Skip to main content
Erschienen in: Cancer and Metastasis Reviews 1/2011

01.03.2011

Innovations therapy: mammalian target of rapamycin (mTOR) inhibitors for the treatment of neuroendocrine tumors

verfasst von: Jaume Capdevila, Ramón Salazar, Irene Halperín, Albert Abad, James C. Yao

Erschienen in: Cancer and Metastasis Reviews | Sonderheft 1/2011

Einloggen, um Zugang zu erhalten

Abstract

Gastroenteropancreatic neuroendocrine tumors (GEP-NETs) are rare neoplasms that require a multidisciplinary approach for an optimal management. The lack of effectiveness of traditional DNA-damaging agents has led to the exploration of new targeted drugs in order to exploit phenotypical features of GEP-NET therapy. However, due to the orphan setting of these tumors, deeper characterization of molecular features and pathways that characterize cell growth, apoptosis, angiogenesis, and invasion are lacking, particularly genetic mutations or epigenetic alterations that generate oncogenic dependency or even addiction. The PI3K-AKT-mTOR pathway has been implicated as having a crucial role in GEP-NETs not only due to the overexpression of several growth factors and their receptors that finally activate this axis but also hereditary syndromes with constitutive activation of the mTOR pathway with high incidence of GEP-NETs. In this article, we aim to review the recent development of the main molecules that target mTOR complex and have showed promising activity in the treatment of GEPNETs.
Literatur
1.
Zurück zum Zitat Hansel, D. E., Rahman, A., Hermans, J., et al. (2003). Liver metastases arising from well-differentiated pancreatic endocrine neoplasms demonstrate increased VEGF-C expression. Modern Pathology, 16(7), 652–659.PubMedCrossRef Hansel, D. E., Rahman, A., Hermans, J., et al. (2003). Liver metastases arising from well-differentiated pancreatic endocrine neoplasms demonstrate increased VEGF-C expression. Modern Pathology, 16(7), 652–659.PubMedCrossRef
2.
Zurück zum Zitat Zhang, J., Jia, Z., Li, Q., et al. (2007). Elevated expression of vascular endothelial growth factor correlates with increased angiogenesis and decreased progression-free survival among patients with low-grade neuroendocrine tumors. Cancer, 109(8), 1478–1486.PubMedCrossRef Zhang, J., Jia, Z., Li, Q., et al. (2007). Elevated expression of vascular endothelial growth factor correlates with increased angiogenesis and decreased progression-free survival among patients with low-grade neuroendocrine tumors. Cancer, 109(8), 1478–1486.PubMedCrossRef
3.
Zurück zum Zitat Furukawa, M., Raffeld, M., Mateo, C., et al. (2005). Increased expression of insulin-like growth factor I and/or its receptor in gastrinomas is associated with low curability, increased growth, and development of metastases. Clinical Cancer Research, 11(9), 3233–3242.PubMedCrossRef Furukawa, M., Raffeld, M., Mateo, C., et al. (2005). Increased expression of insulin-like growth factor I and/or its receptor in gastrinomas is associated with low curability, increased growth, and development of metastases. Clinical Cancer Research, 11(9), 3233–3242.PubMedCrossRef
4.
Zurück zum Zitat Chaudhry, A., Papanicolaou, V., Oberg, K., et al. (1992). Expression of platelet-derived growth factor and its receptors in neuroendocrine tumors of the digestive system. Cancer Research, 52(4), 1006–1012.PubMed Chaudhry, A., Papanicolaou, V., Oberg, K., et al. (1992). Expression of platelet-derived growth factor and its receptors in neuroendocrine tumors of the digestive system. Cancer Research, 52(4), 1006–1012.PubMed
5.
Zurück zum Zitat Nilsson, O., Wangberg, B., Kolby, L., et al. (1995). Expression of transforming growth factor alpha and its receptor in human neuroendocrine tumours. International Journal of Cancer, 60(5), 645–651.CrossRef Nilsson, O., Wangberg, B., Kolby, L., et al. (1995). Expression of transforming growth factor alpha and its receptor in human neuroendocrine tumours. International Journal of Cancer, 60(5), 645–651.CrossRef
6.
Zurück zum Zitat Fjällskog, M. L., Lejonklou, M. H., Oberg, K. E., et al. (2003). Expression of molecular targets for tyrosine kinase receptor antagonists in malignant endocrine pancreatic tumors. Clinical Cancer Research, 9(4), 1469–1473.PubMed Fjällskog, M. L., Lejonklou, M. H., Oberg, K. E., et al. (2003). Expression of molecular targets for tyrosine kinase receptor antagonists in malignant endocrine pancreatic tumors. Clinical Cancer Research, 9(4), 1469–1473.PubMed
7.
Zurück zum Zitat Wulbrand, U., Remmert, G., Zofel, P., et al. (2000). mRNA expression patterns of insulin-like growth factor system components in human neuroendocrine tumours. European Journal of Clinical Investigation, 30(8), 729–739.PubMedCrossRef Wulbrand, U., Remmert, G., Zofel, P., et al. (2000). mRNA expression patterns of insulin-like growth factor system components in human neuroendocrine tumours. European Journal of Clinical Investigation, 30(8), 729–739.PubMedCrossRef
8.
Zurück zum Zitat Kaltsas, G. A., Besser, G. M., & Grossman, A. B. (2004). The diagnosis and medical management of advanced neuroendocrine tumors. Endocrine Reviews, 25(3), 458–511.PubMedCrossRef Kaltsas, G. A., Besser, G. M., & Grossman, A. B. (2004). The diagnosis and medical management of advanced neuroendocrine tumors. Endocrine Reviews, 25(3), 458–511.PubMedCrossRef
9.
Zurück zum Zitat O’Toole, D., Couvelard, A., Rebours, V., et al. (2010). Molecular markers associated with response to chemotherapy in gastro-entero-pancreatic neuroendocrine tumors. Endocrine-Related Cancer, 17(4), 847–856.PubMedCrossRef O’Toole, D., Couvelard, A., Rebours, V., et al. (2010). Molecular markers associated with response to chemotherapy in gastro-entero-pancreatic neuroendocrine tumors. Endocrine-Related Cancer, 17(4), 847–856.PubMedCrossRef
10.
Zurück zum Zitat Kolby, L., Persson, G., Franzen, S., et al. (2003). Randomized clinical trial of the effect of interferon alpha on survival in patients with disseminated midgut carcinoid tumours. The British Journal of Surgery, 90(6), 687–693.PubMedCrossRef Kolby, L., Persson, G., Franzen, S., et al. (2003). Randomized clinical trial of the effect of interferon alpha on survival in patients with disseminated midgut carcinoid tumours. The British Journal of Surgery, 90(6), 687–693.PubMedCrossRef
11.
Zurück zum Zitat Moertel, C. G., Rubin, J., & Kvols, L. K. (1989). Therapy of metastatic carcinoid tumor and the malignant carcinoid syndrome with recombinant leukocyte A interferon. Journal of Clinical Oncology, 7(7), 865–868.PubMed Moertel, C. G., Rubin, J., & Kvols, L. K. (1989). Therapy of metastatic carcinoid tumor and the malignant carcinoid syndrome with recombinant leukocyte A interferon. Journal of Clinical Oncology, 7(7), 865–868.PubMed
12.
Zurück zum Zitat Öberg, K., Norheim, I., Lind, E., et al. (1986). Treatment of malignant carcinoid tumors with human leukocyte interferon: Long-term results. Cancer Treatment Reports, 70(11), 1297–1304.PubMed Öberg, K., Norheim, I., Lind, E., et al. (1986). Treatment of malignant carcinoid tumors with human leukocyte interferon: Long-term results. Cancer Treatment Reports, 70(11), 1297–1304.PubMed
13.
Zurück zum Zitat Dirix, L. Y., Vermeulen, P. B., Fierens, H., et al. (1996). Long-term results of continuous treatment with recombinant interferon-alpha in patients with metastatic carcinoid tumors—an antiangiogenic effect? Anti-Cancer Drugs, 7(2), 175–181.PubMedCrossRef Dirix, L. Y., Vermeulen, P. B., Fierens, H., et al. (1996). Long-term results of continuous treatment with recombinant interferon-alpha in patients with metastatic carcinoid tumors—an antiangiogenic effect? Anti-Cancer Drugs, 7(2), 175–181.PubMedCrossRef
14.
Zurück zum Zitat Rinke, A., Muller, H. H., Schade-Brittinger, C., et al. (2009). 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. Journal of Clinical Oncology, 27(28), 4656–4663.PubMedCrossRef Rinke, A., Muller, H. H., Schade-Brittinger, C., et al. (2009). 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. Journal of Clinical Oncology, 27(28), 4656–4663.PubMedCrossRef
15.
Zurück zum Zitat Moertel, C. G., Lefkopoulo, M., Lipsitz, S., et al. (1992). Streptozocin-doxorubicin, streptozocin-fluorouracil or chlorozotocin in the treatment of advanced islet-cell carcinoma. The New England Journal of Medicine, 326(8), 519–523.PubMedCrossRef Moertel, C. G., Lefkopoulo, M., Lipsitz, S., et al. (1992). Streptozocin-doxorubicin, streptozocin-fluorouracil or chlorozotocin in the treatment of advanced islet-cell carcinoma. The New England Journal of Medicine, 326(8), 519–523.PubMedCrossRef
16.
Zurück zum Zitat Kouvaraki, M. A., Ajani, J. A., Hoff, P., et al. (2004). Fluorouracil, doxorubicin, and streptozocin in the treatment of patients with locally advanced and metastatic pancreatic endocrine carcinomas. Journal of Clinical Oncology, 22(23), 4762–4771.PubMedCrossRef Kouvaraki, M. A., Ajani, J. A., Hoff, P., et al. (2004). Fluorouracil, doxorubicin, and streptozocin in the treatment of patients with locally advanced and metastatic pancreatic endocrine carcinomas. Journal of Clinical Oncology, 22(23), 4762–4771.PubMedCrossRef
17.
Zurück zum Zitat Sun, W., Lipsitz, S., Catalano, P., et al. (2005). Phase II/III study of doxorubicin with fluorouracil compared with streptozocin with fluorouracil or dacarbazine in the treatment of advanced carcinoid tumors: Eastern Cooperative Oncology Group Study E1281. Journal of Clinical Oncology, 23(22), 4897–4904.PubMedCrossRef Sun, W., Lipsitz, S., Catalano, P., et al. (2005). Phase II/III study of doxorubicin with fluorouracil compared with streptozocin with fluorouracil or dacarbazine in the treatment of advanced carcinoid tumors: Eastern Cooperative Oncology Group Study E1281. Journal of Clinical Oncology, 23(22), 4897–4904.PubMedCrossRef
18.
Zurück zum Zitat Öberg, K., & Jelic, S. (2009). Neuroendocrine gastroenteropancreatic tumors: ESMO clinical recommendation for diagnosis, treatment and follow-up. Annals of Oncology, 20(Suppl 4), 150–153.PubMed Öberg, K., & Jelic, S. (2009). Neuroendocrine gastroenteropancreatic tumors: ESMO clinical recommendation for diagnosis, treatment and follow-up. Annals of Oncology, 20(Suppl 4), 150–153.PubMed
19.
Zurück zum Zitat Öberg, K., Kvols, L., Caplin, M., et al. (2004). Consensus report on the use of somatostatin analogs for the management of neuroendocrine tumors of the gastroenteropancreatic system. Annals of Oncology, 15(6), 966–973.PubMedCrossRef Öberg, K., Kvols, L., Caplin, M., et al. (2004). Consensus report on the use of somatostatin analogs for the management of neuroendocrine tumors of the gastroenteropancreatic system. Annals of Oncology, 15(6), 966–973.PubMedCrossRef
20.
Zurück zum Zitat Capdevila, J., & Salazar, R. (2009). Molecular targeted therapies in the treatment of gastroenteropancreatic neuroendocrine tumors. Target Oncology, 4(4), 287–296.CrossRef Capdevila, J., & Salazar, R. (2009). Molecular targeted therapies in the treatment of gastroenteropancreatic neuroendocrine tumors. Target Oncology, 4(4), 287–296.CrossRef
21.
Zurück zum Zitat Wullschleger, S., Loewith, R., & Hall, M. N. (2006). TOR signaling in growth and metabolism. Cell, 124(3), 471–484.PubMedCrossRef Wullschleger, S., Loewith, R., & Hall, M. N. (2006). TOR signaling in growth and metabolism. Cell, 124(3), 471–484.PubMedCrossRef
22.
Zurück zum Zitat Meric-Bernstam, F., & Gonzalez-Angulo, A. M. (2009). Targeting the mTOR signaling network for cancer therapy. Journal of Clinical Oncology, 27(13), 2278–2287.PubMedCrossRef Meric-Bernstam, F., & Gonzalez-Angulo, A. M. (2009). Targeting the mTOR signaling network for cancer therapy. Journal of Clinical Oncology, 27(13), 2278–2287.PubMedCrossRef
23.
Zurück zum Zitat Arcaro, A., & Guerreiro, A. S. (2007). The phosphoinositide 3-kinase pathway in human cancer: genetic alterations and therapeutic implications. Current Genomics, 8(5), 271–306.PubMedCrossRef Arcaro, A., & Guerreiro, A. S. (2007). The phosphoinositide 3-kinase pathway in human cancer: genetic alterations and therapeutic implications. Current Genomics, 8(5), 271–306.PubMedCrossRef
24.
Zurück zum Zitat Yuan, R., Kay, A., Berg, W. J., et al. (2009). Targeting tumorigenesis: development and use of mTOR inhibitors in cancer therapy. Journal of Hematology and Oncology, 2, 45.PubMedCrossRef Yuan, R., Kay, A., Berg, W. J., et al. (2009). Targeting tumorigenesis: development and use of mTOR inhibitors in cancer therapy. Journal of Hematology and Oncology, 2, 45.PubMedCrossRef
25.
Zurück zum Zitat Vignot, S., Faivre, S., Aguirre, D., et al. (2005). mTOR-targeted therapy of cancer with rapamycin derivatives. Annals of Oncology, 16(4), 525–537.PubMedCrossRef Vignot, S., Faivre, S., Aguirre, D., et al. (2005). mTOR-targeted therapy of cancer with rapamycin derivatives. Annals of Oncology, 16(4), 525–537.PubMedCrossRef
26.
Zurück zum Zitat Franke, T. F., Hornik, C. P., Segev, L., et al. (2003). PI3K/Akt and apoptosis: size matters. Oncogene, 22(56), 8983–8998.PubMedCrossRef Franke, T. F., Hornik, C. P., Segev, L., et al. (2003). PI3K/Akt and apoptosis: size matters. Oncogene, 22(56), 8983–8998.PubMedCrossRef
27.
Zurück zum Zitat Paez, J., & Sellers, W. R. (2003). PI3K/PTEN/AKT pathway. A critical mediator of oncogenic signaling. Cancer Treatment and Research, 115, 145–167.PubMedCrossRef Paez, J., & Sellers, W. R. (2003). PI3K/PTEN/AKT pathway. A critical mediator of oncogenic signaling. Cancer Treatment and Research, 115, 145–167.PubMedCrossRef
28.
Zurück zum Zitat LoPiccolo, J., Blumenthal, G. M., Bernstein, W. B., et al. (2008). Targeting the PI3K/Akt/mTOR pathway: effective combinations and clinical considerations. Drug Resistance Updates, 11(1–2), 32–50.PubMedCrossRef LoPiccolo, J., Blumenthal, G. M., Bernstein, W. B., et al. (2008). Targeting the PI3K/Akt/mTOR pathway: effective combinations and clinical considerations. Drug Resistance Updates, 11(1–2), 32–50.PubMedCrossRef
29.
Zurück zum Zitat von Wichert, G., Jehle, P. M., Hoeflich, A., et al. (2000). Insulin-like growth factor-I is an autocrine regulator of chromogranin A secretion and growth in human neuroendocrine tumor cells. Cancer Research, 60(16), 4573–4581. von Wichert, G., Jehle, P. M., Hoeflich, A., et al. (2000). Insulin-like growth factor-I is an autocrine regulator of chromogranin A secretion and growth in human neuroendocrine tumor cells. Cancer Research, 60(16), 4573–4581.
30.
Zurück zum Zitat Van Gompel, J. J., & Chen, H. (2004). Insulin-like growth factor 1 signaling in human gastrointestinal carcinoid tumor cells. Surgery, 136(6), 1297–1302.PubMedCrossRef Van Gompel, J. J., & Chen, H. (2004). Insulin-like growth factor 1 signaling in human gastrointestinal carcinoid tumor cells. Surgery, 136(6), 1297–1302.PubMedCrossRef
31.
Zurück zum Zitat Zatelli, M. C., Minoia, M., Martini, C., et al. (2010). Everolimus as a new potential antiproliferative agent in aggressive human bronchial carcinoids. Endocrine-Related Cancer, 17(3), 719–729.PubMedCrossRef Zatelli, M. C., Minoia, M., Martini, C., et al. (2010). Everolimus as a new potential antiproliferative agent in aggressive human bronchial carcinoids. Endocrine-Related Cancer, 17(3), 719–729.PubMedCrossRef
32.
Zurück zum Zitat Tee, A. R., Fingar, D. C., Manning, B. D., et al. (2002). Tuberous sclerosis complex-1 and -2 gene products function together to inhibit mammalian target of rapamycin (mTOR)-mediated downstream signaling. Proceedings of the National Academy of Sciences of the United States of America, 99(21), 13571–13576.PubMedCrossRef Tee, A. R., Fingar, D. C., Manning, B. D., et al. (2002). Tuberous sclerosis complex-1 and -2 gene products function together to inhibit mammalian target of rapamycin (mTOR)-mediated downstream signaling. Proceedings of the National Academy of Sciences of the United States of America, 99(21), 13571–13576.PubMedCrossRef
33.
Zurück zum Zitat Verhoef, S., van Diemen-Steenvoorde, R., Akkersdijk, W. L., et al. (1999). Malignant pancreatic tumour within the spectrum of tuberous sclerosis complex in childhood. European Journal of Pediatrics, 158(4), 284–287.PubMedCrossRef Verhoef, S., van Diemen-Steenvoorde, R., Akkersdijk, W. L., et al. (1999). Malignant pancreatic tumour within the spectrum of tuberous sclerosis complex in childhood. European Journal of Pediatrics, 158(4), 284–287.PubMedCrossRef
34.
Zurück zum Zitat Eledrisi, M. S., Stuart, C. A., & Alshanti, M. (2002). Insulinoma in a patient with tuberous sclerosis: Is there an association? Endocrine Practice, 8(2), 109–112.PubMed Eledrisi, M. S., Stuart, C. A., & Alshanti, M. (2002). Insulinoma in a patient with tuberous sclerosis: Is there an association? Endocrine Practice, 8(2), 109–112.PubMed
35.
Zurück zum Zitat Dworakowska, D., & Grossman, A. B. (2009). Are neuroendocrine tumours a feature of tuberous sclerosis? A systematic review. Endocrine-Related Cancer, 16(1), 45–58.PubMedCrossRef Dworakowska, D., & Grossman, A. B. (2009). Are neuroendocrine tumours a feature of tuberous sclerosis? A systematic review. Endocrine-Related Cancer, 16(1), 45–58.PubMedCrossRef
36.
Zurück zum Zitat Hattori, S., Maekawa, M., & Nakamura, S. (1992). Identification of neurofibromatosis type I gene product as an insoluble GTPase-activating protein toward ras p21. Oncogene, 7(3), 481–485.PubMed Hattori, S., Maekawa, M., & Nakamura, S. (1992). Identification of neurofibromatosis type I gene product as an insoluble GTPase-activating protein toward ras p21. Oncogene, 7(3), 481–485.PubMed
37.
Zurück zum Zitat Johannessen, C. M., Reczek, E. E., James, M. F., et al. (2005). The NF1 tumor suppressor critically regulates TSC2 and mTOR. Proceedings of the National Academy of Sciences of the United States of America, 102(24), 8573–8578.PubMedCrossRef Johannessen, C. M., Reczek, E. E., James, M. F., et al. (2005). The NF1 tumor suppressor critically regulates TSC2 and mTOR. Proceedings of the National Academy of Sciences of the United States of America, 102(24), 8573–8578.PubMedCrossRef
38.
Zurück zum Zitat Blansfield, J. A., Choyke, L., Morita, S. Y., et al. (2007). Clinical, genetic and radiographic analysis of 108 patients with von Hippel–Lindau disease (VHL) manifested by pancreatic neuroendocrine neoplasms (PNETs). Surgery, 142(6), 814–818. discussion 818 e1–2.PubMedCrossRef Blansfield, J. A., Choyke, L., Morita, S. Y., et al. (2007). Clinical, genetic and radiographic analysis of 108 patients with von Hippel–Lindau disease (VHL) manifested by pancreatic neuroendocrine neoplasms (PNETs). Surgery, 142(6), 814–818. discussion 818 e1–2.PubMedCrossRef
39.
Zurück zum Zitat Forsythe, J. A., Jiang, B. H., Iyer, N. V., et al. (1996). Activation of vascular endothelial growth factor gene transcription by hypoxia-inducible factor 1. Molecular and Cellular Biology, 16(9), 4604–4613.PubMed Forsythe, J. A., Jiang, B. H., Iyer, N. V., et al. (1996). Activation of vascular endothelial growth factor gene transcription by hypoxia-inducible factor 1. Molecular and Cellular Biology, 16(9), 4604–4613.PubMed
40.
Zurück zum Zitat Woodward, E. R., & Maher, E. R. (2006). Von Hippel–Lindau disease and endocrine tumour susceptibility. Endocrine-Related Cancer, 13(2), 415–425.PubMedCrossRef Woodward, E. R., & Maher, E. R. (2006). Von Hippel–Lindau disease and endocrine tumour susceptibility. Endocrine-Related Cancer, 13(2), 415–425.PubMedCrossRef
41.
Zurück zum Zitat Thakker, R. V. (2010). Multiple endocrine neoplasia type 1 (MEN1). Best Practice & Research. Clinical Endocrinology & Metabolism, 24(3), 355–370.CrossRef Thakker, R. V. (2010). Multiple endocrine neoplasia type 1 (MEN1). Best Practice & Research. Clinical Endocrinology & Metabolism, 24(3), 355–370.CrossRef
42.
Zurück zum Zitat Debelenko, L. V., Zhuang, Z., Emmert-Buck, M. R., et al. (1997). Allelic deletions on chromosome 11q13 in multiple endocrine neoplasia type 1-associated and sporadic gastrinomas and pancreatic endocrine tumors. Cancer Research, 57(11), 2238–2243.PubMed Debelenko, L. V., Zhuang, Z., Emmert-Buck, M. R., et al. (1997). Allelic deletions on chromosome 11q13 in multiple endocrine neoplasia type 1-associated and sporadic gastrinomas and pancreatic endocrine tumors. Cancer Research, 57(11), 2238–2243.PubMed
43.
Zurück zum Zitat Jakobovitz, O., Nass, D., DeMarco, L., et al. (1996). Carcinoid tumors frequently display genetic abnormalities involving chromosome 11. The Journal of Clinical Endocrinology and Metabolism, 81(9), 3164–3167.PubMedCrossRef Jakobovitz, O., Nass, D., DeMarco, L., et al. (1996). Carcinoid tumors frequently display genetic abnormalities involving chromosome 11. The Journal of Clinical Endocrinology and Metabolism, 81(9), 3164–3167.PubMedCrossRef
44.
Zurück zum Zitat Öberg, K. (2009). Genetics and molecular pathology of neuroendocrine gastrointestinal and pancreatic tumors (gastroenteropancreatic neuroendocrine tumors). Current Opinion in Endocrinology, Diabetes and Obesity, 16(1), 72–78.CrossRef Öberg, K. (2009). Genetics and molecular pathology of neuroendocrine gastrointestinal and pancreatic tumors (gastroenteropancreatic neuroendocrine tumors). Current Opinion in Endocrinology, Diabetes and Obesity, 16(1), 72–78.CrossRef
45.
Zurück zum Zitat Wang, E. H., Ebrahimi, S. A., Wu, A. Y., et al. (1998). Mutation of the MENIN gene in sporadic pancreatic endocrine tumors. Cancer Research, 58(19), 4417–4420.PubMed Wang, E. H., Ebrahimi, S. A., Wu, A. Y., et al. (1998). Mutation of the MENIN gene in sporadic pancreatic endocrine tumors. Cancer Research, 58(19), 4417–4420.PubMed
46.
Zurück zum Zitat Missiaglia, E., Dalai, I., Barbi, S., et al. (2010). Pancreatic endocrine tumors: expression profiling evidences a role for AKT-mTOR pathway. Journal of Clinical Oncology, 28(2), 245–255.PubMedCrossRef Missiaglia, E., Dalai, I., Barbi, S., et al. (2010). Pancreatic endocrine tumors: expression profiling evidences a role for AKT-mTOR pathway. Journal of Clinical Oncology, 28(2), 245–255.PubMedCrossRef
48.
Zurück zum Zitat Durán, I., Kortmansky, J., Singh, D., et al. (2006). A phase II clinical and pharmacodynamic study of temsirolimus in advanced neuroendocrine carcinomas. British Journal of Cancer, 95(9), 1148–1154.PubMedCrossRef Durán, I., Kortmansky, J., Singh, D., et al. (2006). A phase II clinical and pharmacodynamic study of temsirolimus in advanced neuroendocrine carcinomas. British Journal of Cancer, 95(9), 1148–1154.PubMedCrossRef
50.
Zurück zum Zitat Zitzmann, K., De Toni, E. N., Brand, S., et al. (2007). The novel mTOR inhibitor RAD001 (everolimus) induces antiproliferative effects in human pancreatic neuroendocrine tumor cells. Neuroendocrinology, 85(1), 54–60.PubMedCrossRef Zitzmann, K., De Toni, E. N., Brand, S., et al. (2007). The novel mTOR inhibitor RAD001 (everolimus) induces antiproliferative effects in human pancreatic neuroendocrine tumor cells. Neuroendocrinology, 85(1), 54–60.PubMedCrossRef
51.
Zurück zum Zitat Tabernero, J., Rojo, F., Calvo, E., et al. (2008). Dose- and schedule-dependent inhibition of the mammalian target of rapamycin pathway with everolimus: a phase I tumor pharmacodynamic study in patients with advanced solid tumors. Journal of Clinical Oncology, 26(10), 1603–1610.PubMedCrossRef Tabernero, J., Rojo, F., Calvo, E., et al. (2008). Dose- and schedule-dependent inhibition of the mammalian target of rapamycin pathway with everolimus: a phase I tumor pharmacodynamic study in patients with advanced solid tumors. Journal of Clinical Oncology, 26(10), 1603–1610.PubMedCrossRef
52.
Zurück zum Zitat Yao, J. C., Phan, A. T., Chang, D. Z., et al. (2008). Efficacy of RAD001 (Everolimus) and octreotide LAR in advanced low- to intermediate-grade neuroendocrine tumors: results of a phase II study. Journal of Clinical Oncology, 26(26), 4311–4318.PubMedCrossRef Yao, J. C., Phan, A. T., Chang, D. Z., et al. (2008). Efficacy of RAD001 (Everolimus) and octreotide LAR in advanced low- to intermediate-grade neuroendocrine tumors: results of a phase II study. Journal of Clinical Oncology, 26(26), 4311–4318.PubMedCrossRef
53.
Zurück zum Zitat Yao, J. C., Lombard-Bohas, C., Baudin, E., et al. (2010). Daily oral everolimus activity in patients with metastatic pancreatic neuroendocrine tumors after failure of cytotoxic chemotherapy: a phase II trial. Journal of Clinical Oncology, 28(1), 69–76.PubMedCrossRef Yao, J. C., Lombard-Bohas, C., Baudin, E., et al. (2010). Daily oral everolimus activity in patients with metastatic pancreatic neuroendocrine tumors after failure of cytotoxic chemotherapy: a phase II trial. Journal of Clinical Oncology, 28(1), 69–76.PubMedCrossRef
54.
Zurück zum Zitat Pavel, M., Hainsworth, J.D., Baudin, E., et al. (2010). A randomized, double-blind, placebo-controlled, multicenter phase III trial of everolimus plus octreotide LAR vs ersus placebo plus octreotide LAR in patients with advanced neuroendocrine tumors (NET) (RADIANT-2). In: 35th European Medical Oncology Society; 2010; Milan, Italy. Pavel, M., Hainsworth, J.D., Baudin, E., et al. (2010). A randomized, double-blind, placebo-controlled, multicenter phase III trial of everolimus plus octreotide LAR vs ersus placebo plus octreotide LAR in patients with advanced neuroendocrine tumors (NET) (RADIANT-2). In: 35th European Medical Oncology Society; 2010; Milan, Italy.
55.
Zurück zum Zitat Yao, J.C., Shah, M.H., Ito, T., et al. (2010). A randomized, double-blind, placebo-controlled, multicenter phase III trial of everolimus in patients with advanced pancreatic neuroendocrine tumors (PNET) (RADIANT-3). In: 35th European Society Medical Oncology; 2010; Milan, Italy Yao, J.C., Shah, M.H., Ito, T., et al. (2010). A randomized, double-blind, placebo-controlled, multicenter phase III trial of everolimus in patients with advanced pancreatic neuroendocrine tumors (PNET) (RADIANT-3). In: 35th European Society Medical Oncology; 2010; Milan, Italy
Metadaten
Titel
Innovations therapy: mammalian target of rapamycin (mTOR) inhibitors for the treatment of neuroendocrine tumors
verfasst von
Jaume Capdevila
Ramón Salazar
Irene Halperín
Albert Abad
James C. Yao
Publikationsdatum
01.03.2011
Verlag
Springer US
Erschienen in
Cancer and Metastasis Reviews / Ausgabe Sonderheft 1/2011
Print ISSN: 0167-7659
Elektronische ISSN: 1573-7233
DOI
https://doi.org/10.1007/s10555-011-9290-3

Weitere Artikel der Sonderheft 1/2011

Cancer and Metastasis Reviews 1/2011 Zur Ausgabe

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Alectinib verbessert krankheitsfreies Überleben bei ALK-positivem NSCLC

25.04.2024 NSCLC Nachrichten

Das Risiko für Rezidiv oder Tod von Patienten und Patientinnen mit reseziertem ALK-positivem NSCLC ist unter einer adjuvanten Therapie mit dem Tyrosinkinase-Inhibitor Alectinib signifikant geringer als unter platinbasierter Chemotherapie.

Bei Senioren mit Prostatakarzinom auf Anämie achten!

24.04.2024 DGIM 2024 Nachrichten

Patienten, die zur Behandlung ihres Prostatakarzinoms eine Androgendeprivationstherapie erhalten, entwickeln nicht selten eine Anämie. Wer ältere Patienten internistisch mitbetreut, sollte auf diese Nebenwirkung achten.

ICI-Therapie in der Schwangerschaft wird gut toleriert

Müssen sich Schwangere einer Krebstherapie unterziehen, rufen Immuncheckpointinhibitoren offenbar nicht mehr unerwünschte Wirkungen hervor als andere Mittel gegen Krebs.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.