская литература. Новинки


 

 

 

 

 

 
вce журналы << Эндокринная хирургия << 2017 год << №4 <<
стр.178
отметить
статью

Радионуклидная визуализация и терапия у пациентов с нейроэндокринными опухолями

Баранова О. Д., Румянцев П. О., Слащук К. Ю., Петров Л. О.
Вы можете загрузить полный текст статьи в формате pdf
Баранова Ольга Дмитриевна - клинический ординатор, МНИОИ им. П.А. Герцена - филиал ФГБУ “Национальный медицинский исследовательский центр радиологии” Минздрава России, olga.dm1091@gmail.com, 125284, г. Москва, 2-й Боткинский пр-д, 3
Румянцев Павел Олегович - д.м.н., заведующий отделом радионуклидной диагностики и терапии, ФГБУ “Национальный медицинский исследовательский центр эндокринологии” Минздрава России, pavelrum@gmail.com, 117036, г. Москва, ул. Дм.Ульянова, д.11
Слащук Константин Юрьевич - научный сотрудник отделения радионуклидной диагностики, ФГБУ “Национальный медицинский исследовательский центр эндокринологии” Минздрава России, slashuk911@gmail.com, 117036, г. Москва, ул. Дм.Ульянова, д.11
Петров Леонид Олегович - к.м.н., старший научный сотрудник абдоминального отделения, МНИОИ им. П.А. Герцена - филиал ФГБУ “Национальный медицинский исследовательский центр радиологии” Минздрава России, leonid_petrov@mail.ru, 125284, г. Москва, 2-й Боткинский пр-д, 3

Нейроэндокринные опухоли представляют собой гетерогенную группу опухолей, возникающих из энтерохромаффинных клеток диффузной нейроэндокринной системы и встречающихся в 0,5% случаев всех новообразований. За последние годы отмечается значительный рост заболеваемости нейроэндокринными опухолями, что безусловно связано с усовершенствованием методов их диагностики. Однако, несмотря на значительные успехи в изучении биологических и молекулярных механизмов поведения данной группы заболеваний, формирование единого алгоритма диагностики и лечения нейроэндокринных опухолей остается затруднительным и по сей день. Лечение нейроэндокринных опухолей во многом зависит от их функционального статуса и стадии заболевания. Наиболее предпочтительным подходом в лечении локализованных форм нейроэндокринных опухолей остается хирургический. В то же время для больных с распространенными процессами, характеризующимися наличием отдаленных метастазов, на сегодняшний день доступен широкий спектр терапевтических стратегий. Среди них можно выделить контроль над синдромами, ассоциированными с гиперсекрецией тех или иных гормонов, выполнение циторедуктивных вмешательств, системную химиотерапию, применение аналогов соматостатина, а также радиотаргетную терапию. Однако алгоритм выбора того или иного терапевтического подхода в современной клинической практике у разнородной группы больных с нейроэндокринными опухолями требует дальнейшего обсуждения.

Ключевые слова:
нейроэндокринные опухоли, радионуклидная диагностика, радиотаргетная терапия, аналоги соматостатина, октреотид, онкология, радиология, neuroendocrine tumors, radionuclide imaging, radiolabeled therapy, somatostatin analogues, octreotide, oncology, radiology

Литература:
1.1. Taal BG, Visser O. Epidemiology of neuroendocrine tumours. Neuroendocrinology. 2004;80 Suppl 1:3-7. doi: 10.1159/000080731.
2.2. Modlin IM, Oberg K, Chung DC, et al. Gastroenteropancreatic neuroendocrine tumours. Lancet Oncol. 2008;9(1):61-72. doi: 10.1016/s1470-2045(07)70410-2.
3.3. Kloppel G, Perren A, Heitz PU. The gastroenteropancreatic neuroendocrine cell system and its tumors: the WHO classification. Ann N Y Acad Sci. 2004;1014(1):13-27. doi: 10.1196/annals.1294.002.
4.4. Yao JC, Hassan M, Phan A, et al. One hundred years after “carcinoid”: epidemiology of and prognostic factors for neuroendocrine tumors in 35,825 cases in the United States. J Clin Oncol. 2008;26(18):3063-3072. doi: 10.1200/JCO.2007.15.4377.
5.5. Solcia E, Kloppel G, Sobin LH. Histological Typing of Endocrine Tumors. Berlin Heidelberg: Springer; 2000. doi: 10.1007/978-3-642-59655-1.
6.6. Ito T, Sasano H, Tanaka M, et al. Epidemiological study of gastroenteropancreatic neuroendocrine tumors in Japan. J Gastroenterol. 2010;45(2):234-243. doi: 10.1007/s00535-009-0194-8.
7.7. Garcia-Carbonero R, Capdevila J, Crespo-Herrero G, et al. Incidence, patterns of care and prognostic factors for outcome of gastroenteropancreatic neuroendocrine tumors (GEP-NETs): results from the National Cancer Registry of Spain (RGETNE). Ann Oncol. 2010;21(9):1794-1803. doi: 10.1093/annonc/mdq022.
8.8. Lepage C, Bouvier AM, Manfredi S, et al. Incidence and management of primary malignant small bowel cancers: a well-defined French population study. Am J Gastroenterol. 2006;101(12):2826-2832. doi: 10.1111/j.1572-0241.2006.00854.x.
9.9. Ploeckinger U, Kloeppel G, Wiedenmann B, et al. The German NET-registry: an audit on the diagnosis and therapy of neuroendocrine tumors. Neuroendocrinology. 2009;90(4):349-363. doi: 10.1159/000242109.
10.10. Bergestuen DS, Aabakken L, Holm K, et al. Small intestinal neuroendocrine tumors: prognostic factors and survival. Scand J Gastroenterol. 2009;44(9):1084-1091. doi: 10.1080/00365520903082432.
11.11. Williams E. The classification of carcinoid tumours. Lancet. 1963;281(7275):238-239. doi: 10.1016/s0140-6736(63)90951-6.
12.12. Rindi G, Arnold R, Bosman FT, et al. Nomenclature and classification of neuroendocrine neoplasms of the digestive system. In: Bosman FT, Carneiro F, Hruban RH, et al., editors. WHO classification of tumors of the digestive system. Lyon: IARC; 2010. p. S13-S14.
13.13. Van Eeden S, Quaedvlieg PF, Taal BG, et al. Classification of low-grade neuroendocrine tumors of midgut and unknown origin. Hum Pathol. 2002;33(11):1126-1132. doi: 10.1053/hupa.2002.129204.
14.14. Kulke MH. Are neuroendocrine tumors going mainstream? J Clin Oncol. 2013;31(4):404-405. doi: 10.1200/jco.2012.47.3884.
15.15. Sorbye H, Welin S, Langer SW, et al. Predictive and prognostic factors for treatment and survival in 305 patients with advanced gastrointestinal neuroendocrine carcinoma (WHO G3): the NORDIC NEC study. Ann Oncol. 2013;24(1):152-160. doi: 10.1093/annonc/mds276.
16.16. Velayoudom-Cephise FL, Duvillard P, Foucan L, et al. Are G3 ENETS neuroendocrine neoplasms heterogeneous? Endocr Relat Cancer. 2013;20(5):649-657. doi: 10.1530/ERC-13-0027.
17.17. Heetfeld M, Chougnet CN, Olsen IH, et al. Characteristics and treatment of patients with G3 gastroenteropancreatic neuroendocrine neoplasms. Endocr Relat Cancer. 2015; 22(4):657-664. doi: 10.1530/ERC-15-0119.
18.18. Basturk O, Yang Z, Tang LH, et al. The high-grade (WHO G3) pancreatic neuroendocrine tumor category is morphologically and biologically heterogenous and includes both well differentiated and poorly differentiated neoplasms. Am J Surg Pathol. 2015;39(5):683-690. doi: 10.1097/PAS.0000000000000408.
19.19. 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):1192-1202. doi: 10.1097/PAS.0000000000000662.
20.20. Rindi G, Luinetti O, Cornaggia M, et al. Three subtypes of gastric argyrophil carcinoid and the gastric neuroendocrine carcinoma: A clinicopathologic study. Gastroenterology. 1993; 104(4):994-1006. doi: 10.1016/0016-5085(93)90266-f.
21.21. Landerholm K, Falkmer S, Jarhult J. Epidemiology of small bowel carcinoids in a defined population. World J Surg. 2010;34(7):1500-1505. doi: 10.1007/s00268-010-0519-z.
22.22. Sitaraman SV, Goldfinger SE. Clinical characteristics of primary carcinoid tumors. In: Tannabe KK, Whitcomb DC, editors. UpToDate. Waltham: UpToDate; 2008.
23.23. Wolf P, Winhofer Y, Smajis S, et al. Clinical presentation in insulinoma predicts histopathological tumour characteristics. Clin Endocrinol (Oxf). 2015;83(1):67-71. doi: 10.1111/cen.12777.
24.24. Yang RH, Chu YK. Zollinger-Ellison syndrome: Revelation of the gastrinoma triangle. Radiol Case Rep. 2015;10(1):827. doi: 10.2484/rcr.v10i1.827.
25.25. Nikou GC, Toubanakis C, Nikolaou P, et al. Gastrinomas associated with MEN-1 syndrome: new insights for the diagnosis and management in a series of 11 patients. Hepatogastroenterology. 2005;52(66):1668-1676.
26.26. Mortenson M, Bold RJ. Symptomatic pancreatic polypeptide-secreting tumor of the distal pancreas (PPoma). Int J Gastrointest Cancer. 2002;32(2-3):153-156. doi: 10.1385/ijgc:32:2-3:153.
27.27. Verner JV, Morrison AB. Islet cell tumor and a syndrome of refractory watery diarrhea and hypokalemia. Am J Med. 1958;25(3):374-380. doi: 10.1016/0002-9343(58)90075-5.
28.28. Castro PG, de Leon AM, Trancon JG, et al. Glucagonoma syndrome: a case report. J Med Case Rep. 2011;5:402. doi: 10.1186/1752-1947-5-402.
29.29. Soga J, Yakuwa Y. Somatostatinoma/inhibitory syndrome: a statistical evaluation of 173 reported cases as compared to other pancreatic endocrinomas. J Exp Clin Cancer Res. 1999;18(1):13-22.
30.30. Hajdu SI, Tang P. A note from history: the saga of carcinoid and oat-cell carcinoma. Ann Clin Lab Sci. 2008;38(4): 414-417.
31.31. Masson P. Carcinoids (argentaffin-cell tumors) and nerve hyperplasia of the appendicular mucosa. Am J Pathol. 1928;4(3):181-212.
32.32. Vinik AI, Silva MP, Woltering EA, et al. Biochemical testing for neuroendocrine tumors. Pancreas. 2009;38(8):876-889. doi: 10.1097/MPA.0b013e3181bc0e77.
33.33. Modlin IM, Champaneria MC, Bornschein J, Kidd M. Evolution of the diffuse neuroendocrine system - clear cells and cloudy origins. Neuroendocrinology. 2006;84(2):69-82. doi: 10.1159/000096997.
34.34. Oberg K. Circulating biomarkers in gastroenteropancreatic neuroendocrine tumours. Endocr Relat Cancer. 2011;18 Suppl 1:S17-25. doi: 10.1530/ERC-10-0280.
35.35. Kloppel G, Couvelard A, Perren A, et al. ENETS Consensus Guidelines for the Standards of Care in Neuroendocrine Tumors: towards a standardized approach to the diagnosis of gastroenteropancreatic neuroendocrine tumors and their prognostic stratification. Neuroendocrinology.2009;90(2): 162-166. doi: 10.1159/000182196.
36.36. Oberg K, Modlin IM, De Herder W, et al. Consensus on biomarkers for neuroendocrine tumour disease. Lancet Oncol. 2015;16(9):e435-e446. doi: 10.1016/s1470-2045(15)00186-2.
37.37. Modlin IM, Gustafsson BI, Moss SF, et al. Chromogranin A - biological function and clinical utility in neuroendocrine tumor disease. Ann Surg Oncol. 2010;17(9):2427-2443. doi: 10.1245/s10434-010-1006-3.
38.38. Giusti M, Sidoti M, Augeri C, et al. Effect of short-term treatment with low dosages of the proton-pump inhibitor omeprazole on serum chromogranin A levels in man. Eur J Endocrinol. 2004;150(3):299-303. doi: 10.1530/eje.0.1500299.
39.39. Yang X, Yang Y, Li Z, et al. Diagnostic value of circulating chromogranin a for neuroendocrine tumors: a systematic review and meta-analysis. PLoS One. 2015;10(4):e0124884. doi: 10.1371/journal.pone.0124884.
40.40. Zatelli MC, Torta M, Leon A, et al. Chromogranin A as a marker of neuroendocrine neoplasia: an Italian Multicenter Study. Endocr Relat Cancer. 2007;14(2):473-482. doi: 10.1677/ERC-07-0001.
41.41. Janson ET, Holmberg L, Stridsberg M, et al. Carcinoid tumors: Analysis of prognostic factors and survival in 301 patients from a referral center. Ann Oncol. 1997;8(7):685-690. doi: 10.1023/a:1008215730767.
42.42. Boudreaux JP, Klimstra DS, Hassan MM, et al. The NANETS Consensus Guideline for the Diagnosis and Management of Neuroendocrine Tumors. Pancreas. 2010;39(6):753-766. doi: 10.1097/MPA.0b013e3181ebb2a5.
43.43. Frilling A, Modlin IM, Kidd M, et al. Recommendations for management of patients with neuroendocrine liver metastases. Lancet Oncol. 2014;15(1):e8-e21. doi: 10.1016/s1470-2045(13)70362-0.
44.44. Berna MJ, Hoffmann KM, Serrano J, et al. Serum gastrin in Zollinger-Ellison syndrome: I. Prospective study of fasting serum gastrin in 309 patients from the National Institutes of Health and comparison with 2229 cases from the literature. Medicine (Baltimore). 2006;85(6):295-330. doi: 10.1097/01.md.0000236956.74128.76.
45.45. Massironi S, Conte D, Sciola V, et al. Plasma chromogranin A response to octreotide test: prognostic value for clinical outcome in endocrine digestive tumors. Am J Gastroenterol. 2010;105(9):2072-2078. doi: 10.1038/ajg.2010.154.
46.46. Massironi S, Rossi RE, Casazza G, et al. Chromogranin A in diagnosing and monitoring patients with gastroenteropancreatic neuroendocrine neoplasms: a large series from a single institution. Neuroendocrinology. 2014;100(2-3):240-249. doi: 10.1159/000369818.
47.47. Rossi RE, Garcia-Hernandez J, Meyer T, et al. Chromogranin A as a predictor of radiological disease progression in neuroendocrine tumours. Ann Transl Med. 2015;3(9):118. doi: 10.3978/j.issn.2305-5839.2015.04.23.
48.48. Kunz PL, Reidy-Lagunes D, Anthony LB, et al. Consensus guidelines for the management and treatment of neuroendocrine tumors. Pancreas. 2013;42(4):557-577. doi: 10.1097/MPA.0b013e31828e34a4.
49.49. Ardill J. The importance of the measurement of circulating markers in patients with neuroendocrine tumours of the pancreas and gut. Endocr Relat Cancer. 2003;10(4):459-462. doi: 10.1677/erc.0.0100459.
50.50. Feldman JM, O'Dorisio TM. Role of neuropeptides and serotonin in the diagnosis of carcinoid tumors. Am J Med. 1986;81(6):41-48. doi: 10.1016/0002-9343(86)90583-8.
51.51. O'Toole D, Grossman A, Gross D, et al. ENETS Consensus Guidelines for the Standards of Care in Neuroendocrine Tumors: biochemical markers. Neuroendocrinology. 2009; 90(2):194-202. doi: 10.1159/000225948.
52.52. Kaltsas G, Rockall A, Papadogias D, et al. Recent advances in radiological and radionuclide imaging and therapy of neuroendocrine tumours. Eur J Endocrinol. 2004;151(1):15-27. doi: 10.1530/eje.0.1510015.
53.53. Ricke J, Klose KJ, Mignon M, et al. Standardisation of imaging in neuroendocrine tumours: results of a European delphi process. Eur J Radiol. 2001;37(1):8-17. doi: 10.1016/s0720-048x(00)00187-x.
54.54. Binderup T, Knigge U, Mellon Mogensen A, et al. Quantitative gene expression of somatostatin receptors and noradrenaline transporter underlying scintigraphic results in patients with neuroendocrine tumors. Neuroendocrinology. 2008;87(4): 223-232. doi: 10.1159/000113128.
55.55. Reubi J, Waser B, Schaer J-C, Laissue JA. Somatostatin receptor sst1-sst5 expression in normal and neoplastic human tissues using receptor autoradiography with subtype-selective ligands. Eur J Nucl Med. 2001;28(7):836-846. doi: 10.1007/s002590100541.
56.56. Oberg K, Jelic S, Group EGW. Neuroendocrine gastroenteropancreatic tumors: ESMO clinical recommendations for diagnosis, treatment and follow-up. Ann Oncol. 2008;19 Suppl 2:ii104-105. doi: 10.1093/annonc/mdn117.
57.57. Raderer M, Kurtaran A, Leimer M, et al. Value of peptide receptor scintigraphy using (123)I-vasoactive intestinal peptide and (111)In-DTPA-D-Phe1-octreotide in 194 carcinoid patients: Vienna University Experience, 1993 to 1998. J Clin Oncol. 2000;18(6):1331-1336. doi: 10.1200/JCO.2000.18.6.1331.
58.58. Lu SJ, Gnanasegaran G, Buscombe J, Navalkissoor S. Single photon emission computed tomography/computed tomography in the evaluation of neuroendocrine tumours: a review of the literature. Nucl Med Commun. 2013;34(2):98-107. doi: 10.1097/MNM.0b013e32835bd59d.
59.59. Gabriel M, Muehllechner P, Decristoforo C, et al. 99mTc-EDDA/HYNIC-Tyr(3)-octreotide for staging and follow-up of patients with neuroendocrine gastro-entero-pancreatic tumors. Q J Nucl Med Mol Imaging. 2005;49(3):237-244.
60.60. Hubalewska-Dydejczyk A, Fross-Baron K, Mikolajczak R, et al. 99mTc-EDDA/HYNIC-octreotate scintigraphy, an efficient method for the detection and staging of carcinoid tumours: results of 3 years' experience. Eur J Nucl Med Mol Imaging. 2006;33(10):1123-1133. doi: 10.1007/s00259-006-0113-7.
61.61. Gabriel M, Decristoforo C, Donnemiller E, et al. An intrapatient comparison of 99mTc-EDDA/HYNIC-TOC with 111In-DTPA-octreotide for diagnosis of somatostatin receptor-expressing tumors. J Nucl Med. 2003;44(5):708-716.
62.62. Reubi JC, Schar J-C, Waser B, et al. Affinity profiles for human somatostatin receptor subtypes SST1-SST5 of somatostatin radiotracers selected for scintigraphic and radiotherapeutic use. Eur J Nucl Med Mol Imaging. 2000;27(3): 273-282. doi: 10.1007/s002590050034.
63.63. Antunes P, Ginj M, Zhang H, et al. Are radiogallium-labelled DOTA-conjugated somatostatin analogues superior to those labelled with other radiometals? Eur J Nucl Med Mol Imaging. 2007;34(7):982-993. doi: 10.1007/s00259-006-0317-x.
64.64. Besig S, Voland P, Baur DM, et al. Vascular endothelial growth factors, angiogenesis, and survival in human ileal enterochromaffin cell carcinoids. Neuroendocrinology. 2009;90(4): 402-415. doi: 10.1159/000245900.
65.65. Voland P, Besig S, Rad R, et al. Correlation of matrix metalloproteinases and tissue inhibitors of matrix metalloproteinase expression in ileal carcinoids, lymph nodes and liver metastasis with prognosis and survival. Neuroendocrinology. 2009;89(1):66-78. doi: 10.1159/000151482.
66.66. Hassan MM, Phan A, Li D, et al. Family history of cancer and associated risk of developing neuroendocrine tumors: a case-control study. Cancer Epidemiol Biomarkers Prev. 2008;17(4):959-965. doi: 10.1158/1055-9965.EPI-07-0750.
67.67. Hemminki K, Li X. Familial carcinoid tumors and subsequent cancers: a nation-wide epidemiologic study from Sweden. Int J Cancer. 2001;94(3):444-448. doi: 10.1002/ijc.1473.
68.68. Jarhult J, Landerholm K, Falkmer S, et al. First report on metastasizing small bowel carcinoids in first-degree relatives in three generations. Neuroendocrinology. 2010; 91(4):318-323. doi: 10.1159/000299790.
69.69. Tonnies H, Toliat MR, Ramel C, et al. Analysis of sporadic neuroendocrine tumours of the enteropancreatic system by comparative genomic hybridisation. Gut. 2001;48(4):536-541. doi: 10.1136/gut.48.4.536.
70.70. Jann H, Roll S, Couvelard A, et al. Neuroendocrine tumors of midgut and hindgut origin: tumor-node-metastasis classification determines clinical outcome. Cancer. 2011;117(15): 3332-3341. doi: 10.1002/cncr.25855.
71.71. Schindl M, Kaczirek K, Passler C, et al. Treatment of small intestinal neuroendocrine tumors: is an extended multimodal approach justified? World J Surg. 2002;26(8):976-984. doi: 10.1007/s00268-002-6628-6.
72.72. Rindi G, Kloppel G, Couvelard A, et al. TNM staging of midgut and hindgut (neuro) endocrine tumors: a consensus proposal including a grading system. Virchows Arch. 2007;451(4):757-762. doi: 10.1007/s00428-007-0452-1.
73.73. Metz DC, Jensen RT. Gastrointestinal neuroendocrine tumors: pancreatic endocrine tumors. Gastroenterology. 2008;135(5):1469-1492. doi: 10.1053/j.gastro.2008.05.047.
74.74. Saxena A, Chua TC, Chu F, et al. Optimizing the surgical effort in patients with advanced neuroendocrine neoplasm hepatic metastases: a critical analysis of 40 patients treated by hepatic resection and cryoablation. Am J Clin Oncol. 2012; 35(5):439-445. doi: 10.1097/COC.0b013e31821bc8dd.
75.75. Birnbaum DJ, Turrini O, Vigano L, et al. Surgical management of advanced pancreatic neuroendocrine tumors: short-term and long-term results from an international multi-institutional study. Ann Surg Oncol. 2015;22(3):1000-1007. doi: 10.1245/s10434-014-4016-8.
76.76. Yuan CH, Wang J, Xiu DR, et al. Meta-analysis of liver resection versus nonsurgical treatments for pancreatic neuroendocrine tumors with liver metastases. Ann Surg Oncol. 2016;23(1):244-249. doi: 10.1245/s10434-015-4654-5.
77.77. Moller LN, Stidsen CE, Hartmann B, Holst JJ. Somatostatin receptors. Biochim Biophys Acta. 2003;1616(1):1-84. doi: 10.1016/s0005-2736(03)00235-9.
78.78. Rinke A, Muller HH, Schade-Brittinger C, et al. Placebo-controlled, double-blind, prospective, randomized study on the effect of octreotide LAR in the control of tumor growth in patients with metastatic neuroendocrine midgut tumors: a report from the PROMID Study Group. J ClinOncol. 2009;27(28):4656-4663. doi: 10.1200/JCO.2009.22.8510.
79.79. Laplante M, Sabatini DM. mTOR signaling in growth control and disease. Cell. 2012;149(2):274-293. doi: 10.1016/j.cell.2012.03.017.
80.80. Oberg KE. Gastrointestinal neuroendocrine tumors. Ann Oncol. 2010;21 Suppl 7:vii72-80. doi: 10.1093/annonc/mdq290.
81.81. Yao JC, Shah MH, Ito T, et al. Everolimus for advanced pancreatic neuroendocrine tumors. N Engl J Med. 2011; 364(6):514-523. doi: 10.1056/NEJMoa1009290.
82.82. Yao JC, Fazio N, Singh S, et al. Everolimus for the treatment of advanced, non-functional neuroendocrine tumours of the lung or gastrointestinal tract (RADIANT-4): a randomised, placebo-controlled, phase 3 study. Lancet. 2016;387(10022): 968-977. doi: 10.1016/s0140-6736(15)00817-x.
83.83. Raymond E, Dahan L, Raoul JL, et al. Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. N Engl J Med. 2011;364(6):501-513. doi: 10.1056/NEJMoa1003825.
84.84. Phan AT, Halperin DM, Chan JA, et al. Pazopanib and depot octreotide in advanced, well-differentiated neuroendocrine tumours: a multicentre, single-group, phase 2 study. Lancet Oncol. 2015;16(6):695-703. doi: 10.1016/s1470-2045(15)70136-1.
85.85. Yao JC, Guthrie KA, Moran C, et al. Phase III prospective randomized comparison trial of depot octreotide plus interferon alfa-2b versus depot octreotide plus bevacizumab in patients with advanced carcinoid tumors: SWOG S0518. J Clin Oncol. 2017;35(15):1695-1703. doi: 10.1200/JCO.2016.70.4072.
86.86. Strosberg J, El-Haddad G, Wolin E, et al. Phase 3 trial of (177)Lu-Dotatate for midgut neuroendocrine tumors. N Engl J Med. 2017;376(2):125-135. doi: 10.1056/NEJMoa1607427.
87.87. Garcia-Carbonero R, Sorbye H, Baudin E, et al. ENETS Consensus Guidelines for High-Grade Gastroenteropancreatic Neuroendocrine Tumors and Neuroendocrine Carcinomas. Neuroendocrinology. 2016;103(2):186-194. doi: 10.1159/000443172.
88.88. Moertel CG, Kvols LK, O'Connell MJ, Rubin J. Treatment of neuroendocrine carcinomas with combined etoposide and cisplatin. Evidence of major therapeutic activity in the anaplastic variants of these neoplasms. Cancer. 1991;68(2): 227-232. doi: 10.1002/1097-0142(19910715)68:2227::aid-cncr2820680202>3.0.co;2-i.
89.89. Hentic O, Hammel P, Couvelard A, et al. FOLFIRI regimen: an effective second-line chemotherapy after failure of etoposide-platinum combination in patients with neuroendocrine carcinomas grade 3. Endocr Relat Cancer. 2012;19(6): 751-757. doi: 10.1530/ERC-12-0002.
90.90. Hadoux J, Malka D, Planchard D, et al. Post-first-line FOLFOX chemotherapy for grade 3 neuroendocrine carcinoma. Endocr Relat Cancer. 2015;22(3):289-298. doi: 10.1530/ERC-15-0075.
91.91. Pavel M, O'Toole D, Costa F, 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. 2016;103(2):172-185. doi: 10.1159/000443167.
92.92. Ezziddin S, Opitz M, Attassi M, et a

Radionuclide imaging and therapy in patients with neuroendocrine tumors

Baranova O. D., Roumiantsev P. O., Slashchuk K. Y., Petrov L. O.

Neuroendocrine tumors (NETs) are heterogeneous group of the tumors that arise from the enterochromaffin cells of the diffuse neuroendocrine system and occurr in 0.5% of all neoplasms. Recently there has been a significant increase in the incidence of neuroendocrine tumors, which is undoubtedly associated with the improvement of diagnostic methods. However, despite significant success in studying the biological and molecular mechanisms of its behavior, a single algorithm for the diagnosis and treatment of neuroendocrine tumors remains unclear today. Treatment of neuroendocrine tumors largely depends on their functional status and the stage of the disease. While the treatment of localized NETs is surgical resection, varieties of therapeutic options are available for patients with advanced NETs. These include medical control of excess hormone levels and associated symptoms, cytoreductive surgery for patients with advanced disease, systemic chemotherapy, somatostatin analogues, and peptide receptor-targeted radionuclide therapy. However, the right choice of the therapeutic approach in current clinical practice in heterogeneous group of patients with neuroendocrine tumors requires further discussion.

Keywords:
нейроэндокринные опухоли, радионуклидная диагностика, радиотаргетная терапия, аналоги соматостатина, октреотид, онкология, радиология, neuroendocrine tumors, radionuclide imaging, radiolabeled therapy, somatostatin analogues, octreotide, oncology, radiology

Новости   Магазин   Журналы   Контакты   Правила   Доставка   О компании  
ООО Издательский дом ВИДАР-М, 2024