Применение таргетной терапии в лечении пациентов с нейроэндокринными опухолями желудочно- кишечного тракта и поджелудочной железы

Автор: Исянгулова А. З., Хасанов Р. Ш., Еникеев Р. Ф.

Журнал: Злокачественные опухоли @malignanttumors

Рубрика: Обзоры и аналитика

Статья в выпуске: 4 т.9, 2019 года.

Бесплатный доступ

Проблема нейроэндокринных опухолей (НЭО) стала углубленно изучаться только в последние годы. Возросшее внимание к ней обусловлено увеличением частоты обнаружения НЭО в связи с совершенствованием методов диагностики. Ограниченная эффективность химиотерапии у высокодифференцированных распространенных метастатических НЭО желудочно-кишечного тракта (ЖКТ) и поджелудочной железы (ПЖ) подчеркивает необходимость поиска новых и более эффективных вариантов лекарственного лечения. Недавние исследования специфических биологических особенностей НЭО привели к разработке новых целевых методов лечения, которые учитывают высокую васкуляризацию и гиперэкспрессию специфических факторов роста и родственных рецепторов тирозинкиназы. Таким образом, с изучением сигнальных путей mTOR TSC2, PTEN и PIK3CA открываются новые возможности в лечении пациентов с НЭО ЖКТ и ПЖ, особенно неоперабельных и метастатических форм. Таргетная терапия, которая специфически ингибирует рецепторы факторов роста и связанные с ними сигнальные пути, является многообещающим подходом лекарственного лечения НЭО ЖКТ и ПЖ. В этом обзоре кратко изложено состояние и перспективы использования таргетной терапии, описаны клинические исследования в лечении НЭО ЖКТ и ПЖ.

Еще

Нейроэндокринная опухоль, таргетная терапия, карциноид, рак поджелудочной железы, рак желудочно-кишечного тракта

Короткий адрес: https://sciup.org/140249284

IDR: 140249284   |   DOI: 10.18027/2224-5057-2019-9-4-49-58

Список литературы Применение таргетной терапии в лечении пациентов с нейроэндокринными опухолями желудочно- кишечного тракта и поджелудочной железы

  • Diagnosis and management of gastrointestinal neuroendocrine tumors: an evidence-based Canadian consensus / Singh S, Asa SL, Dey C, Kennecke H, Laidley D, Law C. et al. // Cancer Treat. 2016. V. 47. P. 32 - 45.
  • One hundred years after "carcinoid": epidemiology of and prognostiс factors for neuroendocrine tumors in 35,825 cases in the United States / Yao J., Hassan M., Phan A., Dagohoy C., Leary C., Mares JE. et al. // Clin. Oncol. 2008. V. 26. N18. P. 3063 - 3072.
  • Горбунова В. А. Нейроэндокринные опухоли. Общие принципы диагностики и лечения. М.: Кодекс, 2015. 456 с.
  • Gorbunova V. A. Neuroendocrine tumors. General principles of diagnostics and treatment. M.: Kodeks; 2015. 456 р. (In Russian).
  • UKNETwork for neuroendocrine tumours. Guidelines for the management of gastroenteropancreatic neuroendocrine (including carcinoid) tumours / Ramage J., Davies A., Ardill J. et al. // Gut. 2005. V. 54. N4. P. 1 - 16.
  • Pancreatic neuroendocrine tumors (PNETs): incidence, prognosis and recent trend toward improved survival / Halfdanarson T., Rabe K., Rubin J. et al. // Annals of Oncology. 2008. V. 19. N10. P. 1727 - 1733.
  • Modlin I. M., Lye K. D., Kidd M. A. 5-decade analysis of 13,715 carcinoid tumors // Cancer. 2003. V. 97. P. 934 - 959.
  • Lepage C, Rachet B, Coleman MP. Survival from malignant digestive endocrine tumors in England and Wales: a population-based study // Gastroenterology. 2007. V. 132. P. 899 - 904.
  • Population-based study of islet cell carcinoma / Yao J., Eisner M., Leary C. et al. // Ann Surg Oncol. 2007. V. 14. P. 492 - 500.
  • Moertel C., Kvols L., O'Connell M., 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. V. 68. P. 227 - 232.
  • Moertel C., Hanley J. Combination chemotherapy trials in metastatic carcinoid tumor and the malignant carcinoid syndrome // Cancer clinical trials. 1979. V. 2. P. 327 - 334.
  • Практические рекомендации Российского общества клинической онкологии. Лекарственное лечение злокачественных опухолей. Поддерживающая терапия в онкологии / под ред. В. М. Моисеенко. // Злокачественные опухоли. Спецвыпуск. 2019. 776 с.
  • Practical recommendations of the Russian Society of Clinical Oncology. Drug treatment of malignant tumors. Supportive care in oncology / ed. V. M. Moiseenko. // Malignant tumors. Special issue. 2019.776 p. (In Russian).
  • O'Toole D., Kiaуmanesh R., Caplin M. ENETS 2016 Consensus Guidelines for the Management of Patients with Digestive Neuroendocrine Tumors: An Update // Neuroendocrinology. 2016. V. 103. P. 117 - 118.
  • Susini C, Buscail L. Rationale for the use of somatostatin analogs as antitumor agents // Annals Oncology. 2006. V. 17. N12. P. 1733 - 1742.
  • Wimmel A., Wiedenmann B., Rosewicz S. Autocrine growth inhibition by transforming growth factor beta-1 (TGFbeta-1) in human neuroendocrine tumour cells // Gut. 2003. V. 52. P. 1308 - 1316.
  • Effects of interferon alpha on vascular endothelial growth factor gene transcription and tumor angiogenesis / von Marschall Z., Scholz A., Cramer T., Schafer G., Schirner M., Oberg K, et al. // Natl Cancer Inst. 2003. V. 95. P. 437 - 448.
  • Chaudhry A., Papanicolaou V., Oberg K., Heldin C., Funa K. Expression of platelet-derived growth factor and its receptors in neuroendocrine tumors of the digestive system // Cancer Res. 1992. V. 52. P. 1006 - 1012.
  • Wulbrand U., Remmert G., Zofel P., Wied M., Arnold R., Fehmann H. mRNA expression patterns of insulin-like growth factor system components in human neuroendocrine tumours // Eur J Clin Invest. 2000. V. 30. P. 729 - 739.
  • Insulin-like growth factor-I is an autocrine regulator of chromogranin A secretion and growth in human neuroendocrine tumor cells / von Wichert G., Jehle P., Hoeflich A., Koschnick S., Dralle H, Wolf E. et al. // Cancer Res. 2000. V. 60. P. 4573 - 4581.
  • Nilsson O., Wangberg B., Theodorsson E., Skottner A., Ahlman H. Presence of IGF-I in human midgut carcinoid tumours-an autocrine regulator of carcinoid tumour growth? // Int J Cancer. 1992. V. 51. P. 195 - 203.
  • Zhang H., Yee D. The therapeutic potential of agents targeting the type I insulin-like growth factor receptor // Expert Opin Investig Drugs. 2004. V. 13. P. 1569 - 1577.
  • Gastrointestinal Neuroendocrine Tumors: Pancreatic Endocrine Tumors / Metz, David C. et al. // Gastroenterology. 2008. V. 135. N5. P. 1469 - 1492.
  • Wiedmann M., Caca K. Molecularly targeted therapy for gastrointestinal cancer // Curr Cancer Drug Targets. 2005. V. 5. P. 171 - 193.
  • Smolewski P. Recent developments in targeting the mammalian target of rapamycin (mTOR) kinase pathway // Anticancer Drugs. 2006. V. 17. P. 487 - 494.
  • Dancey J. Therapeutic targets: MTOR and related pathways // Cancer Biol Ther. 2006. V. 5. P. 1065 - 1073.
  • EGFR targeting of solid tumors / Rocha-Lima C., Soares H., Raez L. et al. // Cancer Control. 2007. V. 14. P. 295 - 304.
  • Harari P. Epidermal growth factor receptor inhibition strategies in oncology // Endocr Relat Cancer. 2004. V. 11. P. 689 - 708.
  • Bjornsti M., Houghton P. The TOR pathway: a target for cancer therapy // Nat Rev Cancer. 2004. V. 4. P. 335 - 348.
  • BAY 43 - 9006 exhibits broad spectrum oral antitumor activity and targets the RAF / MEK / ERK pathway and receptor tyrosine kinases involved in tumor progression and angiogenesis / Wilhelm S., Carter C., Tang L., Wilkie D., McNabola A., Rong H. et al.// Cancer Res. 2004. V. 64. P. 7099 - 7109.
  • Interaction between the epidermal growth factor receptor (EGFR) and the vascular endothelial growth factor (VEGF) pathways: a rational approach for multi-target anticancer therapy / Ciardiello F., Troiani T., Bianco R., Orditura M., Morgillo F., Martinelli E. et al. // Annals of Oncology. 2006. V. 17. N 7. P. 109 - 114.
  • Maione P., Gridelli C., Troiani T., Ciardiello F. Combining targeted therapies and drugs with multiple targets in the treatment of NSCLC// Oncologist. 2006. V. 11. P. 274 - 284.
  • Burgaud JL, Baserga R. Intracellular transactivation of the insulin-like growth factor I receptor by an epidermal growth factor receptor // Exp Cell Res. 1996. V. 223. P. 412 - 419.
  • Activation of BAD by therapeutic inhibition of epidermal growth factor receptor and transactivation by insulin-like growth factor receptor / Gilmore A., Valentijn A., Wang P., Ranger A., Bundred N., O'Hare M. et al.//J Biol Chem. 2002. V. 277. P. 27643 - 27650.
  • Jain R., Duda D., Clark J., Loeffler J. Lessons from phase III clinical trials on anti-VEGF therapy for cancer // Nat Clin Pract Oncol. 2006. V. 3. P. 24 - 40.
  • Morabito A., De Maio E., Di Maio M., Normanno N., Perrone F. Tyrosine kinase inhibitors of vascular endothelial growth factor receptors in clinical trials: current status and future directions // Oncologist. 2006. V. 11. P. 753 - 764.
  • Hopfner M., Sutter A., Gerst B., Zeitz M., Scherubl H. A novel approach in the treatment of neuroendocrine gastrointestinal tumours. Targeting the epidermal growth factor receptor by gefitinib (ZD1839) // Br J Cancer. 2003. V. 89. P. 1766 - 1775.
  • Hopfner M., Baradari V., Huether A., Schofl C., Scherubl H. The insulin-like growth factor receptor 1 is a promising target for novel treatment approaches in neuroendocrine gastrointestinal tumours // Endocr Relat Cancer. 2006. V. 13. P. 135 - 149.
  • Elevated expression of vascular endothelial growth factor correlates with increased angiogenesis and decreased progression-free survival among patients with low-grade neuroendocrine tumors / Zhang J., Jia Z., Li Q., Wang L., Rashid A, Zhu Z. et al. // Cancer. 2007. V. 109. P. 1478 - 1486.
  • Pavel M., Hassler G., Baum U., Hahn E., Lohmann T., Schuppan D. Circulating levels of angiogenic cytokines can predict tumour progression and prognosis in neuroendocrine carcinomas // Clin Endocrinol (Oxf). 2005. V. 62. P. 434 - 443.
  • Carmeliet P., Jain R. Molecular mechanisms and clinical applications of angiogenesis // Nature 473. 2011. P. 298 - 307.
  • Anti-vascular endothelial growth factor antibody single therapy for pancreatic neuroendocrine carcinoma exhibits a marked tumor growth-inhibitory effect / Kasuya K., Nagakawa Y., Suzuki M., Tanaka H., Ohta H., Itoi T. et al. // Exp Ther Med 2. 2011. P. 1047 - 1052.
  • Prospective study of bevacizumab plus temozolomide in patients with advanced neuroendocrine tumors / Chan J., Stuart K., Earle C., Clark J., Bhargava P., Miksad R. et al. // J Clin Oncol. 2012. V. 30. P. 2963 - 2968.
  • Reinacher-Schick, ASCO-update 2015 - highlights of the 51 meeting of the american society of clinical oncology // Lorenzen S., Arnold D., Fottner C., Leichsenring J., Moehler M., Seufferlein T., et al. //ASCO 2015, Z Gastroenterol. 2016. V. 54. N 02. P. 167 - 172.
  • (2015b) Randomized phase II study of everolimus (E) versus everolimus plus bevacizumab (E+B) in patients (Pts) with locally advanced or metastatic pancreatic neuroendocrine tumors (pNET), CALGB 80701 (Alliance) / Kulke M., Niedzwiecki D., Foster N., Fruth B., Kunz P., Kennecke H. et al.// J Clin Oncol. 2015. V. 33. N 15. P. 4005.
  • Randomized phase II study of everolimus (E) versus everolimus plus bevacizumab (E+B) in patients (Pts) with locally advanced or metastatic pancreatic neuroendocrine tumors (pNET), CALGB 80701 (Alliance) / Matthew H. Kulke, Donna Niedzwiecki, Nathan R. Foster, Briant Fruth, Pamela L. Kunz, Hagen F. Kennecke et al. // J Clin Oncol. 2015. V. 33. N 15. P. 4005.
  • 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 / Yao J., Guthrie K., Moran C. et al. // J Clin Oncol. 2017. 10.1200 / JCO. 2016. V. 70. P. 4072
  • DOI: 10.1200/JCO.2016.V.70.P.4072
  • Перспективы использования бевацизумаба при НЭО. Клинический случай / Г. С. Емельянова, Н. Ф. Орел, В. А. Горбунова, А. А. Коломейцева, А. А. Кузнецова, А. Е. Кузьминов и др. // Сибирский онкологический журнал. 2017. Т. 16. № 6. C. 100 - 104.
  • Perspectives on use of bevacizumab in patients with neuroendocrine tumors. Case report / G. S. Emelianova, N. F. Orel, V. A. Gorbunova, A. A. Kolomeytseva, A. A. Kuznetsova, A. E. Kuzminov et al. // Siberian Journal of Oncology. 2017. V. 16. N 6. P. 100 - 104 (In Russian).
  • Bevacizumab plus octreotide and metronomic capecitabine in patients with metastatic well-to-moderately differentiated neuroendocrine tumors: the xelbevoct study / Berruti A., Fazio N., Ferrero A., Brizzi M., Volante M., Nobili E. et al. // BMC Cancer. 2014. V. 14. P. 184.
  • Bevacizumab plus capecitabine in patients with progressive advanced well-differentiated neuroendocrine tumors of the gastro-intestinal (GI-NETs) tract (BETTER trial) - a phase II non-randomised trial / Mitry E., Walter T., Baudin E., Kurtz J. E., Ruszniewski P., Dominguez-Tinajero S. et al. // Eur J Cancer. 2014. V. 50. N 18. P. 3107 - 3115.
  • Resistance to antiangiogenic therapy is directed by vascular phenotype, vessel stabilization, and maturation in malignant melanoma / Helfrich I., Scheffrahn I., Bartling S., Weis J., von Felbert V., Middleton M. et al. // J Exp Med 207. 2010. P. 491 - 503.
  • Sitohy B., Nagy J., Dvorak H. Anti-VEGF / VEGFR therapy for cancer: Reassessing the target // Cancer Res 72. 2012. P. 1909 - 1914.
  • Combined inhibition of VEGF and PDGF signaling enforces tumor vessel regression by interfering with pericyte- mediated endothelial cell survival mechanisms / Erber R., Thurnher A., Katsen A., Groth G., Kerger H., Hammes H. et al. // FASEB J 18. 2004. P. 338 - 340.
  • Wilhelm S., Chien D. BAY 43 - 9006: preclinical data // Curr Pharm Des. 2002. V. 8. P. 2255 - 2257.
  • Role of Raf kinase in cancer: therapeutic potential of targeting the Raf / MEK / ERK signal transduction pathway / Gollob J., Wilhelm S., Carter C., Kelley S. //Semin Oncol. 2006. V. 33. P. 392 - 406.
  • Sorafenib blocks the RAF / MEK / ERK pathway, inhibits tumor angiogenesis, and induces tumor cell apoptosis in hepatocellular carcinoma model PLC / PRF / 5 / Liu L., Cao Y., Chen C., Zhang X., McNabola A., Wilkie D. et al. // Cancer Res. 2006. V. 66. P. 11851 - 11858.
  • Hobday TJ, Rubin J, Holen K. MC044h, a phase II trial of sorafenib in patients with metastatic neuroendocrine tumors (NET): A Phase II Consortium (P2C) study // Clin Oncol. 2007, ASCO Annual Meeting Proceedings Part 1. 2007. V. 25. N 18. P. 4504.
  • Activity of Sunitinib in Patients With Advanced Neuroendocrine Tumors / Matthew H. Kulke, Heinz-Josef Lenz, Neal J. Meropol et al. // Clin Oncol. 2008. V. 20. P. 3404 - 3410.
  • Sunitinib malate for the treatment of pancreatic neuroendocrine tumors Raymond E., Dahan L., Raoul J. et al. // N Engl J Med. 2011. V. 364. Р. 501 - 513.
  • Motzer R., Bukowski R. Targeted therapy for metastatic renal cell carcinoma // Clin Oncol. 2006. V. 24. P. 5601 - 5608.
  • Kulke M., Bergsland E., Ryan D. A Phase II study to evaluate the safety and efficacy of SU11248 in patients with unresectable neuroendocrine tumors // Proc Am Soc Clin Oncol. 2003. V. 22. P. 958.
  • A phase two study to evaluate the efficacy and safety of SU11248 in patients (pts) with unresectable neuroendocrine tumors (NET) // Kulke M., Lenz H., Meropol N., Posey J., Ryan D., Picus J. et al. // J Clin Oncol. 2005. V. 23. P. 4008.
  • Updated results of the phase III trial of sunitinib (SU) versus placebo (PBO) for treatment of advanced pancreatic neuroendocrine tumors (NET) / Raymond E., Niccoli-Sire P., Bang Y. et al. // ASCO Gastrointestinal Cancers Symposium. Book of abstracts. 2010. Abstract 127.
  • Pietras K., Hanahan D. A multitargeted, metronomic, and maximum-tolerated dose "chemo-switch" regimen is antiangiogenic, producing objective responses and survival benefit in a mouse model of cance // Clin Oncol. 2005. V. 23. P. 939 - 952.
  • Combined anti-VEGFR and anti-PDGFR actions of sunitinib on blood vessels in preclinical tumor models / Yao V., Sennino B., Davis R. et al.// Eur J Cancer. 2006. V. 4. P. 27 - 28.
  • Activity of sunitinib in patients with advanced neuroendocrine tumors / Kulke M., Lenz H., Meropol N. et al. // Clin Oncol. 2008. V. 26. P. 3403 - 3410.
  • Phase 11 study of sunitinib (SU) in Japanese patients with unresectable or metastatic, well-differentiated pancreatic neuroendocrine tumors (NET) / Okusaka T., Ito T., Nishida T. et al. // Clin Oncol. 2012. V. 30. P. 381.
  • Chan J., Kulke M. Targeting the mTOR signaling pathway in neuroendocrine tumors// Curr Treat Options Oncol. 2014. V. 15. N3. P. 365 - 379. 10.1007 / s11864-014-0294-4.
  • DOI: 10.1007/s11864-014-0294-4
  • Симоненко В. Б., Дулин П. А., Маканин М. А. Возможности таргетной терапии нейроэндокринных опухолей // Клиническая медицина. 2014. Т. 92. № 8. С. 5 - 14.
  • Simonenko V. B., Dulin P. A., Makanin M. A. Possibilities for targeted therapy of neuroendocrine tumours // Klinicheskaya meditsina. 2014. V. 92. N 8. P. 5 - 14. (In Russian)].
  • mTOR inhibition induces upstream receptor tyrosine kinase signaling and activates / O'Reilly K., Rojo F., She Q., Solit D., Mills G., Smith D. et al. // Akt. Cancer Research 66. 2006. P. 1500 - 1508.
  • A randomized, double-blind, placebo-controlled, multicenter phase III trial of everolimus + octreotide LAR vs placebo + octreotide lar in patients with advanced neuroendocrine tumors (NET) (RADIANT-2) / Pavel M., Hainsworth J., Baudin E. et al. // 35th ESMO Congress. Book of abstracts. 2010. Abstract LBA8.
  • Everolimus treatment for neuroendocrine tumors: latest results and clinical potential / Pusceddu S., Verzoni E., Prinzi N. et al. // Therapeutic Advances in Medical Oncology. 2017. V. 9. N 3. P. 183 - 188. 10.1177 / 1758834016683905.
  • DOI: 10.1177/1758834016683905
  • Everolimus for advanced pancreatic neuroendocrine tumors / Yao J., Shah M., Ito T. et al. // N Engl J Med. 2011. V. 364. P. 514 - 523.
  • Кузьминов А. Е., Полозкова С. А., Орел Н. Ф., Горбунова В. А. Нейроэндокринные опухоли // Эффективная фармакотерапия. Онкология, гематология и радиология. 2012. N1. С. 44 - 48.
  • Kuzminov A. E., Polozkova S. A., Orel N. F., Gorbunova V. A. Neuroendocrine tumors // Effective pharmacotherapy. Oncology, hematology and radiology. 2012. N 1. P. 44 - 48. (In Russian).
  • A randomized, double-blind, placebo-controlled, multicenter phase III trial of everolimus in patients with advanced pancreatic neuroendocrine tumors (PNET) (RADIANT-3) / Yao J., Shah M., Ito T. et al. // 35th ESMO Congress. Book of abstracts. 2010. Abstract LBA9.
  • Jensen R., Delle Fave G. Promising advances in the treatment of malignant pancreatic endocrine tumors // N Engl J Med. 2011. V. 364. P. 564 - 565.
  • 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 / Yao C., Fazio N., Singh S., Buzzoni R., Carnaghi C., Wolin E. et al. // The Lancet. 5 - 11 March 2016. V. 387. N10022. P. 968 - 977.
  • Health-related quality of life for everolimus versus placebo in patients with advanced, non-functional, well-differentiated gastrointestinal or lung neuroendocrine tumours (RADIANT-4): a multicentre, randomised, double-blind, placebo- controlled, phase 3 trial / Pavel M., Singh S., Strosberg J., Bubuteishvili-Pacaud L., Degtyarev E., Neary M. et al. // The Lancet. October 2017. V. 18, N10, P. 1411 - 1422.
  • Chan J., Kulke M. Progressing in the treatment of neuroendocrine tumors // Curr. Oncol. Rep. 2009. V. 11. P. 193 - 199.
  • Targeted Systemic Treatment of Neuroendocrine Tumors: Current Options and Future Perspectives / Herrera-Martínez, Aura D et al.// Drugs. 2019. V. 79. N1. P. 21 - 42.
Еще
Статья научная