Skip to main content
Erschienen in: Cancer and Metastasis Reviews 1/2014

01.03.2014

Systemic therapeutic strategies for GEP-NETS: what can we expect in the future?

verfasst von: E. Raymond, R. García-Carbonero, B. Wiedenmann, E. Grande, M. Pavel

Erschienen in: Cancer and Metastasis Reviews | Ausgabe 1/2014

Einloggen, um Zugang zu erhalten

Abstract

Over the last few years, there have been important advances in the understanding of the molecular biology of neuroendocrine tumors (NETs) that have already translated into relevant advances in the clinic. Several studies have extensively assessed the mutational profile of NETs, and have shown the key roles that angiogenesis and the PI3K-AKT-mTOR pathway play in the pathogenesis of these tumors. Recent data has also revealed the potential relevance of transcription factors such as death domain-associated protein, x-linked mental retardation, and α-thalassemia syndrome protein or ataxia telangiectasia-mutated in NETs of pancreatic origin. This fast progress is leading to a rapidly increasing number of new agents being explored in the field of NETs. However, and despite some unquestionable success, objective remission rates remain low, and evidence of a substantial survival impact is lacking. Thus, there is an important need to improve our ability to identify patients most likely to benefit from specific therapies, and to incorporate biomarkers in the management of NETs. In addition, further efforts to understand mechanisms of escape and acquired resistance to the different available agents is of utmost importance, and will likely require performing paired tumor biopsies (prior and after treatment) or sequential sampling of surrogate tissues. Combinations of approved agents with new agents, either in a rational or biomarker-driven manner, are certainly warranted in this field. Likewise, sequential strategies to modulate and compensate for escape phenomenons are also of great interest. It should also be noted, however, that targeted agents are not innocuous and frequently yield toxicities that need to be adequately addressed by experienced specialists, particularly when drug combinations are considered. This review summarizes the salient data on biomarker and new agent development for the treatment of NETs.
Literatur
1.
Zurück zum Zitat Corbo, V., et al. (2012). Pancreatic endocrine tumours: mutational and immunohistochemical survey of protein kinases reveals alterations in targetable kinases in cancer cell lines and rare primaries. Annals of Oncology, 23, 127–134.PubMedCentralPubMedCrossRef Corbo, V., et al. (2012). Pancreatic endocrine tumours: mutational and immunohistochemical survey of protein kinases reveals alterations in targetable kinases in cancer cell lines and rare primaries. Annals of Oncology, 23, 127–134.PubMedCentralPubMedCrossRef
2.
Zurück zum Zitat Corbo, V., et al. (2010). MEN1 in pancreatic endocrine tumors: analysis of gene and protein status in 169 sporadic neoplasms reveals alterations in the vast majority of cases. Endocrine-Related Cancer, 17, 771–783.PubMedCrossRef Corbo, V., et al. (2010). MEN1 in pancreatic endocrine tumors: analysis of gene and protein status in 169 sporadic neoplasms reveals alterations in the vast majority of cases. Endocrine-Related Cancer, 17, 771–783.PubMedCrossRef
3.
Zurück zum Zitat Jiao, Y., et al. (2011). DAXX/ATRX, MEN1, and mTOR pathway genes are frequently altered in pancreatic neuroendocrine tumors. Science, 331, 1199–1203.PubMedCentralPubMedCrossRef Jiao, Y., et al. (2011). DAXX/ATRX, MEN1, and mTOR pathway genes are frequently altered in pancreatic neuroendocrine tumors. Science, 331, 1199–1203.PubMedCentralPubMedCrossRef
4.
Zurück zum Zitat Missiaglia, E., et al. (2010). Pancreatic endocrine tumors: expression profiling evidences a role for AKT-mTOR pathway. Journal of Clinical Oncology, 28, 245–255.PubMedCrossRef Missiaglia, E., et al. (2010). Pancreatic endocrine tumors: expression profiling evidences a role for AKT-mTOR pathway. Journal of Clinical Oncology, 28, 245–255.PubMedCrossRef
5.
Zurück zum Zitat Faivre, S., Sablin, M. P., Dreyer, C., & Raymond, E. (2010). Novel anticancer agents in clinical trials for well-differentiated neuroendocrine tumors. Endocrinology and Metabolism Clinics of North America, 39, 811–826.PubMedCrossRef Faivre, S., Sablin, M. P., Dreyer, C., & Raymond, E. (2010). Novel anticancer agents in clinical trials for well-differentiated neuroendocrine tumors. Endocrinology and Metabolism Clinics of North America, 39, 811–826.PubMedCrossRef
6.
Zurück zum Zitat Couvelard, A., et al. (2005). Microvascular density and hypoxia-inducible factor pathway in pancreatic endocrine tumours: negative correlation of microvascular density and VEGF expression with tumour progression. British Journal of Cancer, 92, 94–101.PubMedCentralPubMedCrossRef Couvelard, A., et al. (2005). Microvascular density and hypoxia-inducible factor pathway in pancreatic endocrine tumours: negative correlation of microvascular density and VEGF expression with tumour progression. British Journal of Cancer, 92, 94–101.PubMedCentralPubMedCrossRef
7.
Zurück zum Zitat Rinke, A., 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, 4656–4663.PubMedCrossRef Rinke, A., 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, 4656–4663.PubMedCrossRef
8.
Zurück zum Zitat Raymond, E., et al. (2011). Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. The New England Journal of Medicine, 364, 501–513.PubMedCrossRef Raymond, E., et al. (2011). Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. The New England Journal of Medicine, 364, 501–513.PubMedCrossRef
9.
Zurück zum Zitat Yao, J. C., et al. (2011). Everolimus for advanced pancreatic neuroendocrine tumors. The New England Journal of Medicine, 364, 514–523.PubMedCrossRef Yao, J. C., et al. (2011). Everolimus for advanced pancreatic neuroendocrine tumors. The New England Journal of Medicine, 364, 514–523.PubMedCrossRef
10.
Zurück zum Zitat Ekeblad, S., et al. (2007). Temozolomide as monotherapy is effective in treatment of advanced malignant neuroendocrine tumors. Clinical Cancer Research, 13, 2986–2991.PubMedCrossRef Ekeblad, S., et al. (2007). Temozolomide as monotherapy is effective in treatment of advanced malignant neuroendocrine tumors. Clinical Cancer Research, 13, 2986–2991.PubMedCrossRef
11.
Zurück zum Zitat Kulke, M. H., et al. (2009). O6-methylguanine DNA methyltransferase deficiency and response to temozolomide-based therapy in patients with neuroendocrine tumors. Clinical Cancer Research, 15, 338–345.PubMedCentralPubMedCrossRef Kulke, M. H., et al. (2009). O6-methylguanine DNA methyltransferase deficiency and response to temozolomide-based therapy in patients with neuroendocrine tumors. Clinical Cancer Research, 15, 338–345.PubMedCentralPubMedCrossRef
12.
Zurück zum Zitat Strosberg, J. R., et al. (2011). First-line chemotherapy with capecitabine and temozolomide in patients with metastatic pancreatic endocrine carcinomas. Cancer, 117, 268–275.PubMedCrossRef Strosberg, J. R., et al. (2011). First-line chemotherapy with capecitabine and temozolomide in patients with metastatic pancreatic endocrine carcinomas. Cancer, 117, 268–275.PubMedCrossRef
13.
Zurück zum Zitat Tijeras-Raballand, A., et al. (2012). Resistance to targeted therapies in pancreatic neuroendocrine tumors (PNETs): molecular basis, preclinical data, and counteracting strategies. Targeted Oncology, 7, 173–181.PubMedCrossRef Tijeras-Raballand, A., et al. (2012). Resistance to targeted therapies in pancreatic neuroendocrine tumors (PNETs): molecular basis, preclinical data, and counteracting strategies. Targeted Oncology, 7, 173–181.PubMedCrossRef
14.
Zurück zum Zitat Maira, S. M., et al. (2008). Identification and characterization of NVP-BEZ235, a new orally available dual phosphatidylinositol 3-kinase/mammalian target of rapamycin inhibitor with potent in vivo antitumor activity. Molecular Cancer Therapeutics, 7, 1851–1863.PubMedCrossRef Maira, S. M., et al. (2008). Identification and characterization of NVP-BEZ235, a new orally available dual phosphatidylinositol 3-kinase/mammalian target of rapamycin inhibitor with potent in vivo antitumor activity. Molecular Cancer Therapeutics, 7, 1851–1863.PubMedCrossRef
15.
Zurück zum Zitat Fraedrich, K., et al. (2012). Targeting aurora kinases with danusertib (PHA-739358) inhibits growth of liver metastases from gastroenteropancreatic neuroendocrine tumors in an orthotopic xenograft model. Clinical Cancer Research, 18, 4621–4632.PubMedCrossRef Fraedrich, K., et al. (2012). Targeting aurora kinases with danusertib (PHA-739358) inhibits growth of liver metastases from gastroenteropancreatic neuroendocrine tumors in an orthotopic xenograft model. Clinical Cancer Research, 18, 4621–4632.PubMedCrossRef
16.
Zurück zum Zitat Fendrich, V., et al. (2011). Hedgehog inhibition with cyclopamine represses tumor growth and prolongs survival in a transgenic mouse model of islet cell tumors. Annals of Surgery, 253, 546–552.PubMedCrossRef Fendrich, V., et al. (2011). Hedgehog inhibition with cyclopamine represses tumor growth and prolongs survival in a transgenic mouse model of islet cell tumors. Annals of Surgery, 253, 546–552.PubMedCrossRef
17.
Zurück zum Zitat Waser, B., et al. (2012). Glucose- dependent insulinotropic polypeptide receptors in most gastroenteropancreatic and bronchial neuroendocrine tumors. Journal of Clinical Endocrinology and Metabolism, 97, 482–488.PubMedCrossRef Waser, B., et al. (2012). Glucose- dependent insulinotropic polypeptide receptors in most gastroenteropancreatic and bronchial neuroendocrine tumors. Journal of Clinical Endocrinology and Metabolism, 97, 482–488.PubMedCrossRef
18.
Zurück zum Zitat O’Dorisio, B., et al. (2012) Relief of bowel-related symptoms with telotristat etiprate in octreotide refractory carcinoid syndrome: Preliminary results of a double-blind, placebo-controlled multicenter study. Journal Clinical Oncology, 30, suppl; abstr 4085. O’Dorisio, B., et al. (2012) Relief of bowel-related symptoms with telotristat etiprate in octreotide refractory carcinoid syndrome: Preliminary results of a double-blind, placebo-controlled multicenter study. Journal Clinical Oncology, 30, suppl; abstr 4085.
19.
Zurück zum Zitat Sennino, B., et al. (2012). Suppression of tumor invasion and metastasis by concurrent inhibition of c-Met and VEGF signaling in pancreatic neuroendocrine tumors. Cancer Discovery, 2, 270–287.PubMedCentralPubMedCrossRef Sennino, B., et al. (2012). Suppression of tumor invasion and metastasis by concurrent inhibition of c-Met and VEGF signaling in pancreatic neuroendocrine tumors. Cancer Discovery, 2, 270–287.PubMedCentralPubMedCrossRef
20.
Zurück zum Zitat Yap, T. A., et al. (2008). Targeting the PI3K-AKT-mTOR pathway: progress, pitfalls, and promises. Current Opinion in Pharmacology, 8, 393–412.PubMedCrossRef Yap, T. A., et al. (2008). Targeting the PI3K-AKT-mTOR pathway: progress, pitfalls, and promises. Current Opinion in Pharmacology, 8, 393–412.PubMedCrossRef
21.
Zurück zum Zitat Serra, V., et al. (2008). NVP-BEZ235, a dual PI3K/mTOR inhibitor, prevents PI3K signaling and inhibits the growth of cancer cells with activating PI3K mutations. Cancer Research, 68, 8022–8030.PubMedCrossRef Serra, V., et al. (2008). NVP-BEZ235, a dual PI3K/mTOR inhibitor, prevents PI3K signaling and inhibits the growth of cancer cells with activating PI3K mutations. Cancer Research, 68, 8022–8030.PubMedCrossRef
22.
Zurück zum Zitat Schnell, C. R., et al. (2008). Effects of the dual phosphatidylinositol 3-kinase/mammalian target of rapamycin inhibitor NVP-BEZ235 on the tumor vasculature: implications for clinical imaging. Cancer Research, 68, 6598–6607.PubMedCrossRef Schnell, C. R., et al. (2008). Effects of the dual phosphatidylinositol 3-kinase/mammalian target of rapamycin inhibitor NVP-BEZ235 on the tumor vasculature: implications for clinical imaging. Cancer Research, 68, 6598–6607.PubMedCrossRef
23.
Zurück zum Zitat Pavel, M. (2013). Translation of molecular pathways into clinical trials of neuroendocrine tumors. Neuroendocrinology, 97(1), 99–112.PubMedCrossRef Pavel, M. (2013). Translation of molecular pathways into clinical trials of neuroendocrine tumors. Neuroendocrinology, 97(1), 99–112.PubMedCrossRef
24.
Zurück zum Zitat Chiu, C. W., Nozawa, H., & Hanahan, D. (2010). Survival benefit with proapoptotic molecular and pathologic responses from dual targeting of mammalian target of rapamycin and epidermal growth factor receptor in a preclinical model of pancreatic neuroendocrine carcinogenesis. Journal of Clinical Oncology, 28, 4425–4433.PubMedCentralPubMedCrossRef Chiu, C. W., Nozawa, H., & Hanahan, D. (2010). Survival benefit with proapoptotic molecular and pathologic responses from dual targeting of mammalian target of rapamycin and epidermal growth factor receptor in a preclinical model of pancreatic neuroendocrine carcinogenesis. Journal of Clinical Oncology, 28, 4425–4433.PubMedCentralPubMedCrossRef
25.
Zurück zum Zitat Niraula, S., et al. (2012). The price we pay for progress: a meta-analysis of harms of newly approved anticancer drugs. Journal of Clinical Oncology, 30, 3012–3019.PubMedCrossRef Niraula, S., et al. (2012). The price we pay for progress: a meta-analysis of harms of newly approved anticancer drugs. Journal of Clinical Oncology, 30, 3012–3019.PubMedCrossRef
26.
Zurück zum Zitat Schutz, F. A., Je, Y., Richards, C. J., & Choueiri, T. K. (2012). Meta-analysis of randomized controlled trials for the incidence and risk of treatment-related mortality in patients with cancer treated with vascular endothelial growth factor tyrosine kinase inhibitors. Journal of Clinical Oncology, 30, 871–877.PubMedCrossRef Schutz, F. A., Je, Y., Richards, C. J., & Choueiri, T. K. (2012). Meta-analysis of randomized controlled trials for the incidence and risk of treatment-related mortality in patients with cancer treated with vascular endothelial growth factor tyrosine kinase inhibitors. Journal of Clinical Oncology, 30, 871–877.PubMedCrossRef
Metadaten
Titel
Systemic therapeutic strategies for GEP-NETS: what can we expect in the future?
verfasst von
E. Raymond
R. García-Carbonero
B. Wiedenmann
E. Grande
M. Pavel
Publikationsdatum
01.03.2014
Verlag
Springer US
Erschienen in
Cancer and Metastasis Reviews / Ausgabe 1/2014
Print ISSN: 0167-7659
Elektronische ISSN: 1573-7233
DOI
https://doi.org/10.1007/s10555-013-9467-z

Weitere Artikel der Ausgabe 1/2014

Cancer and Metastasis Reviews 1/2014 Zur Ausgabe

Update Onkologie

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