asco-gi: the efficacy and safety of sunitinib in patients with advanced well-differentiated...

1
Presented at the 2017 American Society of Clinical Oncology Gastrointestinal Cancer Symposium (ASCO GI), January 19–21, 2017, San Francisco, CA CONCLUSIONS n Median PFS of 13.2 months (95% CI, 10.9–16.7) and ORR of 24.5% (95% CI, 16.7–33.8) observed in this phase IV trial support the outcomes of the pivotal phase III trial of sunitinib in pNETs and confirm its activity in this setting. n OS data were not mature at the time of the primary analysis. n AEs were consistent with the known safety profile of sunitinib. n The study confirmed sunitinib is an efficacious and safe treatment option in progressive, locally advanced and/or metastatic, well-differentiated, unresectable pNETs. The Efficacy and Safety of Sunitinib in Patients With Advanced Well-Differentiated Pancreatic Neuroendocrine Tumors Eric Raymond 1 , Matthew H Kulke 2 , Shukui Qin 3 , Michael Schenker 4 , Antonio Cubillo 5 , Wenhui Lou 6 , Jiri Tomasek 7 , Espen Thiis-Evensen 8 , Jianming Xu 9 , Karoly Racz 10 , Adina E Croitoru 11 , Mustafa Khasraw 12 , Eva Sedlackova 13 , Ivan Borbath 14 , Paul Ruff 15 , Paul E Oberstein 16 , Tetsuhide Ito 17 , Kathrine C Fernandez 18 , Brad Rosbrook 19 , Nicola Fazio 20 1 Paris Saint-Joseph Hospital Group, Paris, France; 2 Dana-Farber Cancer Institute, Boston, MA, USA; 3 PLA Cancer Center of Nanjing Bayi Hospital, Nanjing, China; 4 Centrul de Oncologie Sf. Nectarie, Oncologie Medicala, Craiova, Romania; 5 Hospital Universitario Madrid Sanchinarro, Centro Integral Oncológico Clara Campal, Madrid, Spain; 6 Zhongshan Hospital, Fudan University, Shanghai, China; 7 Masaryk Memorial Cancer Institute, Faculty of Medicine, Masaryk University, Brno, Czech Republic; 8 Oslo University Hospital, Department of Gastroenterology, Rikshospitalet, Oslo, Norway; 9 No.307 Hospital, Academy of Military Medical Sciences, Beijing, China; 10 Semmelweis University, Faculty of Medicine, 2nd Department of Internal Medicine, Budapest, Hungary; 11 Fundeni Clinical Institute, Department of Medical Oncology, Bucharest, Romania; 12 Andrew Love Cancer Center, Geelong Hospital, Victoria, Australia; 13 Všeobecné Fakultní Nemocnice v Praze Onkologická Klinika, Praha, Czech Republic; 14 Cliniques Universitaires Saint-Luc, King Albert II Institute Cancerology and Hematology, Brussels, Belgium; 15 University of the Witwatersrand, Faculty of Health Sciences, Johannesburg, South Africa; 16 Columbia University Medical Center, Division of Hematology/Oncology, New York, NY, USA; 17 Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; 18 Pfizer Inc, Cambridge, MA, USA; 19 Pfizer Inc, San Diego, CA, USA; 20 IEO, European Institute of Oncology, Unit of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, Milan, Italy 380 REFERENCES 1. American Cancer Society. Cancer Facts & Figures 2016. Atlanta, GA. 2. Raymond E, et al. N Engl J Med 2011;364:501-13. 3. Faivre S, et al. Ann Oncol 2016; Nov 10 [Epub ahead of print]. 4. Sutent ® (sunitinib malate) prescribing information. New York, NY: Pfizer Inc; 2015. ACKNOWLEDGMENTS We thank the participating patients and their families, as well as the global network of research nurses, trial coordinators, and operations staff for their contributions, and investigators who participated in this trial, including: M. Michael (Australia); X. Yu, Y. Shen and L. Jia (China); P. Hammel (France); S. Skrikhande (India); C. Morizane (Japan); J. Sufliarsky (Slovakia); and G. Khan (United States). This study was sponsored by Pfizer. Medical writing support was provided by Mariko Nagashima, PhD, and Anne Marie Reid, PhD, of Engage Scientific Solutions, and was funded by Pfizer. For information on this poster, contact x xxxxx at xxxx.xxxx Copyright © 2016 INTRODUCTION Pancreatic neuroendocrine tumors (pNETs) are rare tumors. Of an estimated 53,070 new cases of pancreatic cancers expected in the US in 2016, pNETs account for fewer than 5%. 1 Due to their relatively indolent nature, the majority of patients are diagnosed with metastatic pNETs, for whom treatment options are limited. pNETs are highly vascularized tumors; aberrant expression of vascular endothelial growth factor (VEGF) and its receptors, which play vital roles in tumor angiogenesis, has been observed. Sunitinib, a multi-targeted tyrosine kinase inhibitor (TKI) of angiogenesis, showed a significant increase in progression-free survival (PFS) over placebo in well-differentiated, advanced and/or metastatic pNETs in a pivotal phase III clinical trial (ClinicalTrials.gov NCT00428597). 2 The trial was terminated early after more-serious adverse events (AEs) and deaths were observed in the placebo arm and a difference in PFS favored the sunitinib arm (hazard ratio [HR] 0.42; 95% confidence interval [CI], 0.26–0.66; P<0.001; median: 11.4 vs 5.5 months). The median (95% CI) 5-year overall survival (OS) was 38.6 (25.6–56.4) months for sunitinib and 29.1 (16.4–36.8) months for placebo (HR 0.73; 95% CI, 0.50–1.06; P=0.094), with 69% of placebo patients having crossed over to sunitinib. 3 In 2010 and 2011, sunitinib was approved by the European Medicines Agency and US Food and Drug Administration (FDA), respectively, for the treatment of patients with progressive, locally advanced and/or metastatic, well-differentiated, unresectable pNETs. 4 OBJECTIVES This phase IV clinical trial (ClinicalTrials.gov NCT01525550) was conducted as post-approval commitments to the FDA and other regulatory agencies to confirm the efficacy and safety of sunitinib in advanced and/or metastatic, well-differentiated, unresectable pNETs. METHODS Study Design This multinational, single-arm, open-label, phase IV clinical trial is ongoing. The primary endpoint is investigator-assessed PFS per the Response Evaluation Criteria in Solid Tumors (RECIST) v1.0. Secondary endpoints include PFS assessed by the independent radiological review, time to tumor progression (TTP), objective response rate (ORR), OS, and safety. Key Eligibility Criteria Histologically or cytologically confirmed, well-differentiated, unresectable or metastatic pNETs with documented progression within 12 months of study enrollment. Not amenable to surgery, radiation, or combined modality therapy with curative intent. Presence of ≥1 measurable target lesion per RECIST v1.0. Eastern Cooperative Oncology Group performance status (ECOG PS) 0 or 1. No pre-existing uncontrolled hypertension (ie, blood pressure >150/100 mmHg despite medical therapy). No prior treatment with TKIs, anti-VEGF, or other angiogenesis inhibitors. Treatment and Assessments Patients received 37.5 mg sunitinib orally once a day on a continuous daily-dosing regimen. Sunitinib dose could be increased to 50 mg daily any time after 8 weeks of treatment initiation in patients without treatment response who experienced only grade 1 or lower nonhematologic or grade 2 or lower hematologic treatment-related AEs. Dose could be temporarily interrupted or reduced to 25 mg daily to manage severe toxicity. Somatostatin analogs for control of symptoms were permitted at the investigator’s discretion. Radiologic tumor assessments were conducted at screening, Cycle 2 Day 1, Cycle 3 Day 1, and every 2 cycles thereafter; tumor responses were evaluated per RECIST v1.0 criteria. Safety was monitored throughout the study and AEs were graded according to the National Cancer Institute Common Terminology Criteria for Adverse Events v3.0. Statistical Analyses More than 80 patients were to be enrolled: 40 treatment-naïve (defined as those who never received systemic antitumor therapy; somatostatin analogs for symptomatic control were allowed) and 40 previously treated patients. Median PFS and 95% CI were estimated using the Kaplan–Meier method for the entire population as well as separately for treatment-naïve and previously treated patients. Descriptive statistics were used to summarize other parameters. RESULTS Patients Of 123 patients screened, 106 (61 treatment-naïve and 45 previously treated) were enrolled at 25 centers in 15 countries (Figure 1). Figure 1: Trial Profile 123 patients screened 106 patients enrolled 61 txt-naïve patients treated 45 previously treated patients treated 39 previously treated patients discontinued txt 23 Objective progression/relapse 8 Adverse events 1 Refusal of txt for reason other than AE 2 Global health deterioration 1 Death 4 Other 43 txt-naïve patients discontinued txt 26 Objective progression/relapse 6 Adverse events 4 Refusal of txt for reason other than AE 2 Global health deterioration 1 Death 4 Other Efficacy analysis: 61 txt-naïve patients Safety analysis: 61 txt-naïve patients 45 previously treated patients 45 previously treated patients AE=adverse event; Txt=treatment Patient demographics and baseline characteristics are summarized in Table 1. More than half (n=64/106, 60.4%) of patients had a nonfunctioning tumor and 17.9% had a functioning tumor (unknown for 21.7% of patients). In regard to prior locoregional treatment, 18.9% of patients had trans-arterial chemoembolization; radiofrequency ablation (3.8%); or trans-arterial embolization, percutaneous injections, or microwave ablation (2.8% each). Median (range) treatment duration in the total population was 11.7 months (0.2–40.3): 12.2 months (0.2–35.9) in treatment-naïve vs 10.2 months (0.5–40.3) in previously treated patients. Efficacy Median PFS as assessed by investigators was 13.2 months (95% CI, 10.9–16.7); median PFS was similar in treatment-naïve and previously treated patients (13.2 months [95% CI, 7.4–16.8] and 13.0 months [95% CI, 9.2–20.4], respectively; Figure 2). Median PFS as assessed by independent radiological review was 11.1 months (95% CI, 7.4–16.6), in treatment-naive patients 11.1 months (95% CI, 5.5–16.7), and in previously treated patients 9.5 months (95% CI, 7.4–18.4). 7 patients were censored due to lack of adequate baseline assessments. Table 1: Demographics and Patient Baseline Characteristics Characteristics Treatment-naïve n=61 Previously Treated n=45 Total N=106 Age, years, mean (SD) 55.4 (8.9) 53.5 (9.1) 54.6 (9.0) Gender, n (%) Male 30 (49.2) 33 (73.3) 63 (59.4) Female 31 (50.8) 12 (26.7) 43 (40.6) Race, n (%) White 32 (52.5) 35 (77.8) 67 (63.2) Black 2 (3.3) 0 2 (1.9) Asian 27 (44.3) 10 (22.2) 37 (34.9) ECOG PS, n (%) 0 39 (63.9) 29 (64.4) 68 (64.2) 1 22 (36.1) 16 (35.6) 38 (35.8) No. of involved disease sites,* n (%) 1 24 (39.3) 9 (20.0) 33 (31.1) 2 19 (31.1) 22 (48.9) 41 (38.7) 3 11 (18.0) 8 (17.8) 19 (17.9) 4 3 (4.9) 3 (6.7) 6 (5.7) >4 4 (6.6) 3 (6.7) 7 (6.6) Tumor site,* n (%) Liver 57 (93.4) 41 (91.1) 98 (92.5) Pancreas 22 (36.1) 25 (55.6) 47 (44.3) Lymph node, distant 16 (26.2) 13 (28.9) 29 (27.4) Lymph node, regional 13 (21.3) 7 (15.6) 20 (18.9) Lung 3 (4.9) 3 (6.7) 6 (5.7) Other 10 (16.4) 10 (22.2) 20 (18.9) Prior systemic chemotherapy, n (%) Any 0 45 (100) 45 (42.5) Neoadjuvant 0 2 (4.4) 2 (1.9) Adjuvant 0 4 (8.9) 4 (3.8) Advanced/metastatic 0 39 (86.7) 39 (36.8) Prior SSA, n (%) 24 (39.3) 27 (60.0) 51 (48.1) Ki-67 index, mean (SD) 6.7 (5.0) 8.4 (7.2) 7.4 (6.0) * Included both target and nontarget sites; sites with multiple lesions were counted once. † Patients with regimens that consist only of somatostatin analogs were considered treatment-naïve. ECOG PS, Eastern Cooperative Oncology Group performance status; SD=standard deviation; SSA=somatostatin analogs Figure 2: Kaplan–Meier Estimates of PFS in Treatment-Naïve and Previously Treated Patients With pNETs, Assessed by Investigators PFS Distribution Function 1.0 0.9 0.8 0.7 0.6 0.5 0.4 0.3 0.2 0.1 0.0 0 5 10 15 20 25 Time (Months) No. at risk: Treatment-naïve Previously treated 61 45 41 29 32 20 14 10 8 8 6 4 0 4 0 2 0 1 0 0 30 35 40 45 Treatment-naïve 61 37 13.2 (7.4–16.8) Previously treated 45 28 13.0 (9.2–20.4) n Events mPFS (95% CI), mo CI=confidence interval; mPFS=median progression-free survival; PFS=progression-free survival; pNETs=pancreatic neuroendocrine tumors ORR was 24.5% (95% CI, 16.7–33.8) according to the investigator assessment (Table 2). Table 2: Best Observed Response by RECIST, Assessed by Investigators Treatment-naïve n=61 Previously Treated n=45 Total N=106 Best overall response, n (%) Complete response 2 (3.3) 1 (2.2) 3 (2.8) Partial response 11 (18.0) 12 (26.7) 23 (21.7) Stable disease 40 (65.6) 29 (64.4) 69 (65.1) Progressive disease 7 (11.5) 2 (4.4) 9 (8.5) Indeterminate 1 (1.6) 1 (2.2) 2 (1.9) ORR,* n (%) 13 (21.3) 13 (28.9) 26 (24.5) 95% CI 11.9–33.7 16.4–44.3 16.7–33.8 * Complete response + partial response. CI=confidence interval; ORR=objective response rate; RECIST=Response Evaluation Criteria in Solid Tumors Median TTP was 14.5 months (95% CI, 11.0–16.7); median TTP in treatment-naïve and previously treated patients was similar (14.8 [95% CI, 7.5–16.8] and 14.5 [95% CI, 9.2–20.4] months, respectively). OS data were not mature at the time of data cutoff date (Mar 19, 2016); 29 (27.4%) patients had died and median OS was 37.8 months (95% CI, 33.0–not estimable). Safety Most-common treatment-emergent, all-grade AEs experienced by all patients treated with sunitinib included neutropenia, diarrhea, and leukopenia (Table 3). No major differences were observed in the incidence of AEs reported by treatment- naïve vs previously treated patients, except dyspepsia, nausea, and neutropenia. Percentage of treatment-naïve and previously treated patients who experienced Grade 3 or 4 AEs was comparable; serious AEs were also comparable: 24.6% (n=15) vs 24.4% (n=11), respectively. 15 (24.6%) treatment-naïve and 5 (11.1%) previously treated patients had sunitinib dose reductions due to AEs; 8 (13.1%) and 10 (22.2%) patients, respectively, discontinued treatment due to AEs. Please scan this Quick Response (QR) code with your smartphone app to view an electronic version of this poster. If you don’t have a smartphone, access the poster via the internet at: http://congress-download.pfizer.com/asco_gi_2017_american_society_of_clinical_oncology_ gastrointestinal_cancers_symposium_295_sutent_raymond_380.html Copies of this poster obtained through QR Code are for personal use only and may not be reproduced without permission from ASCO ® and the author of this poster. Table 3: Treatment-Emergent, All-Causality Adverse Events Adverse Events,* n (%) Treatment-naïve n=61 Previously Treated n=45 Total N=106 All Grades Grade 3/4 All Grades Grade 3/4 All Grades Grade 3/4 Neutropenia 37 (60.7) 13 (21.3) 22 (48.9) 10 (22.2) 59 (55.7) 23 (21.7) Diarrhea 32 (52.5) 7 (11.5) 22 (48.9) 3 (6.7) 54 (50.9) 10 (9.4) Leukopenia 25 (41.0) 4 (6.6) 21 (46.7) 3 (6.7) 46 (43.4) 7 (6.6) Fatigue 19 (31.1) 1 (1.6) 14 (31.1) 0 (0.0) 33 (31.1) 1 (0.9) Hand–foot syndrome 19 (31.1) 5 (8.2) 14 (31.1) 2 (4.4) 33 (31.1) 7 (6.6) Thrombocytopenia 18 (29.5) 6 (9.8) 14 (31.1) 2 (4.4) 32 (30.2) 8 (7.5) Hypertension 16 (26.2) 4 (6.6) 11 (24.4) 2 (4.4) 27 (25.5) 6 (5.7) Abdominal pain 16 (26.2) 2 (3.3) 10 (22.2) 3 (6.7) 26 (24.5) 5 (4.7) Dysgeusia 14 (23.0) 0 (0.0) 11 (24.4) 0 (0.0) 25 (23.6) 0 (0.0) Nausea 11 (18.0) 0 (0.0) 14 (31.1) 1 (2.2) 25 (23.6) 1 (0.9) Dyspepsia 7 (11.5) 0 (0.0) 14 (31.1) 0 (0.0) 21 (19.8) 0 (0.0) Headache 12 (19.7) 0 (0.0) 9 (20.0) 0 (0.0) 21 (19.8) 0 (0.0) Stomatitis 13 (21.3) 2 (3.3) 6 (13.3) 1 (2.2) 19 (17.9) 3 (2.8) Vomiting 8 (13.1) 1 (1.6) 10 (22.2) 1 (2.2) 18 (17.0) 2 (1.9) Asthenia 10 (16.4) 0 (0.0) 7 (15.6) 2 (4.4) 17 (16.0) 2 (1.9) Abdominal pain upper 5 (8.2) 1 (1.6) 7 (15.6) 1 (2.2) 12 (11.3) 2 (1.9) ALT increased 2 (3.3) 1 (1.6) 9 (20.0) 0 (0.0) 11 (10.4) 1 (0.9) AST increased 4 (6.6) 1 (1.6) 7 (15.6) 1 (2.2) 11 (10.4) 2 (1.9) Constipation 3 (4.9) 0 (0.0) 7 (15.6) 0 (0.0) 10 (9.4) 0 (0.0) Dizziness 3 (4.9) 0 (0.0) 7 (15.6) 0 (0.0) 10 (9.4) 0 (0.0) Hypophosphatemia 2 (3.3) 2 (3.3) 7 (15.6) 3 (6.7) 9 (8.5) 5 (4.7) Myalgia 2 (3.3) 0 (0.0) 7 (15.6) 0 (0.0) 9 (8.5) 0 (0.0) Adverse events are listed by highest to lowest % for Total/All Grades patients. * Adverse events reported by ≥15% in any treatment group per MedDRA criteria. ALT=alanine aminotransferase; AST=aspartate aminotransferase; MedDRA=Medical Dictionary for Regulatory Activities

Upload: prof-eric-raymond-oncologie-medicale

Post on 08-Feb-2017

57 views

Category:

Health & Medicine


0 download

TRANSCRIPT

Presented at the 2017 American Society of Clinical Oncology Gastrointestinal Cancer Symposium (ASCO GI), January 19–21, 2017, San Francisco, CA

CONCLUSIONS n Median PFS of 13.2 months (95% CI, 10.9–16.7) and ORR of 24.5% (95% CI,

16.7–33.8) observed in this phase IV trial support the outcomes of the pivotal phase III trial of sunitinib in pNETs and confi rm its activity in this setting.

n OS data were not mature at the time of the primary analysis.

n AEs were consistent with the known safety profi le of sunitinib.

n The study confi rmed sunitinib is an effi cacious and safe treatment option in progressive, locally advanced and/or metastatic, well-differentiated, unresectable pNETs.

The Effi cacy and Safety of Sunitinib in Patients With Advanced Well-Differentiated Pancreatic Neuroendocrine TumorsEric Raymond1, Matthew H Kulke2, Shukui Qin3, Michael Schenker4, Antonio Cubillo5, Wenhui Lou6, Jiri Tomasek7, Espen Thiis-Evensen8, Jianming Xu9, Karoly Racz10, Adina E Croitoru11, Mustafa Khasraw12, Eva Sedlackova13, Ivan Borbath14, Paul Ruff15, Paul E Oberstein16, Tetsuhide Ito17, Kathrine C Fernandez18, Brad Rosbrook19, Nicola Fazio20 1Paris Saint-Joseph Hospital Group, Paris, France; 2Dana-Farber Cancer Institute, Boston, MA, USA; 3PLA Cancer Center of Nanjing Bayi Hospital, Nanjing, China; 4Centrul de Oncologie Sf. Nectarie, Oncologie Medicala, Craiova, Romania; 5Hospital Universitario Madrid Sanchinarro, Centro Integral Oncológico Clara Campal, Madrid, Spain; 6Zhongshan Hospital, Fudan University, Shanghai, China; 7Masaryk Memorial Cancer Institute, Faculty of Medicine, Masaryk University, Brno, Czech Republic; 8Oslo University Hospital, Department of Gastroenterology, Rikshospitalet, Oslo, Norway; 9No.307 Hospital, Academy of Military Medical Sciences, Beijing, China; 10Semmelweis University, Faculty of Medicine, 2nd Department of Internal Medicine, Budapest, Hungary; 11Fundeni Clinical Institute, Department of Medical Oncology, Bucharest, Romania; 12Andrew Love Cancer Center, Geelong Hospital, Victoria, Australia; 13Všeobecné Fakultní Nemocnice v Praze Onkologická Klinika, Praha, Czech Republic; 14Cliniques Universitaires Saint-Luc, King Albert II Institute Cancerology and Hematology, Brussels, Belgium; 15University of the Witwatersrand, Faculty of Health Sciences, Johannesburg, South Africa; 16Columbia University Medical Center, Division of Hematology/Oncology, New York, NY, USA; 17Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; 18Pfi zer Inc, Cambridge, MA, USA; 19Pfi zer Inc, San Diego, CA, USA; 20IEO, European Institute of Oncology, Unit of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, Milan, Italy

380

REFERENCES1. American Cancer Society. Cancer Facts & Figures 2016. Atlanta, GA.2. Raymond E, et al. N Engl J Med 2011;364:501-13.3. Faivre S, et al. Ann Oncol 2016; Nov 10 [Epub ahead of print]. 4. Sutent® (sunitinib malate) prescribing information. New York, NY: Pfi zer Inc; 2015.

ACKNOWLEDGMENTSWe thank the participating patients and their families, as well as the global network of research nurses, trial coordinators, and operations staff for their contributions, and investigators who participated in this trial, including: M. Michael (Australia); X. Yu, Y. Shen and L. Jia (China); P. Hammel (France); S. Skrikhande (India); C. Morizane (Japan); J. Sufl iarsky (Slovakia); and G. Khan (United States). This study was sponsored by Pfi zer. Medical writing support was provided by Mariko Nagashima, PhD, and Anne Marie Reid, PhD, of Engage Scientifi c Solutions, and was funded by Pfi zer.

For information on this poster, contact x xxxxx at xxxx.xxxx

Copyright © 2016

INTRODUCTION • Pancreatic neuroendocrine tumors (pNETs) are rare tumors. Of an estimated

53,070 new cases of pancreatic cancers expected in the US in 2016, pNETs account for fewer than 5%.1

• Due to their relatively indolent nature, the majority of patients are diagnosed with metastatic pNETs, for whom treatment options are limited.

• pNETs are highly vascularized tumors; aberrant expression of vascular endothelial growth factor (VEGF) and its receptors, which play vital roles in tumor angiogenesis, has been observed.

• Sunitinib, a multi-targeted tyrosine kinase inhibitor (TKI) of angiogenesis, showed a signifi cant increase in progression-free survival (PFS) over placebo in well-differentiated, advanced and/or metastatic pNETs in a pivotal phase III clinical trial (ClinicalTrials.gov NCT00428597).2

• The trial was terminated early after more-serious adverse events (AEs) and deaths were observed in the placebo arm and a difference in PFS favored the sunitinib arm (hazard ratio [HR] 0.42; 95% confi dence interval [CI], 0.26–0.66; P<0.001; median: 11.4 vs 5.5 months).

– The median (95% CI) 5-year overall survival (OS) was 38.6 (25.6–56.4) months for sunitinib and 29.1 (16.4–36.8) months for placebo (HR 0.73; 95% CI, 0.50–1.06; P=0.094), with 69% of placebo patients having crossed over to sunitinib.3

• In 2010 and 2011, sunitinib was approved by the European Medicines Agency and US Food and Drug Administration (FDA), respectively, for the treatment of patients with progressive, locally advanced and/or metastatic, well-differentiated, unresectable pNETs.4

OBJECTIVES • This phase IV clinical trial (ClinicalTrials.gov NCT01525550) was conducted

as post-approval commitments to the FDA and other regulatory agencies to confi rm the effi cacy and safety of sunitinib in advanced and/or metastatic, well-differentiated, unresectable pNETs.

METHODSStudy Design

• This multinational, single-arm, open-label, phase IV clinical trial is ongoing.

• The primary endpoint is investigator-assessed PFS per the Response Evaluation Criteria in Solid Tumors (RECIST) v1.0.

• Secondary endpoints include PFS assessed by the independent radiological review, time to tumor progression (TTP), objective response rate (ORR), OS, and safety.

Key Eligibility Criteria • Histologically or cytologically confi rmed, well-differentiated, unresectable or

metastatic pNETs with documented progression within 12 months of study enrollment.

• Not amenable to surgery, radiation, or combined modality therapy with curative intent.

• Presence of ≥1 measurable target lesion per RECIST v1.0.

• Eastern Cooperative Oncology Group performance status (ECOG PS) 0 or 1.

• No pre-existing uncontrolled hypertension (ie, blood pressure >150/100 mmHg despite medical therapy).

• No prior treatment with TKIs, anti-VEGF, or other angiogenesis inhibitors.

Treatment and Assessments • Patients received 37.5 mg sunitinib orally once a day on a continuous daily-dosing

regimen.

– Sunitinib dose could be increased to 50 mg daily any time after 8 weeks of treatment initiation in patients without treatment response who experienced only grade 1 or lower nonhematologic or grade 2 or lower hematologic treatment-related AEs.

– Dose could be temporarily interrupted or reduced to 25 mg daily to manage severe toxicity.

• Somatostatin analogs for control of symptoms were permitted at the investigator’s discretion.

• Radiologic tumor assessments were conducted at screening, Cycle 2 Day 1, Cycle 3 Day 1, and every 2 cycles thereafter; tumor responses were evaluated per RECIST v1.0 criteria.

• Safety was monitored throughout the study and AEs were graded according to the National Cancer Institute Common Terminology Criteria for Adverse Events v3.0.

Statistical Analyses • More than 80 patients were to be enrolled: 40 treatment-naïve (defi ned as

those who never received systemic antitumor therapy; somatostatin analogs for symptomatic control were allowed) and 40 previously treated patients.

• Median PFS and 95% CI were estimated using the Kaplan–Meier method for the entire population as well as separately for treatment-naïve and previously treated patients.

• Descriptive statistics were used to summarize other parameters.

RESULTSPatients

• Of 123 patients screened, 106 (61 treatment-naïve and 45 previously treated) were enrolled at 25 centers in 15 countries (Figure 1).

Figure 1: Trial Profi le

123 patients screened

106 patients enrolled

61 txt-naïve patients treated 45 previously treated patients treated

39 previously treated patients discontinued txt 23 Objective progression/relapse 8 Adverse events 1 Refusal of txt for reason other than AE 2 Global health deterioration 1 Death 4 Other

43 txt-naïve patients discontinued txt 26 Objective progression/relapse 6 Adverse events 4 Refusal of txt for reason other than AE 2 Global health deterioration 1 Death 4 Other

Efficacy analysis: 61 txt-naïve patientsSafety analysis: 61 txt-naïve patients

45 previously treated patients45 previously treated patients

AE=adverse event; Txt=treatment

• Patient demographics and baseline characteristics are summarized in Table 1.

• More than half (n=64/106, 60.4%) of patients had a nonfunctioning tumor and 17.9% had a functioning tumor (unknown for 21.7% of patients).

• In regard to prior locoregional treatment, 18.9% of patients had trans-arterial chemoembolization; radiofrequency ablation (3.8%); or trans-arterial embolization, percutaneous injections, or microwave ablation (2.8% each).

• Median (range) treatment duration in the total population was 11.7 months (0.2–40.3): 12.2 months (0.2–35.9) in treatment-naïve vs 10.2 months (0.5–40.3) in previously treated patients.

Effi cacy • Median PFS as assessed by investigators was 13.2 months (95% CI, 10.9–16.7);

median PFS was similar in treatment-naïve and previously treated patients (13.2 months [95% CI, 7.4–16.8] and 13.0 months [95% CI, 9.2–20.4], respectively; Figure 2).

• Median PFS as assessed by independent radiological review was 11.1 months (95% CI, 7.4–16.6), in treatment-naive patients 11.1 months (95% CI, 5.5–16.7), and in previously treated patients 9.5 months (95% CI, 7.4–18.4).

– 7 patients were censored due to lack of adequate baseline assessments.

Table 1: Demographics and Patient Baseline Characteristics

CharacteristicsTreatment-naïve

n=61

Previously Treated

n=45Total

N=106

Age, years, mean (SD) 55.4 (8.9) 53.5 (9.1) 54.6 (9.0)Gender, n (%) Male 30 (49.2) 33 (73.3) 63 (59.4) Female 31 (50.8) 12 (26.7) 43 (40.6) Race, n (%) White 32 (52.5) 35 (77.8) 67 (63.2) Black 2 (3.3) 0 2 (1.9) Asian 27 (44.3) 10 (22.2) 37 (34.9)ECOG PS, n (%) 0 39 (63.9) 29 (64.4) 68 (64.2) 1 22 (36.1) 16 (35.6) 38 (35.8)No. of involved disease sites,* n (%) 1 24 (39.3) 9 (20.0) 33 (31.1) 2 19 (31.1) 22 (48.9) 41 (38.7) 3 11 (18.0) 8 (17.8) 19 (17.9) 4 3 (4.9) 3 (6.7) 6 (5.7) >4 4 (6.6) 3 (6.7) 7 (6.6) Tumor site,* n (%) Liver 57 (93.4) 41 (91.1) 98 (92.5) Pancreas 22 (36.1) 25 (55.6) 47 (44.3) Lymph node, distant 16 (26.2) 13 (28.9) 29 (27.4) Lymph node, regional 13 (21.3) 7 (15.6) 20 (18.9) Lung 3 (4.9) 3 (6.7) 6 (5.7) Other 10 (16.4) 10 (22.2) 20 (18.9)Prior systemic chemotherapy, n (%) Any 0 45 (100) 45 (42.5) Neoadjuvant 0 2 (4.4) 2 (1.9) Adjuvant 0 4 (8.9) 4 (3.8) Advanced/metastatic 0 39 (86.7) 39 (36.8)Prior SSA,† n (%) 24 (39.3) 27 (60.0) 51 (48.1)Ki-67 index, mean (SD) 6.7 (5.0) 8.4 (7.2) 7.4 (6.0)* Included both target and nontarget sites; sites with multiple lesions were counted once.† Patients with regimens that consist only of somatostatin analogs were considered treatment-naïve.ECOG PS, Eastern Cooperative Oncology Group performance status; SD=standard deviation; SSA=somatostatin analogs

Figure 2: Kaplan–Meier Estimates of PFS in Treatment-Naïve and Previously Treated Patients With pNETs, Assessed by Investigators

PFS

Dis

trib

utio

n Fu

nctio

n

1.0

0.9

0.8

0.7

0.6

0.5

0.4

0.3

0.2

0.1

0.00 5 10 15 20 25

Time (Months)No. at risk:

Treatment-naïvePreviously treated

6145

4129

3220

1410

88

64

04

02

01

00

30 35 40 45

Treatment-naïve 61 37 13.2 (7.4–16.8)Previously treated 45 28 13.0 (9.2–20.4)

n Events mPFS (95% CI), mo

CI=confi dence interval; mPFS=median progression-free survival; PFS=progression-free survival; pNETs=pancreatic neuroendocrine tumors

• ORR was 24.5% (95% CI, 16.7–33.8) according to the investigator assessment (Table 2).

Table 2: Best Observed Response by RECIST, Assessed by Investigators

Treatment-naïve

n=61

Previously Treated

n=45Total

N=106

Best overall response, n (%)

Complete response 2 (3.3) 1 (2.2) 3 (2.8)

Partial response 11 (18.0) 12 (26.7) 23 (21.7)

Stable disease 40 (65.6) 29 (64.4) 69 (65.1)

Progressive disease 7 (11.5) 2 (4.4) 9 (8.5)

Indeterminate 1 (1.6) 1 (2.2) 2 (1.9)

ORR,* n (%) 13 (21.3) 13 (28.9) 26 (24.5)

95% CI 11.9–33.7 16.4–44.3 16.7–33.8* Complete response + partial response. CI=confi dence interval; ORR=objective response rate; RECIST=Response Evaluation Criteria in Solid Tumors

Median TTP was 14.5 months (95% CI, 11.0–16.7); median TTP in treatment-naïve and previously treated patients was similar (14.8 [95% CI, 7.5–16.8] and 14.5 [95% CI, 9.2–20.4] months, respectively).

• OS data were not mature at the time of data cutoff date (Mar 19, 2016); 29 (27.4%) patients had died and median OS was 37.8 months (95% CI, 33.0–not estimable).

Safety • Most-common treatment-emergent, all-grade AEs experienced by all patients treated

with sunitinib included neutropenia, diarrhea, and leukopenia (Table 3). – No major differences were observed in the incidence of AEs reported by treatment-

naïve vs previously treated patients, except dyspepsia, nausea, and neutropenia. • Percentage of treatment-naïve and previously treated patients who experienced

Grade 3 or 4 AEs was comparable; serious AEs were also comparable: 24.6% (n=15) vs 24.4% (n=11), respectively.

• 15 (24.6%) treatment-naïve and 5 (11.1%) previously treated patients had sunitinib dose reductions due to AEs; 8 (13.1%) and 10 (22.2%) patients, respectively, discontinued treatment due to AEs.

Please scan this Quick Response (QR) code with your smartphone app to view an electronic version of this poster. If you don’t have a smartphone, access the poster via the internet at:

http://congress-download.pfi zer.com/asco_gi_2017_american_society_of_clinical_oncology_gastrointestinal_cancers_symposium_295_sutent_raymond_380.html

Copies of this poster obtained through QR Code are for personal use only and may not be reproduced without permission from ASCO® and the author of this poster.

Table 3: Treatment-Emergent, All-Causality Adverse Events

Adverse Events,* n (%)

Treatment-naïven=61

Previously Treatedn=45

TotalN=106

All Grades

Grade 3/4

All Grades

Grade 3/4

All Grades

Grade 3/4

Neutropenia 37 (60.7) 13 (21.3) 22 (48.9) 10 (22.2) 59 (55.7) 23 (21.7)Diarrhea 32 (52.5) 7 (11.5) 22 (48.9) 3 (6.7) 54 (50.9) 10 (9.4)Leukopenia 25 (41.0) 4 (6.6) 21 (46.7) 3 (6.7) 46 (43.4) 7 (6.6)Fatigue 19 (31.1) 1 (1.6) 14 (31.1) 0 (0.0) 33 (31.1) 1 (0.9)Hand–foot syndrome 19 (31.1) 5 (8.2) 14 (31.1) 2 (4.4) 33 (31.1) 7 (6.6)Thrombocytopenia 18 (29.5) 6 (9.8) 14 (31.1) 2 (4.4) 32 (30.2) 8 (7.5)Hypertension 16 (26.2) 4 (6.6) 11 (24.4) 2 (4.4) 27 (25.5) 6 (5.7)Abdominal pain 16 (26.2) 2 (3.3) 10 (22.2) 3 (6.7) 26 (24.5) 5 (4.7)Dysgeusia 14 (23.0) 0 (0.0) 11 (24.4) 0 (0.0) 25 (23.6) 0 (0.0)Nausea 11 (18.0) 0 (0.0) 14 (31.1) 1 (2.2) 25 (23.6) 1 (0.9)Dyspepsia 7 (11.5) 0 (0.0) 14 (31.1) 0 (0.0) 21 (19.8) 0 (0.0)Headache 12 (19.7) 0 (0.0) 9 (20.0) 0 (0.0) 21 (19.8) 0 (0.0)Stomatitis 13 (21.3) 2 (3.3) 6 (13.3) 1 (2.2) 19 (17.9) 3 (2.8)Vomiting 8 (13.1) 1 (1.6) 10 (22.2) 1 (2.2) 18 (17.0) 2 (1.9)Asthenia 10 (16.4) 0 (0.0) 7 (15.6) 2 (4.4) 17 (16.0) 2 (1.9)Abdominal pain upper 5 (8.2) 1 (1.6) 7 (15.6) 1 (2.2) 12 (11.3) 2 (1.9)ALT increased 2 (3.3) 1 (1.6) 9 (20.0) 0 (0.0) 11 (10.4) 1 (0.9)AST increased 4 (6.6) 1 (1.6) 7 (15.6) 1 (2.2) 11 (10.4) 2 (1.9)Constipation 3 (4.9) 0 (0.0) 7 (15.6) 0 (0.0) 10 (9.4) 0 (0.0)Dizziness 3 (4.9) 0 (0.0) 7 (15.6) 0 (0.0) 10 (9.4) 0 (0.0)Hypophosphatemia 2 (3.3) 2 (3.3) 7 (15.6) 3 (6.7) 9 (8.5) 5 (4.7)Myalgia 2 (3.3) 0 (0.0) 7 (15.6) 0 (0.0) 9 (8.5) 0 (0.0)Adverse events are listed by highest to lowest % for Total/All Grades patients.* Adverse events reported by ≥15% in any treatment group per MedDRA criteria.ALT=alanine aminotransferase; AST=aspartate aminotransferase; MedDRA=Medical Dictionary for Regulatory Activities