Determination of an optimal response cut-off able to predict progression-free survival in patients with well-differentiated advanced pancreatic neuroendocrine tumours treated with sunitinib: an alternative to the current RECIST-defined response

Angela Lamarca, Jorge Barriuso, Matthew Kulke, Ivan Borbath, Heinz-Josef Lenz, Jean Luc Raoul, Neal J Meropol, Catherine Lombard-Bohas, James Posey, Sandrine Faivre, Eric Raymond, Juan W Valle, Angela Lamarca, Jorge Barriuso, Matthew Kulke, Ivan Borbath, Heinz-Josef Lenz, Jean Luc Raoul, Neal J Meropol, Catherine Lombard-Bohas, James Posey, Sandrine Faivre, Eric Raymond, Juan W Valle

Abstract

Background: Sunitinib prolongs progression-free survival (PFS) in patients with advanced pancreatic neuroendocrine tumours (pNET). Response Evaluation Criteria in Solid Tumors (RECIST)-defined partial responses (PR; classically defined as ⩾30% size decrease from baseline) are infrequent.

Methods: Individual data of pNET patients from the phase II [NCT00056693] and pivotal phase III [NCT00428597] trials of sunitinib were analysed in this investigator-initiated, post hoc study. The primary objective was to determine the optimal RECIST (v.1.0) response cut-off value to identify patients who were progression-free at 11 months (median PFS in phase III trial); and the most informative time-point (highest area under the curve (AUC) by receiver operating characteristic (ROC) analysis and logistic regression) for prediction of benefit (PFS) from sunitinib.

Results: Data for 237 patients (85 placebo; 152 sunitinib (n=66.50 mg '4-weeks on/2-weeks off' schedule; n=86 '37.5 mg continuous daily dosing (CDD)')) and 788 scans were analysed. The median PFS for sunitinib and placebo were 9.3 months (95% CI 7.6-12.2) and 5.4 months (95% CI 3.5-6.01), respectively (hazard ratio (HR) 0.43 (95% CI 0.29-0.62); P<0.001). A PR was seen in 19 patients (13%) on sunitinib; the median change in the sum of the lesions (vs baseline) was -12.8% (range -100 to +36.4). Month 7 was the most informative time-point (AUC 0.78 (95% CI 0.66-0.9); odds ratio 1.05 (95% CI 1.01-1.08), P=0.002). Reduction of 10% (vs baseline) achieved the highest sensitivity (50%) and specificity (82%), amongst cut-offs tested. A 10% reduction in marker lesions was associated with improved PFS in the whole sunitinib population (HR 0.55 (95 CI 0.3-0.9); P=0.04); mostly in patients on sunitinib CDD (HR 0.33 (95% CI 0.2-0.7); P=0.005). A 10% reduction in marker lesions (P=0.034) and sunitinib treatment (P=0.012) independently impacted on PFS (multivariable analysis).

Conclusions: A 10% reduction within marker lesions identifies pNET patients benefiting from sunitinib treatment with implications for maintenance of dose intensity and future trial design.

Conflict of interest statement

Angela Lamarca has received Travel and Educational Grants from Pfizer; she has also participated in Speakers' Bureaus for Pfizer. Jorge Barriuso has received Travel Grants, and Research funding from Pfizer, he has also participated in Speakers' Bureaus for Pfizer. Juan W Valle has participated in Consulting or Advisory Role and Speakers' Bureaus for Pfizer. Eric Raymond has received Consulting and Research Grants from Pfizer.

Figures

Figure 1
Figure 1
Outcome of patients treated with sunitib/placebo. Progression-free survival in patients with sunitinib and placebo (A); Waterfall-plot showing best changes in sum of marker lesions in patients treated with sunitinib (B) and placebo (C). HR, hazard ratio; 95% CI, 95% confidence interval; n, number of patients.
Figure 2
Figure 2
Progression free-survival in patients treated with 37.5 mg continuously of sunitinib (Phase III clinical trial), both cut-offs (classical 30% and our proposed alternative 10%) are tested. CI=confidence interval; HR=hazard ratio; 95% nr=not-reached.

References

    1. Chen HX, Rubinstein LV, Shankar LK, Abrams JS (2014) Are we ready for the 10% solution? Oncologist 19: 439–440.
    1. Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, Dancey J, Arbuck S, Gwyther S, Mooney M, Rubinstein L, Shankar L, Dodd L, Kaplan R, Lacombe D, Verweij J (2009) New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer 45: 228–247.
    1. EMA Last accessed 23th October (2016) Sunitinib SPC.
    1. Faivre S, Ronot M, Dreyer C, Serrate C, Hentic O, Bouattour M, Bruno O, Couvelard A, Vilgrain V, Raymond E (2012) Imaging response in neuroendocrine tumors treated with targeted therapies: the experience of sunitinib. Target Oncol 7: 127–133.
    1. Houk BE, Bello CL, Poland B, Rosen LS, Demetri GD, Motzer RJ (2010) Relationship between exposure to sunitinib and efficacy and tolerability endpoints in patients with cancer: results of a pharmacokinetic/pharmacodynamic meta-analysis. Cancer Chemother Pharmacol 66: 357–371.
    1. Krajewski KM, Franchetti Y, Nishino M, Fay AP, Ramaiya N, Van den Abbeele AD, Choueiri TK (2014) 10% Tumor diameter shrinkage on the first follow-up computed tomography predicts clinical outcome in patients with advanced renal cell carcinoma treated with angiogenesis inhibitors: a follow-up validation study. Oncologist 19: 507–514.
    1. Krajewski KM, Guo M, Van den Abbeele AD, Yap J, Ramaiya N, Jagannathan J, Heng DY, Atkins MB, McDermott DF, Schutz FA, Pedrosa I, Choueiri TK (2011) Comparison of four early posttherapy imaging changes (EPTIC; RECIST 1.0, tumor shrinkage, computed tomography tumor density, Choi criteria) in assessing outcome to vascular endothelial growth factor-targeted therapy in patients with advanced renal cell carcinoma. Eur Urol 59: 856–862.
    1. Kulke MH, Lenz HJ, Meropol NJ, Posey J, Ryan DP, Picus J, Bergsland E, Stuart K, Tye L, Huang X, Li JZ, Baum CM, Fuchs CS (2008) Activity of sunitinib in patients with advanced neuroendocrine tumors. J Clin Oncol 26: 3403–3410.
    1. Luo YCJ (2017) Optimisation of the Size Variation Threshold for CT Evaluation of Response in Advanced Gastroenteropancreatic Nauroendocrine Tumors Treated with Octreotide LAR. ENETS Annual Coference 2017; abstract K11.
    1. Ornstein MC, Wood L, Elson P, Allman K, Beach J, Martin A, Gilligan T, Garcia JA, Rini BI (2016) Clinical effect of dose escalation after disease progression in patients with metastatic renal cell carcinoma. Clin Genitourin Cancer 15: e275–e280.
    1. Patel S (2012) Managing progressive disease in patients with GIST: factors to consider besides acquired secondary tyrosine kinase inhibitor resistance. Cancer Treat Rev 38: 467–472.
    1. Raymond E, Dahan L, Raoul JL, Bang YJ, Borbath I, Lombard-Bohas C, Valle J, Metrakos P, Smith D, Vinik A, Chen JS, Horsch D, Hammel P, Wiedenmann B, Van Cutsem E, Patyna S, Lu DR, Blanckmeister C, Chao R, Ruszniewski P (2011) Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. N Engl J Med 364: 501–513.
    1. Shi HZ, Tian J, Chen X, Wang D, Li CL (2016) Clinical experience of escalated sunitinib dose in select patients with metastatic renal cell carcinoma. Clin Genitourin Cancer 15: 139–144.
    1. Sundin A, Rockall A (2012) Therapeutic monitoring of gastroenteropancreatic neuroendocrine tumors: the challenges ahead. Neuroendocrinology 96: 261–271.
    1. Therasse P, Arbuck SG, Eisenhauer EA, Wanders J, Kaplan RS, Rubinstein L, Verweij J, Van Glabbeke M, van Oosterom AT, Christian MC, Gwyther SG (2000) New guidelines to evaluate the response to treatment in solid tumors. European Organization for Research and Treatment of Cancer, National Cancer Institute of the United States, National Cancer Institute of Canada. J Natl Cancer Inst 92: 205–216.
    1. Thiam R, Fournier LS, Trinquart L, Medioni J, Chatellier G, Balvay D, Escudier B, Dromain C, Cuenod CA, Oudard S (2010) Optimizing the size variation threshold for the CT evaluation of response in metastatic renal cell carcinoma treated with sunitinib. Ann Oncol 21: 936–941.
    1. van der Veldt AA, Meijerink MR, van den Eertwegh AJ, Haanen JB, Boven E (2010) Choi response criteria for early prediction of clinical outcome in patients with metastatic renal cell cancer treated with sunitinib. Br J Cancer 102: 803–809.
    1. Yao JC, Shah MH, Ito T, Bohas CL, Wolin EM, Van Cutsem E, Hobday TJ, Okusaka T, Capdevila J, de Vries EG, Tomassetti P, Pavel ME, Hoosen S, Haas T, Lincy J, Lebwohl D, Oberg K (2011) Everolimus for advanced pancreatic neuroendocrine tumors. N Engl J Med 364: 514–523.

Source: PubMed

3
Abonneren