Skip to main content
Top
Published in: Investigational New Drugs 5/2016

01-10-2016 | SHORT REPORT

Prediction of response to everolimus in neuroendocrine tumors: evaluation of clinical, biological and histological factors

Authors: Noura Benslama, Julien Bollard, Cécile Vercherat, Patrick Massoma, Colette Roche, Valérie Hervieu, Julien Peron, Catherine Lombard-Bohas, Jean-Yves Scoazec, Thomas Walter

Published in: Investigational New Drugs | Issue 5/2016

Login to get access

Summary

Objectives Several targeted therapies are available for metastatic neuroendocrine tumours (NETs) but no predictive factor of response to these treatments has been identified yet. Our aim was to identify and evaluate clinical, biological, histological and functional markers of response to everolimus. Methods We retrospectively reviewed 53 patients with NETs treated with everolimus (68 % in clinical trials). Clinical, biological and histological data were analyzed. The functional marker p-p70S6K, a main effector of the mTOR pathway, was studied by immunohistochemistry in 43 cases. Prognostic factors of progression-free survival (PFS) were studied by Kaplan Meier analysis. Results All patients had metastatic and progressive disease before everolimus treatment. Objective response was 9 % and median PFS was 8.1 (4.7–11.5) months. Hypercholesterolemia (HR = 0.13, p < 0.0001) was associated with longer PFS, whereas presence of bone metastases (HR = 3.1, p < 0.001) and overexpression of p-p70S6K by tumor cells (HR = 2.5, p = 0.01) were associated with shorter PFS under everolimus at multivariate analysis. Conclusion Clinical markers are not useful to predict response to everolimus. However, occurrence of hypercholesterolemia under treatment may be an early marker of response. Prospective studies are required to confirm these results and to assess whether p-p70S6K immunostaining is a prognostic or predictive marker of no-response to everolimus.
Literature
1.
go back to reference Yao JC, Hassan M, Phan A, et al. (2008) One hundred years after “carcinoid”: epidemiology of and prognostic factors for neuroendocrine tumors in 35, 825 cases in the United States. J Clin Oncol Off J Am Soc Clin Oncol 26:3063–3072CrossRef Yao JC, Hassan M, Phan A, et al. (2008) One hundred years after “carcinoid”: epidemiology of and prognostic factors for neuroendocrine tumors in 35, 825 cases in the United States. J Clin Oncol Off J Am Soc Clin Oncol 26:3063–3072CrossRef
2.
go back to reference Pavel ME, Hainsworth JD, Baudin E, et al. (2011) Everolimus plus octreotide long-acting repeatable for the treatment of advanced neuroendocrine tumours associated with carcinoid syndrome (RADIANT-2): a randomised, placebo-controlled, phase 3 study. Lancet 378:2005–2012CrossRefPubMed Pavel ME, Hainsworth JD, Baudin E, et al. (2011) Everolimus plus octreotide long-acting repeatable for the treatment of advanced neuroendocrine tumours associated with carcinoid syndrome (RADIANT-2): a randomised, placebo-controlled, phase 3 study. Lancet 378:2005–2012CrossRefPubMed
3.
go back to reference Yao JC, Fazio N, Singh S et al. (2016) 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 387(10022):968–77. doi: 10.1016/S0140-6736(15)00817-X. Yao JC, Fazio N, Singh S et al. (2016) 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 387(10022):968–77. doi: 10.​1016/​S0140-6736(15)00817-X.
5.
go back to reference Galanis E, Buckner JC, Maurer MJ, et al. (2005) Phase II trial of temsirolimus (CCI-779) in recurrent glioblastoma multiforme: a north central cancer treatment group study. J Clin Oncol Off J Am Soc Clin Oncol 23:5294–5304CrossRef Galanis E, Buckner JC, Maurer MJ, et al. (2005) Phase II trial of temsirolimus (CCI-779) in recurrent glioblastoma multiforme: a north central cancer treatment group study. J Clin Oncol Off J Am Soc Clin Oncol 23:5294–5304CrossRef
6.
go back to reference Lee CK, Marschner IC, Simes RJ, et al. (2012) Increase in cholesterol predicts survival advantage in renal cell carcinoma patients treated with temsirolimus. Clin Cancer Res: An Official Journal of the American Association for Cancer Research 18:3188–3196CrossRef Lee CK, Marschner IC, Simes RJ, et al. (2012) Increase in cholesterol predicts survival advantage in renal cell carcinoma patients treated with temsirolimus. Clin Cancer Res: An Official Journal of the American Association for Cancer Research 18:3188–3196CrossRef
7.
go back to reference Ravaud A, Urva SR, Grosch K, et al. (2014) Relationship between everolimus exposure and safety and efficacy: meta-analysis of clinical trials in oncology. Eur J Cancer 50:486–495CrossRefPubMed Ravaud A, Urva SR, Grosch K, et al. (2014) Relationship between everolimus exposure and safety and efficacy: meta-analysis of clinical trials in oncology. Eur J Cancer 50:486–495CrossRefPubMed
8.
go back to reference Neshat MS, Mellinghoff IK, Tran C, et al. (2001) Enhanced sensitivity of PTEN-deficient tumors to inhibition of FRAP/mTOR. Proc Natl Acad Sci U S A 98:10314–10319CrossRefPubMedPubMedCentral Neshat MS, Mellinghoff IK, Tran C, et al. (2001) Enhanced sensitivity of PTEN-deficient tumors to inhibition of FRAP/mTOR. Proc Natl Acad Sci U S A 98:10314–10319CrossRefPubMedPubMedCentral
9.
go back to reference Janku F, Wheler JJ, Westin SN, et al. (2012) PI3K/AKT/mTOR inhibitors in patients with breast and gynecologic malignancies harboring PIK3CA mutations. J Clin Oncol Off J Am Soc Clin Oncol 30:777–782CrossRef Janku F, Wheler JJ, Westin SN, et al. (2012) PI3K/AKT/mTOR inhibitors in patients with breast and gynecologic malignancies harboring PIK3CA mutations. J Clin Oncol Off J Am Soc Clin Oncol 30:777–782CrossRef
11.
go back to reference Bollard J, Couderc C, Blanc M, et al. (2013) Antitumor effect of everolimus in preclinical models of high-grade gastroenteropancreatic neuroendocrine carcinomas. Neuroendocrinology 97:331–340CrossRefPubMed Bollard J, Couderc C, Blanc M, et al. (2013) Antitumor effect of everolimus in preclinical models of high-grade gastroenteropancreatic neuroendocrine carcinomas. Neuroendocrinology 97:331–340CrossRefPubMed
12.
go back to reference Cho D, Signoretti S, Dabora S, et al. (2007) Potential histologic and molecular predictors of response to temsirolimus in patients with advanced renal cell carcinoma. Clin Genitourin Cancer 5:379–385CrossRefPubMed Cho D, Signoretti S, Dabora S, et al. (2007) Potential histologic and molecular predictors of response to temsirolimus in patients with advanced renal cell carcinoma. Clin Genitourin Cancer 5:379–385CrossRefPubMed
13.
go back to reference Duran I, Kortmansky J, Singh D, et al. (2006) A phase II clinical and pharmacodynamic study of temsirolimus in advanced neuroendocrine carcinomas. Br J Cancer 95:1148–1154CrossRefPubMedPubMedCentral Duran I, Kortmansky J, Singh D, et al. (2006) A phase II clinical and pharmacodynamic study of temsirolimus in advanced neuroendocrine carcinomas. Br J Cancer 95:1148–1154CrossRefPubMedPubMedCentral
14.
go back to reference Yoon DH, Ryu MH, Park YS, et al. (2012) Phase II study of everolimus with biomarker exploration in patients with advanced gastric cancer refractory to chemotherapy including fluoropyrimidine and platinum. Br J Cancer 106:1039–1044CrossRefPubMedPubMedCentral Yoon DH, Ryu MH, Park YS, et al. (2012) Phase II study of everolimus with biomarker exploration in patients with advanced gastric cancer refractory to chemotherapy including fluoropyrimidine and platinum. Br J Cancer 106:1039–1044CrossRefPubMedPubMedCentral
15.
go back to reference Lombard-Bohas C, Cariou B, Verges B, et al. (2014) Management of metabolic disorders induced by everolimus in patients with differentiated neuroendocrine tumors: expert proposals. Bull Cancer 101:175–183PubMed Lombard-Bohas C, Cariou B, Verges B, et al. (2014) Management of metabolic disorders induced by everolimus in patients with differentiated neuroendocrine tumors: expert proposals. Bull Cancer 101:175–183PubMed
16.
go back to reference Verges B, Walter T, Cariou B (2014) ENDOCRINE SIDE EFFECTS OF ANTI-CANCER DRUGS effects of anti-cancer targeted therapies on lipid and glucose metabolism. Eur J Endocrinol 170:R43–R55CrossRefPubMed Verges B, Walter T, Cariou B (2014) ENDOCRINE SIDE EFFECTS OF ANTI-CANCER DRUGS effects of anti-cancer targeted therapies on lipid and glucose metabolism. Eur J Endocrinol 170:R43–R55CrossRefPubMed
17.
go back to reference Bilimoria KY, Talamonti MS, Tomlinson JS, et al. (2008) Prognostic score predicting survival after resection of pancreatic neuroendocrine tumors: analysis of 3851 patients. Ann Surg 247:490–500CrossRefPubMed Bilimoria KY, Talamonti MS, Tomlinson JS, et al. (2008) Prognostic score predicting survival after resection of pancreatic neuroendocrine tumors: analysis of 3851 patients. Ann Surg 247:490–500CrossRefPubMed
18.
go back to reference Khan MS, Kirkwood A, Tsigani T, et al. (2013) Circulating tumor cells as prognostic markers in neuroendocrine tumors. J Clin Oncol Off J Am Soc Clin Oncol 31:365–372CrossRef Khan MS, Kirkwood A, Tsigani T, et al. (2013) Circulating tumor cells as prognostic markers in neuroendocrine tumors. J Clin Oncol Off J Am Soc Clin Oncol 31:365–372CrossRef
19.
go back to reference Capurso G, Archibugi L, Delle Fave G (2015) Molecular pathogenesis and targeted therapy of sporadic pancreatic neuroendocrine tumors. J Hepatobiliary Pancreat Sci 22(8):594–601. doi:10.1002/jhbp.210. Capurso G, Archibugi L, Delle Fave G (2015) Molecular pathogenesis and targeted therapy of sporadic pancreatic neuroendocrine tumors. J Hepatobiliary Pancreat Sci 22(8):594–601. doi:10.​1002/​jhbp.​210.
20.
go back to reference Missiaglia E, Dalai I, Barbi S, et al. (2009) Pancreatic endocrine tumors: expression profiling evidences a role for AKT-mTOR pathway. J Clin Oncol Off J Am Soc Clin Oncol 28:245–255CrossRef Missiaglia E, Dalai I, Barbi S, et al. (2009) Pancreatic endocrine tumors: expression profiling evidences a role for AKT-mTOR pathway. J Clin Oncol Off J Am Soc Clin Oncol 28:245–255CrossRef
21.
go back to reference Qian ZR, Ter-Minassian M, Chan JA, et al. (2013) Prognostic significance of MTOR pathway component expression in neuroendocrine tumors. J Clin Oncol Off J Am Soc Clin Oncol 31:3418–3425CrossRef Qian ZR, Ter-Minassian M, Chan JA, et al. (2013) Prognostic significance of MTOR pathway component expression in neuroendocrine tumors. J Clin Oncol Off J Am Soc Clin Oncol 31:3418–3425CrossRef
22.
go back to reference Sharpe LJ, Brown AJ (2008) Rapamycin down-regulates LDL-receptor expression independently of SREBP-2. Biochem Biophys Res Commun 373:670–674CrossRefPubMed Sharpe LJ, Brown AJ (2008) Rapamycin down-regulates LDL-receptor expression independently of SREBP-2. Biochem Biophys Res Commun 373:670–674CrossRefPubMed
23.
go back to reference Custodio AJ-FP, Alonso-Orduña V, López López C, Alonso T, Guillermo C, Carmona-Bayonas A, Álvarez-Escolá CSM, Capdevila J, Grande E, Barriuso J, Feliu J, Aller J (2015) Everolimus (EVE)-induced hyperglycemia (HG) in patients (pts) with advanced G1-G2 neuroendocrine tumors (NETs): clinical relevance and predictive value. ENETS - The European Neuroendocrine Tumor Society. Barcelona, In Custodio AJ-FP, Alonso-Orduña V, López López C, Alonso T, Guillermo C, Carmona-Bayonas A, Álvarez-Escolá CSM, Capdevila J, Grande E, Barriuso J, Feliu J, Aller J (2015) Everolimus (EVE)-induced hyperglycemia (HG) in patients (pts) with advanced G1-G2 neuroendocrine tumors (NETs): clinical relevance and predictive value. ENETS - The European Neuroendocrine Tumor Society. Barcelona, In
24.
go back to reference Rugo HS, Hortobagyi GN, Yao J, Pavel M, Ravaud A, Franz D, Ringeisen F, Gallo J, Rouyrre N, Anak O, Motzer R (2016) Meta-analysis of stomatitis in clinical studies of everolimus: incidence and relationship with efficacy. Ann Oncol 27(3):519–525. doi:10.1093/annonc/mdv595. Rugo HS, Hortobagyi GN, Yao J, Pavel M, Ravaud A, Franz D, Ringeisen F, Gallo J, Rouyrre N, Anak O, Motzer R (2016) Meta-analysis of stomatitis in clinical studies of everolimus: incidence and relationship with efficacy. Ann Oncol 27(3):519–525. doi:10.​1093/​annonc/​mdv595.
25.
go back to reference Templeton AJ, Dutoit V, Cathomas R, et al. (2013) Phase 2 trial of single-agent everolimus in chemotherapy-naive patients with castration-resistant prostate cancer (SAKK 08/08). Eur Urol 64:150–158CrossRefPubMed Templeton AJ, Dutoit V, Cathomas R, et al. (2013) Phase 2 trial of single-agent everolimus in chemotherapy-naive patients with castration-resistant prostate cancer (SAKK 08/08). Eur Urol 64:150–158CrossRefPubMed
Metadata
Title
Prediction of response to everolimus in neuroendocrine tumors: evaluation of clinical, biological and histological factors
Authors
Noura Benslama
Julien Bollard
Cécile Vercherat
Patrick Massoma
Colette Roche
Valérie Hervieu
Julien Peron
Catherine Lombard-Bohas
Jean-Yves Scoazec
Thomas Walter
Publication date
01-10-2016
Publisher
Springer US
Published in
Investigational New Drugs / Issue 5/2016
Print ISSN: 0167-6997
Electronic ISSN: 1573-0646
DOI
https://doi.org/10.1007/s10637-016-0363-6

Other articles of this Issue 5/2016

Investigational New Drugs 5/2016 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