Phase II study of sunitinib in Japanese patients with unresectable or metastatic, well-differentiated pancreatic neuroendocrine tumor

Tetsuhide Ito, Takuji Okusaka, Toshirou Nishida, Kenji Yamao, Hisato Igarashi, Chigusa Morizane, Shunsuke Kondo, Nobumasa Mizuno, Kazuo Hara, Akira Sawaki, Satoshi Hashigaki, Nobuyuki Kimura, Mami Murakami, Emiko Ohki, Richard C Chao, Masayuki Imamura, Tetsuhide Ito, Takuji Okusaka, Toshirou Nishida, Kenji Yamao, Hisato Igarashi, Chigusa Morizane, Shunsuke Kondo, Nobumasa Mizuno, Kazuo Hara, Akira Sawaki, Satoshi Hashigaki, Nobuyuki Kimura, Mami Murakami, Emiko Ohki, Richard C Chao, Masayuki Imamura

Abstract

Background: Pancreatic neuroendocrine tumors (NETs) are rare but are frequently diagnosed at advanced stages and require systemic therapy.

Patients and methods: This multicenter, open-label, phase II study evaluated sunitinib in Japanese patients with well-differentiated pancreatic NET. Patients received sunitinib 37.5 mg/day on a continuous daily dosing (CDD) schedule. The primary endpoint was clinical benefit rate (CBR; percentage of complete responses [CRs] plus partial responses [PRs] plus stable disease [SD] ≥ 24 weeks). Secondary endpoints included objective response rate (ORR), tumor shrinkage, progression-free survival (PFS) probability, safety, pharmacokinetics, and biomarkers.

Results: Twelve patients received treatment. The CBR was 75 % (95 % confidence interval [CI], 43-94) and included 6 patients with a PR and 3 with SD. The ORR was 50 % (95 % CI, 21-79). PFS probability was 91 % (95 % CI, 54-99) at 6 months and 71 % (95 % CI, 34-90) at 12 months. Commonly reported treatment-emergent (all-causality), any-grade adverse events included diarrhea (n=10), hand-foot syndrome and hypertension (both n=8), fatigue and headache (both n=7), and neutropenia (n=6). No deaths on study were reported; one death due to disease progression occurred >28 days after end of treatment. Sunitinib on a CDD schedule resulted in sustained drug concentrations without accumulation across cycles. Tumor responses in all 12 patients did not appear to correlate with decreases in chromogranin A levels.

Conclusions: Sunitinib 37.5 mg/day on a CDD schedule demonstrated antitumor activity in Japanese patients with unresectable, well-differentiated pancreatic NET. Commonly reported adverse events were consistent with the known safety profile of sunitinib.

Trial registration: ClinicalTrials.gov NCT01121562.

Figures

Fig. 1
Fig. 1
Maximum percentage reduction from baseline in target lesion size by patient (N = 12). Although one patient had a maximum percentage change in target tumor size from baseline of −100 %, non-target lesions remained and therefore this was not classified as a complete response. Asterisk stable disease of ≥24 weeks in duration; RECIST Response Evaluation Criteria in Solid Tumors
Fig. 2
Fig. 2
Individual patient profiles and response to treatment (N = 12). a Summary of patient profiles and changes in tumor-size and chromogranin A levels. b Percentage change from baseline in target lesion size over time in individual patients. a Maximum % change = [(minimum value after dosing – baseline)/baseline] x 100; b Based on censored data; F female; M male; PD progressive disease; PFS progression-free survival; PR partial response; SD stable disease
Fig. 3
Fig. 3
Trough concentrations of sunitinib, active metabolite SU12662, and total drug (sunitinib plus SU12662) in Japanese patients with pancreatic neuroendocrine tumor (NET; n = 11; present study) or gastrointestinal stromal tumor (GIST; n = 30) [21] and renal cell carcinoma (RCC; n = 38; pooled data) [25], and in predominantly Western patients with pancreatic NET; n = 57 [17]. The sunitinib dose in each study was dose-corrected to 37.5 mg. The upper and lower box boundaries denote the 75th and 25th percentiles, respectively, with the median shown as a line within the box. Whiskers indicate minimum and maximum values. Outlying values are denoted as circles

References

    1. Ito T, Sasano H, Tanaka M, Osamura RY, Sasaki I, Kimura W, Takano K, Obara T, Ishibashi M, Nakao K, Doi R, Shimatsu A, Nishida T, Komoto I, Hirata Y, Nakamura K, Igarashi H, Jensen RT, Wiedenmann B, Imamura M. Epidemiological study of gastroenteropancreatic neuroendocrine tumors in Japan. J Gastroenterol. 2010;45:234–243. doi: 10.1007/s00535-009-0194-8.
    1. Yao JC, Hassan M, Phan A, Dagohoy C, Leary C, Mares JE, Abdalla EK, Fleming JB, Vauthey JN, Rashid A, Evans DB. One hundred years after “carcinoid”: epidemiology of and prognostic factors for neuroendocrine tumors in 35,825 cases in the United States. J Clin Oncol. 2008;26:3063–3072. doi: 10.1200/JCO.2007.15.4377.
    1. National Comprehensive Cancer Network (NCCN) (2012) National Comprehensive Cancer Network Clinical Practice Guidelines in Oncology. Neuroendocr Tumors v1.2012
    1. Oberg K, Kvols L, Caplin M, Delle Fave G, de Herder W, Rindi G, Ruszniewski P, Woltering EA, Wiedenmann B. Consensus report on the use of somatostatin analogs for the management of neuroendocrine tumors of the gastroenteropancreatic system. Ann Oncol. 2004;15:966–973. doi: 10.1093/annonc/mdh216.
    1. McCollum AD, Kulke MH, Ryan DP, Clark JW, Shulman LN, Mayer RJ, Bartel S, Fuchs CS. Lack of efficacy of streptozocin and doxorubicin in patients with advanced pancreatic endocrine tumors. Am J Clin Oncol. 2004;27:485–488. doi: 10.1097/01.coc.0000135343.06038.eb.
    1. Saltz L, Trochanowski B, Buckley M, Heffernan B, Niedzwiecki D, Tao Y, Kelsen D. Octreotide as an antineoplastic agent in the treatment of functional and nonfunctional neuroendocrine tumors. Cancer. 1993;72:244–248. doi: 10.1002/1097-0142(19930701)72:1<244::AID-CNCR2820720143>;2-Q.
    1. Raymond E, Dahan L, Raoul J-L, Bang Y-J, Borbath I, Lombard-Bohas C, Valle J, Metrakos P, Smith D, Vinik A, Chen J-S, Hörsch D, Hammel P, Wiedenmann B, Van Cutsem E, Patyna S, Lu DR, Blanckmeister C, Chao R, Ruszniewski P. Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. N Engl J Med. 2011;364:501–513. doi: 10.1056/NEJMoa1003825.
    1. Yao JC, Shah MH, Ito T, Bohas CL, Wolin EM, Van CE, Hobday TJ, Okusaka T, Capdevila J, de Vries EG, Tomassetti P, Pavel ME, Hoosen S, Haas T, Lincy J, Lebwohl D, Oberg K. Everolimus for advanced pancreatic neuroendocrine tumors. N Engl J Med. 2011;364:514–523. doi: 10.1056/NEJMoa1009290.
    1. Ito T, Okusaka T, Ikeda M, Igarashi H, Morizane C, Nakachi K, Tajima T, Kasuga A, Fujita Y, Furuse J. Everolimus for advanced pancreatic neuroendocrine tumours: a subgroup analysis evaluating Japanese patients in the RADIANT-3 trial. Jpn J Clin Oncol. 2012;42:903–911. doi: 10.1093/jjco/hys123.
    1. Casanovas O, Hicklin DJ, Bergers G, Hanahan D. Drug resistance by evasion of antiangiogenic targeting of VEGF signaling in late-stage pancreatic islet tumors. Cancer Cell. 2005;8:299–309. doi: 10.1016/j.ccr.2005.09.005.
    1. Inoue M, Hager JH, Ferrara N, Gerber HP, Hanahan D. VEGF-A has a critical, nonredundant role in angiogenic switching and pancreatic beta cell carcinogenesis. Cancer Cell. 2002;1:193–202. doi: 10.1016/S1535-6108(02)00031-4.
    1. Abrams TJ, Lee LB, Murray LJ, Pryer NK, Cherrington JM. SU11248 inhibits KIT and platelet-derived growth factor receptor beta in preclinical models of human small cell lung cancer. Mol Cancer Ther. 2003;2:471–478. doi: 10.4161/cbt.2.5.446.
    1. Mendel DB, Laird AD, Xin X, Louie SG, Christensen JG, Li G, Schreck RE, Abrams TJ, Ngai TJ, Lee LB, Murray LJ, Carver J, Chan E, Moss KG, Haznedar JO, Sukbuntherng J, Blake RA, Sun L, Tang C, Miller T, Shirazian S, McMahon G, Cherrington JM. In vivo antitumor activity of SU11248, a novel tyrosine kinase inhibitor targeting vascular endothelial growth factor and platelet-derived growth factor receptors: determination of a pharmacokinetic/pharmacodynamic relationship. Clin Cancer Res. 2003;9:327–337.
    1. Murray LJ, Abrams TJ, Long KR, Ngai TJ, Olson LM, Hong W, Keast PK, Brassard JA, O’Farrell AM, Cherrington JM, Pryer NK. SU11248 inhibits tumor growth and CSF-1R-dependent osteolysis in an experimental breast cancer bone metastasis model. Clin Exp Metastasis. 2003;20:757–766. doi: 10.1023/B:CLIN.0000006873.65590.68.
    1. Pietras K, Hanahan D. A multitargeted, metronomic, and maximum-tolerated dose “chemo-switch” regimen is antiangiogenic, producing objective responses and survival benefit in a mouse model of cancer. J Clin Oncol. 2005;23:939–952. doi: 10.1200/JCO.2005.07.093.
    1. Yao VJ, Sennino B, Davis RB, Christensen JG, Hu-Lowe D, Roberts G, McDonald DM (2006) Combined anti-VEGFR and anti-PDGFR actions of sunitinib on blood vessels in preclinical tumor models. Presented at the 18th EORTC-NCI-AACR symposium, Prague, Czech Republic (Abstract 78)
    1. Kulke M, Lenz HJ, Meropol N, Posey J, Ryan DP, Picus J, Bergsland E, Stuart K, Tye L, Huang X, Li J, Baum C, Fuchs C. Activity of sunitinib in patients with advanced neuroendocrine tumors. J Clin Oncol. 2008;26:3403–3410. doi: 10.1200/JCO.2007.15.9020.
    1. Vinik A, Cutsem E, Niccoli P, Raoul J-L, Bang Y-J, Borbath I, Valle JW, Metrakos P, Smith D, Chen J-S, Hoersch D, Castellano DE, Kennecke HF, Picus J, Hazel G, Patyna S, Lu D, Chao RC, Raymond E. Updated results from a phase III trial of sunitinib versus placebo in patients with progressive, unresectable, well-differentiated pancreatic neuroendocrine tumor (NET) J Clin Oncol. 2012;30:4118.
    1. Bosman FT, Carnerio F, Hruban RH, et al. WHO classification of tumors of the digestive system (world health classification of tumors) Lyon: IARC Press; 2010.
    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. New guidelines to evaluate the response to treatment in solid tumors. J Natl Cancer Inst. 2000;92:205–216. doi: 10.1093/jnci/92.3.205.
    1. Shirao K, Nishida T, Doi T, Komatsu Y, Muro K, Li Y, Ueda E, Ohtsu A. Phase I/II study of sunitinib malate in Japanese patients with gastrointestinal stromal tumor after failure of prior treatment with imatinib mesylate. Invest New Drugs. 2010;28:866–875. doi: 10.1007/s10637-009-9306-9.
    1. Tomita Y, Shinohara N, Yuasa T, Fujimoto H, Niwakawa M, Mugiya S, Miki T, Uemura H, Nonomura N, Takahashi M, Hasegawa Y, Agata N, Houk B, Naito S, Akaza H. Overall survival and updated results from a phase II study of sunitinib in Japanese patients with metastatic renal cell carcinoma. Jpn J Clin Oncol. 2010;40:1166–1172. doi: 10.1093/jjco/hyq146.
    1. Demetri GD, van Oosterom AT, Garrett CR, Blackstein ME, Shah MH, Verweij J, McArthur G, Judson IR, Heinrich MC, Morgan JA, Desai J, Fletcher CD, George S, Bello CL, Huang X, Baum CM, Casali PG. Efficacy and safety of sunitinib in patients with advanced gastrointestinal stromal tumour after failure of imatinib: a randomised controlled trial. Lancet. 2006;368:1329–1338. doi: 10.1016/S0140-6736(06)69446-4.
    1. Motzer RJ, Hutson TE, Tomczak P, Dror Michaelson M, Bukowski RM, Oudard S, Negrier S, Szczylik C, Pili R, Bjarnason GA, Garcia-del-Muro X, Sosman JA, Solska E, Wilding G, Thompson JA, Kim ST, Chen I, Huang X, Figlin RA. Overall survival and updated results for sunitinib versus interferon alfa in first-line treatment of patients with metastatic renal cell carcinoma. J Clin Oncol. 2009;27:3584–3590. doi: 10.1200/JCO.2008.20.1293.
    1. Uemura H, Shinohara N, Yuasa T, Tomita Y, Fujimoto H, Niwakawa M, Mugiya S, Miki T, Nonomura N, Takahashi M, Hasegawa Y, Agata N, Houk B, Naito S, Akaza H. A phase II study of sunitinib in Japanese patients with metastatic renal cell carcinoma: insights into the treatment, efficacy and safety. Jpn J Clin Oncol. 2010;40:194–202. doi: 10.1093/jjco/hyp146.
    1. SUTENT® (sunitinib malate) prescribing information (2012) Pfizer Inc. New York, NY, USA
    1. Afinitor® (everolimus) prescribing information (2012) Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
    1. Yao JC, Lombard-Bohas C, Baudin E, Kvols LK, Rougier P, Ruszniewski P, Hoosen S, St PJ, Haas T, Lebwohl D, Van CE, Kulke MH, Hobday TJ, O’Dorisio TM, Shah MH, Cadiot G, Luppi G, Posey JA, Wiedenmann B. 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. doi: 10.1200/JCO.2009.24.2669.
    1. Modlin IM, Gustafsson BI, Moss SF, Pavel M, Tsolakis AV, Kidd M. Chromogranin A–biological function and clinical utility in neuro endocrine tumor disease. Ann Surg Oncol. 2010;17:2427–2443. doi: 10.1245/s10434-010-1006-3.

Source: PubMed

3
Se inscrever