Skip to main content
Top
Published in: Targeted Oncology 5/2017

01-10-2017 | Review Article

Predictive Markers of Response to Everolimus and Sunitinib in Neuroendocrine Tumors

Authors: Diana Martins, Francesca Spada, Ioana Lambrescu, Manila Rubino, Chiara Cella, Bianca Gibelli, Chiara Grana, Dario Ribero, Emilio Bertani, Davide Ravizza, Guido Bonomo, Luigi Funicelli, Eleonora Pisa, Dario Zerini, Nicola Fazio, IEO ENETS Center of Excellence for GEP NETs

Published in: Targeted Oncology | Issue 5/2017

Login to get access

Abstract

Neuroendocrine tumors (NETs) represent a large and heterogeneous group of malignancies with various biological and clinical characteristics, depending on the site of origin and the grade of tumor proliferation. In NETs, as in other cancer types, molecularly targeted therapies have radically changed the therapeutic landscape. Recently two targeted agents, the mammalian target of rapamycin inhibitor everolimus and the tyrosine kinase inhibitor sunitinib, have both demonstrated significantly prolonged progression free survival in patients with advanced pancreatic NETs. Despite these important therapeutic developments, there are still significant limitations to the use of these agents due to the lack of accurate biomarkers for predicting tumor response and efficacy of therapy. In this review, we provide an overview of the current clinical data for the evaluation of predictive factors of response to/efficacy of everolimus and sunitinib in advanced pancreatic NETs. Surrogate indicators discussed include circulating and tissue markers, as well as non-invasive imaging techniques.
Literature
1.
go back to reference van der Zwan JM, Trama A, Otter R, et al. Rare neuroendocrine tumours: results of the surveillance of rare cancers in Europe project. Eur J Cancer. 2013;49:2565–78.CrossRefPubMed van der Zwan JM, Trama A, Otter R, et al. Rare neuroendocrine tumours: results of the surveillance of rare cancers in Europe project. Eur J Cancer. 2013;49:2565–78.CrossRefPubMed
2.
go back to reference Bosman FT, Carneiro F, Hruban RH et al. WHO classification of tumours of the digestive system. 4th ed. World Health Organization; 2010. Bosman FT, Carneiro F, Hruban RH et al. WHO classification of tumours of the digestive system. 4th ed. World Health Organization; 2010.
4.
go back to reference Raymond E, Hammel P, Dreyer C. Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. N Engl J Med. 2011;364:501–13.CrossRefPubMed Raymond E, Hammel P, Dreyer C. Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. N Engl J Med. 2011;364:501–13.CrossRefPubMed
5.
go back to reference Yao JC, Fazio N, Singh S, et al. RAD001 in advanced neuroendocrine Tumours, fourth trial (RADIANT-4) study group. Lancet. 2016;387:968–77.CrossRefPubMed Yao JC, Fazio N, Singh S, et al. RAD001 in advanced neuroendocrine Tumours, fourth trial (RADIANT-4) study group. Lancet. 2016;387:968–77.CrossRefPubMed
6.
go back to reference Yao JC, Pavel M, Phan AT, et al. Chromogranin a and neuron-specific enolase as prognostic markers in patients with advanced pNET treated with Everolimus. J Clin Endocrinol Metab. 2011;96:3741–9.CrossRefPubMed Yao JC, Pavel M, Phan AT, et al. Chromogranin a and neuron-specific enolase as prognostic markers in patients with advanced pNET treated with Everolimus. J Clin Endocrinol Metab. 2011;96:3741–9.CrossRefPubMed
7.
go back to reference Baudin E, Wolin E, Castellano D. Correlation of PFS with early response of chromogranin a and 5-hydroxyindoleacetic acid levels in patients with advanced neuroendocrine tumors: phase III RADIANT-2 study results. Eur J Cancer. 2011;47(Suppl 1):S460.CrossRef Baudin E, Wolin E, Castellano D. Correlation of PFS with early response of chromogranin a and 5-hydroxyindoleacetic acid levels in patients with advanced neuroendocrine tumors: phase III RADIANT-2 study results. Eur J Cancer. 2011;47(Suppl 1):S460.CrossRef
8.
go back to reference Yao JC, Shah M, Panneerselvam A, et al. The VEGF pathway in patients with pancreatic neuroendocrine tumors: efficacy of everolimus by baseline marker level, and prognostic and predictive effect analyses from RADIANT-3. Ann Oncol. 2012;23(Suppl 9):376.CrossRef Yao JC, Shah M, Panneerselvam A, et al. The VEGF pathway in patients with pancreatic neuroendocrine tumors: efficacy of everolimus by baseline marker level, and prognostic and predictive effect analyses from RADIANT-3. Ann Oncol. 2012;23(Suppl 9):376.CrossRef
9.
go back to reference Bellister SA, Zhou Y, Sceusi E, et al. Prediction of prognosis in patients treated with everolimus for extrapancreatic neuroendocrine tumors by a single nucleotide polymorphism in PHLPP2. J Clin Oncol. 2013;31(Suppl 4):163.CrossRef Bellister SA, Zhou Y, Sceusi E, et al. Prediction of prognosis in patients treated with everolimus for extrapancreatic neuroendocrine tumors by a single nucleotide polymorphism in PHLPP2. J Clin Oncol. 2013;31(Suppl 4):163.CrossRef
10.
go back to reference Serra S, Zheng L, Hassan M, et al. The FGFR4-G388R single-nucleotide polymorphism alters pancreatic neuroendocrine tumor progression and response to mTOR inhibition therapy. Cancer Res. 2012;72:5683–91.CrossRefPubMed Serra S, Zheng L, Hassan M, et al. The FGFR4-G388R single-nucleotide polymorphism alters pancreatic neuroendocrine tumor progression and response to mTOR inhibition therapy. Cancer Res. 2012;72:5683–91.CrossRefPubMed
11.
go back to reference Meric-Bernstam F, Akcakanat A, Chen H, et al. PIK3CA/PTEN mutations and Akt activation as markers of sensitivity to allosteric mTOR inhibitor. Clin Cancer Res. 2012;18:1777–89.CrossRefPubMedPubMedCentral Meric-Bernstam F, Akcakanat A, Chen H, et al. PIK3CA/PTEN mutations and Akt activation as markers of sensitivity to allosteric mTOR inhibitor. Clin Cancer Res. 2012;18:1777–89.CrossRefPubMedPubMedCentral
12.
go back to reference Spada F, Fazio N, Capurso G, et al. PI3K-AKT-mTOR pathway disregulation and its correlation with clinical outcome in patients with advanced neuroendocrine tumors treated with everolimus. Pancreas. 2014;43:493. Spada F, Fazio N, Capurso G, et al. PI3K-AKT-mTOR pathway disregulation and its correlation with clinical outcome in patients with advanced neuroendocrine tumors treated with everolimus. Pancreas. 2014;43:493.
13.
go back to reference Gagliano T, Bellio M, Gentilin E, et al. mTOR, p70S6K, AKT, and ERK1/2 levels predict sensitivity to mTOR and PI3K/mTOR inhibitors in human bronchial carcinoids. Endocr Relat Cancer. 2013;20:463–75.CrossRefPubMed Gagliano T, Bellio M, Gentilin E, et al. mTOR, p70S6K, AKT, and ERK1/2 levels predict sensitivity to mTOR and PI3K/mTOR inhibitors in human bronchial carcinoids. Endocr Relat Cancer. 2013;20:463–75.CrossRefPubMed
14.
go back to reference Casanovas O, Capdevila J, Barriuso J. Potential role of mTOR phosphorylation status as a negative predictor to everolimus plus octreotide in NETs. J Clin Oncol. 2014;32(Suppl 3):484.CrossRef Casanovas O, Capdevila J, Barriuso J. Potential role of mTOR phosphorylation status as a negative predictor to everolimus plus octreotide in NETs. J Clin Oncol. 2014;32(Suppl 3):484.CrossRef
15.
go back to reference Cros J, Moati E, Raffenne J, et al. Gly388Arg FGFR4 polymorphism is not predictive of Everolimus efficacy in well-differentiated digestive neuroendocrine tumors. Neuroendocrinology. 2015;103:495–9.CrossRefPubMed Cros J, Moati E, Raffenne J, et al. Gly388Arg FGFR4 polymorphism is not predictive of Everolimus efficacy in well-differentiated digestive neuroendocrine tumors. Neuroendocrinology. 2015;103:495–9.CrossRefPubMed
16.
go back to reference Benslama N, Bollard J, Vercherat C, et al. Prediction of response to everolimus in neuroendocrine tumors: evaluation of clinical, biological and histological factors. Investig New Drugs. 2016;34:654–62.CrossRef Benslama N, Bollard J, Vercherat C, et al. Prediction of response to everolimus in neuroendocrine tumors: evaluation of clinical, biological and histological factors. Investig New Drugs. 2016;34:654–62.CrossRef
17.
go back to reference Falletta S, Partelli S. Rubini et al. mTOR inhibitors response and mTOR pathway in pancreatic neuroendocrine tumors. Endocr Relat Cancer. 2016;23:883–91.CrossRefPubMed Falletta S, Partelli S. Rubini et al. mTOR inhibitors response and mTOR pathway in pancreatic neuroendocrine tumors. Endocr Relat Cancer. 2016;23:883–91.CrossRefPubMed
18.
go back to reference van Asselt SJ, Oosting SF, Brouwers AH, et al. Everolimus reduces (89)Zr bevacizumab tumor uptake in patients with neuroendocrine tumors. J Nucl Med. 2014;55:1087–92.CrossRefPubMed van Asselt SJ, Oosting SF, Brouwers AH, et al. Everolimus reduces (89)Zr bevacizumab tumor uptake in patients with neuroendocrine tumors. J Nucl Med. 2014;55:1087–92.CrossRefPubMed
19.
go back to reference Yao JC, Phan AT, Hess K, et al. Perfusion computed tomography as functional biomarker in randomized run-in study of bevacizumab and everolimus in well-differentiated neuroendocrine tumors. Pancreas. 2015;44:190–7.CrossRefPubMed Yao JC, Phan AT, Hess K, et al. Perfusion computed tomography as functional biomarker in randomized run-in study of bevacizumab and everolimus in well-differentiated neuroendocrine tumors. Pancreas. 2015;44:190–7.CrossRefPubMed
20.
go back to reference Bello CL, DePrimo SE, Friece C, et al. Analysis of circulating biomarkers of sunitinib malate in patients with unresectable neuroendocrine tumors (NET): VEGF, IL-8, and soluble VEGF receptors 2 and 3. J Clin Oncol. 2006;24(Suppl 18):4045. Bello CL, DePrimo SE, Friece C, et al. Analysis of circulating biomarkers of sunitinib malate in patients with unresectable neuroendocrine tumors (NET): VEGF, IL-8, and soluble VEGF receptors 2 and 3. J Clin Oncol. 2006;24(Suppl 18):4045.
21.
go back to reference Zurita AJ, Khajavi M, Wu HK, et al. Circulating cytokines and monocyte subpopulations as biomarkers of outcome and biological activity in sunitinib-treated patients with advanced neuroendocrine tumours. Br J Cancer. 2015;112:1199–205.CrossRefPubMedPubMedCentral Zurita AJ, Khajavi M, Wu HK, et al. Circulating cytokines and monocyte subpopulations as biomarkers of outcome and biological activity in sunitinib-treated patients with advanced neuroendocrine tumours. Br J Cancer. 2015;112:1199–205.CrossRefPubMedPubMedCentral
22.
go back to reference Dreyer C, Couvelard A, Walter T, et al. Clinical and biomarker evaluations of sunitinib in patients (pts) with advanced well-differentiated grade 3 (G3) and poorly differentiated neuroendocrine neoplasms (PD-NEN). J Clin Oncol. 2016;34(Suppl 4):274.CrossRef Dreyer C, Couvelard A, Walter T, et al. Clinical and biomarker evaluations of sunitinib in patients (pts) with advanced well-differentiated grade 3 (G3) and poorly differentiated neuroendocrine neoplasms (PD-NEN). J Clin Oncol. 2016;34(Suppl 4):274.CrossRef
23.
go back to reference Faivre S, Ronot M, Dreyer C, et al. Imaging response in neuroendocrine tumors treated with targeted therapies: the experience of sunitinib. Target Oncol. 2012;7:127–33.CrossRefPubMed Faivre S, Ronot M, Dreyer C, et al. Imaging response in neuroendocrine tumors treated with targeted therapies: the experience of sunitinib. Target Oncol. 2012;7:127–33.CrossRefPubMed
24.
go back to reference Oberg K, Krenning E, Sundin A, et al. A Delphic consensus assessment: imaging and biomarkers in gastroenteropancreatic neuroendocrine tumor disease management. Endocr Connect. 2016;5:174–87.CrossRefPubMedPubMedCentral Oberg K, Krenning E, Sundin A, et al. A Delphic consensus assessment: imaging and biomarkers in gastroenteropancreatic neuroendocrine tumor disease management. Endocr Connect. 2016;5:174–87.CrossRefPubMedPubMedCentral
25.
go back to reference O’Reilly KE, Rojo F, She QB, et al. mTOR inhibition induces upstream receptor tyrosine kinase signaling and activates Akt. Cancer Res. 2006;66:1500–8.CrossRefPubMedPubMedCentral O’Reilly KE, Rojo F, She QB, et al. mTOR inhibition induces upstream receptor tyrosine kinase signaling and activates Akt. Cancer Res. 2006;66:1500–8.CrossRefPubMedPubMedCentral
26.
go back to reference Yao JC, Lombard-Bohas C, Baudin E, et al. Daily oral everolimus activity in patients with metastatic pancreatic neuroendocrine tumors after failure of cytotoxic chemotherapy: a phase II trial. J Clin Oncol. 2010;28:69–76.CrossRefPubMed Yao JC, Lombard-Bohas C, Baudin E, et al. Daily oral everolimus activity in patients with metastatic pancreatic neuroendocrine tumors after failure of cytotoxic chemotherapy: a phase II trial. J Clin Oncol. 2010;28:69–76.CrossRefPubMed
27.
go back to reference Yao JC. Neuroendocrine tumors molecular targeted therapy for carcinoid and islet-cell carcinoma. Best Pract Res Clin Endocrinol Metab. 2007;21:163–72.CrossRefPubMed Yao JC. Neuroendocrine tumors molecular targeted therapy for carcinoid and islet-cell carcinoma. Best Pract Res Clin Endocrinol Metab. 2007;21:163–72.CrossRefPubMed
28.
go back to reference Missiaglia E, Dalai I, Barbi S, et al. Pancreatic endocrine tumors: expression profiling evidences a role for AKT-Mtor pathway. J Clin Oncol. 2010;28:245–55.CrossRefPubMed Missiaglia E, Dalai I, Barbi S, et al. Pancreatic endocrine tumors: expression profiling evidences a role for AKT-Mtor pathway. J Clin Oncol. 2010;28:245–55.CrossRefPubMed
29.
30.
go back to reference Abraham RT, Eng CH. Mammalian target of rapamycin as a therapeutic target in oncology. Expert Opin Ther Targets. 2008;12:209–22.CrossRefPubMed Abraham RT, Eng CH. Mammalian target of rapamycin as a therapeutic target in oncology. Expert Opin Ther Targets. 2008;12:209–22.CrossRefPubMed
31.
go back to reference Zatelli MC, Fanciulli G, Malandrino P, et al. Predictive factors of response to mTOR inhibitors in neuroendocrine tumours. Endocr Relat Cancer. 2016;23:173–83.CrossRef Zatelli MC, Fanciulli G, Malandrino P, et al. Predictive factors of response to mTOR inhibitors in neuroendocrine tumours. Endocr Relat Cancer. 2016;23:173–83.CrossRef
32.
go back to reference Fazio N. Neuroendocrine tumors resistant to mammalian target of rapamycin inhibitors: a difficult conversion from biology to the clinic. World J Clin Oncol. 2015;6:194–7.CrossRefPubMedPubMedCentral Fazio N. Neuroendocrine tumors resistant to mammalian target of rapamycin inhibitors: a difficult conversion from biology to the clinic. World J Clin Oncol. 2015;6:194–7.CrossRefPubMedPubMedCentral
33.
go back to reference Modlin IM, Gustafsson BI, Moss SF, et al. Chromogranin A-biological function and clinical utility in neuroendocrine tumor disease. Ann Surg Oncol. 2010;17:2427–43.CrossRefPubMed Modlin IM, Gustafsson BI, Moss SF, et al. Chromogranin A-biological function and clinical utility in neuroendocrine tumor disease. Ann Surg Oncol. 2010;17:2427–43.CrossRefPubMed
34.
go back to reference Qian ZR, Ter-Minassian M, Chan JA, et al. Prognostic significance of MTOR pathway component expression in neuroendocrine tumors. J Clin Oncol. 2013;31:3418–25.CrossRefPubMedPubMedCentral Qian ZR, Ter-Minassian M, Chan JA, et al. Prognostic significance of MTOR pathway component expression in neuroendocrine tumors. J Clin Oncol. 2013;31:3418–25.CrossRefPubMedPubMedCentral
35.
go back to reference Capurso G, Archibugi L, Delle FG. Molecular pathogenesis and targeted therapy of sporadic pancreatic neuroendocrine tumors. J Hepatobiliary Pancreat Sci. 2015;22:594–601.CrossRefPubMed Capurso G, Archibugi L, Delle FG. Molecular pathogenesis and targeted therapy of sporadic pancreatic neuroendocrine tumors. J Hepatobiliary Pancreat Sci. 2015;22:594–601.CrossRefPubMed
36.
go back to reference Villaume K, Blanc M, Gouysse G, et al. VEGF secretion by neuroendocrine tumor cells is inhibited by octreotide and by inhibitors of the PI3K/AKT/mTOR pathway. Neuroendocrinology. 2010;91:268–78.CrossRefPubMed Villaume K, Blanc M, Gouysse G, et al. VEGF secretion by neuroendocrine tumor cells is inhibited by octreotide and by inhibitors of the PI3K/AKT/mTOR pathway. Neuroendocrinology. 2010;91:268–78.CrossRefPubMed
37.
go back to reference Yao JC, Phan A, Hoff PM, et al. Targeting vascular endothelial growth factor in advanced carcinoid tumor: a random assignment phase II study of depot octreotide with bevacizumab and pegylated interferon alpha-2b. J Clin Oncol. 2008;26:1316–23.CrossRefPubMed Yao JC, Phan A, Hoff PM, et al. Targeting vascular endothelial growth factor in advanced carcinoid tumor: a random assignment phase II study of depot octreotide with bevacizumab and pegylated interferon alpha-2b. J Clin Oncol. 2008;26:1316–23.CrossRefPubMed
38.
go back to reference Kulke MH, Chan JA, Meyerhardt JA, et al. A prospective phase II study of 2-methoxyestradiol administered in combination with bevacizumab in patients with metastatic carcinoid tumors. Cancer Chemother Pharmacol. 2011;68:293–300.CrossRefPubMed Kulke MH, Chan JA, Meyerhardt JA, et al. A prospective phase II study of 2-methoxyestradiol administered in combination with bevacizumab in patients with metastatic carcinoid tumors. Cancer Chemother Pharmacol. 2011;68:293–300.CrossRefPubMed
39.
go back to reference Chan JA, Stuart K, Earle CC, et al. Prospective study of bevacizumab plus temozolomide in patients with advanced neuroendocrine tumors. J Clin Oncol. 2012;30:2963–8.CrossRefPubMedPubMedCentral Chan JA, Stuart K, Earle CC, et al. Prospective study of bevacizumab plus temozolomide in patients with advanced neuroendocrine tumors. J Clin Oncol. 2012;30:2963–8.CrossRefPubMedPubMedCentral
40.
go back to reference Ng CS, Charnsangavej C, Wei W, et al. Perfusion CT findings in patients with metastatic carcinoid tumors undergoing bevacizumab and interferon therapy. AJR Am J Roentgenol. 2007;196:569–76.CrossRef Ng CS, Charnsangavej C, Wei W, et al. Perfusion CT findings in patients with metastatic carcinoid tumors undergoing bevacizumab and interferon therapy. AJR Am J Roentgenol. 2007;196:569–76.CrossRef
41.
go back to reference Faivre S, Djelloul S, Raymond E. New paradigms in anticancer therapy: targeting multiple signalling pathways with kinase inhibitors. Semin Oncol. 2006;33:407–20.CrossRefPubMed Faivre S, Djelloul S, Raymond E. New paradigms in anticancer therapy: targeting multiple signalling pathways with kinase inhibitors. Semin Oncol. 2006;33:407–20.CrossRefPubMed
42.
go back to reference Vinik AI, Raymond E. Pancreatic neuroendocrine tumors: approach to treatment with focus on sunitinib. Therap Adv Gastroenterol. 2013;5:396–411.CrossRef Vinik AI, Raymond E. Pancreatic neuroendocrine tumors: approach to treatment with focus on sunitinib. Therap Adv Gastroenterol. 2013;5:396–411.CrossRef
43.
go back to reference Raymond E, Kulke M, Qin S, et al. The efficacy and safety of sunitinib in patients with advanced well-differentiated pancreatic neuroendocrine tumors. Pancreas. 2016;46(Suppl 3):427–51. Raymond E, Kulke M, Qin S, et al. The efficacy and safety of sunitinib in patients with advanced well-differentiated pancreatic neuroendocrine tumors. Pancreas. 2016;46(Suppl 3):427–51.
44.
go back to reference DePrimo SE, Bello C. Surrogate biomarkers in evaluating response to anti-angiogenic agents: focus on sunitinib. Ann Oncol. 2007;18:11–9.CrossRef DePrimo SE, Bello C. Surrogate biomarkers in evaluating response to anti-angiogenic agents: focus on sunitinib. Ann Oncol. 2007;18:11–9.CrossRef
45.
go back to reference Gerger A, LaBonte M, Lenz HJ. Molecular predictors of response to antiangiogenesis therapies. Cancer J. 2011;17:134–41.CrossRefPubMed Gerger A, LaBonte M, Lenz HJ. Molecular predictors of response to antiangiogenesis therapies. Cancer J. 2011;17:134–41.CrossRefPubMed
46.
go back to reference Norden-Zfoni A, Desai J, Manola J, et al. Blood-based biomarkers of SU11248 activity and clinical outcome in patients with metastatic imatinib-resistant gastrointestinal stromal tumor. Clin Cancer Res. 2007;13:2643–50.CrossRefPubMed Norden-Zfoni A, Desai J, Manola J, et al. Blood-based biomarkers of SU11248 activity and clinical outcome in patients with metastatic imatinib-resistant gastrointestinal stromal tumor. Clin Cancer Res. 2007;13:2643–50.CrossRefPubMed
47.
go back to reference DePrimo SE, Bello CL, Smeraglia J, et al. Circulating protein biomarkers of pharmacodynamic activity of sunitinib in patients with metastatic renal cell carcinoma: modulation of VEGF and VEGF-related proteins. J Transl Med. 2007;5:32.CrossRefPubMedPubMedCentral DePrimo SE, Bello CL, Smeraglia J, et al. Circulating protein biomarkers of pharmacodynamic activity of sunitinib in patients with metastatic renal cell carcinoma: modulation of VEGF and VEGF-related proteins. J Transl Med. 2007;5:32.CrossRefPubMedPubMedCentral
48.
go back to reference Grande E, Casanovas O, Earl J, et al. sVEGFR2 and circulating tumor cells to predict for the efficacy of pazopanib in neuroendocrine tumors (NETs): the PAZONET subgroup analysis. J Clin Oncol. 2013;31(Suppl 15):4140. Grande E, Casanovas O, Earl J, et al. sVEGFR2 and circulating tumor cells to predict for the efficacy of pazopanib in neuroendocrine tumors (NETs): the PAZONET subgroup analysis. J Clin Oncol. 2013;31(Suppl 15):4140.
49.
go back to reference Takahashi Y, Akishima-Fukasawa Y, Kobayashi N, et al. Prognostic value of tumor architecture, tumor-associated vascular characteristics, and expression of angiogenic molecules in pancreatic endocrine tumors. Clin Cancer Res. 2007;13:187–96.CrossRefPubMed Takahashi Y, Akishima-Fukasawa Y, Kobayashi N, et al. Prognostic value of tumor architecture, tumor-associated vascular characteristics, and expression of angiogenic molecules in pancreatic endocrine tumors. Clin Cancer Res. 2007;13:187–96.CrossRefPubMed
50.
go back to reference You D, Song SH, Cho YM, et al. Predictive role of tissue-based molecular markers in patients treated with sunitinib for metastatic renal cell carcinoma. World J Urol. 2015;33:111–8.CrossRefPubMed You D, Song SH, Cho YM, et al. Predictive role of tissue-based molecular markers in patients treated with sunitinib for metastatic renal cell carcinoma. World J Urol. 2015;33:111–8.CrossRefPubMed
51.
go back to reference Choi H, Charnsangavej C, Faria SC, et al. Correlation of computed tomography and positron emission tomography in patients with metastatic gastrointestinal stromal tumor treated at a single institution with imatinib mesylate: proposal of new computed tomography response criteria. J Clin Oncol. 2007;25:1753–9.CrossRefPubMed Choi H, Charnsangavej C, Faria SC, et al. Correlation of computed tomography and positron emission tomography in patients with metastatic gastrointestinal stromal tumor treated at a single institution with imatinib mesylate: proposal of new computed tomography response criteria. J Clin Oncol. 2007;25:1753–9.CrossRefPubMed
52.
go back to reference Vercellino L, Bousquet G, Baillet G, et al. 18F-FDG PET/CT imaging for an early assessment of response to sunitinib in metastatic renal carcinoma: preliminary study. Cancer Biother Radiopharm. 2009;24:137–44.CrossRefPubMed Vercellino L, Bousquet G, Baillet G, et al. 18F-FDG PET/CT imaging for an early assessment of response to sunitinib in metastatic renal carcinoma: preliminary study. Cancer Biother Radiopharm. 2009;24:137–44.CrossRefPubMed
53.
go back to reference Mordente A, Meucci E, Martorana G, E et al. Cancer biomarkers discovery and validation: state of the art, problems and future perspectives. Adv Exp Med Biol. 2015;867:9–26. Mordente A, Meucci E, Martorana G, E et al. Cancer biomarkers discovery and validation: state of the art, problems and future perspectives. Adv Exp Med Biol. 2015;867:9–26.
54.
Metadata
Title
Predictive Markers of Response to Everolimus and Sunitinib in Neuroendocrine Tumors
Authors
Diana Martins
Francesca Spada
Ioana Lambrescu
Manila Rubino
Chiara Cella
Bianca Gibelli
Chiara Grana
Dario Ribero
Emilio Bertani
Davide Ravizza
Guido Bonomo
Luigi Funicelli
Eleonora Pisa
Dario Zerini
Nicola Fazio
IEO ENETS Center of Excellence for GEP NETs
Publication date
01-10-2017
Publisher
Springer International Publishing
Published in
Targeted Oncology / Issue 5/2017
Print ISSN: 1776-2596
Electronic ISSN: 1776-260X
DOI
https://doi.org/10.1007/s11523-017-0506-5

Other articles of this Issue 5/2017

Targeted Oncology 5/2017 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine