*Bert Bakker was an employee of Novartis Pharmaceuticals Corporation until June 06, 2014.

Similar documents
TRACTAMENT ONCOLÒGIC DELS TUMORS NEUROENDOCRINS METASTÀSICS

NET εντέρου Τι νεότερο/ Νέες μελέτες. Μαντώ Νικολαΐδη παθολόγος-ογκολόγος ΜΗΤΕΡΑ

Tumor Growth Rate (TGR) A New Indicator of Antitumor Activity in NETS? IPSEN NET Masterclass Athens, 12 th November 2016

Gastrointestinal Neuroendocrine Tumors: A Closer Look at the Characteristics of These Diverse Tumors

Recent developments of oncology in neuroendocrine tumors (NETs)

Jaume Capdevila, MD GI and Endocrine Tumor Unit Vall d Hebron University Hospital Developmental Therapeutics Unit Vall d Hebron Institute of Oncology

Everolimus Plus Octreotide Long-Acting Repeatable in Patients With Advanced Lung Neuroendocrine Tumors

MEDICAL MANAGEMENT OF METASTATIC GEP-NET

Review of Gastrointestinal Carcinoid Tumors: Latest Therapies

Octreotide LAR in neuroendocrine tumours a summary of the experience

Nuevas alternativas en el manejo de TNE avanzados

Le target therapy nei Tumori Neuroendocrini

QOL Improvements in NETTER-1 Phase III Trial in Patients With Progressive Midgut Neuroendocrine Tumors

Cutting Edge Treatment of Neuroendocrine Tumors

Cutting Edge Treatment of Neuroendocrine Tumors

Long-Term Management of a Patient with Well-Differentiated Pulmonary Neuroendocrine Carcinoma: A Case Report

Teresa Alonso Gordoa Servicio Oncología Médica Hospital Universitario Ramón y Cajal

2015: Year in Review Results of Recent Trials

ENETS Consensus Guidelines for the Standard of Care in

Lanreotide in Metastatic Enteropancreatic Neuroendocrine Tumors

Systematic Review of the Role of Targeted Therapy in Metastatic Neuroendocrine Tumors

NICaN Pancreatic Neuroendocrine Tumour SACT protocols. 1.0 Dr M Eatock Final version issued

Antiangiogenics are effective treatments in NETs

NET ΠΝΕΥΜΟΝΑ: τι νεότερο / νέες μελέτες

Toward More Aggressive Management of Neuroendocrine Tumors: Current and Future Perspectives

CLICK TO GO BACK TO KIOSK MENU

OPTIMISING OUTCOMES IN GASTROINTESTINAL NEUROENDOCRINE TUMOURS

NET und NEC. Endoscopic and oncologic therapy

Evaluation and Management of Neuroendocrine Tumors

Selection of Appropriate Treatment

Prior Authorization Review Panel MCO Policy Submission

Sandostatin LAR. Sandostatin LAR (octreotide acetate) Description

GI CARCINOID Dr Mussawar Iqbal Consultant Oncologist Hull and East Yorkshire Hospitals NHS Trust

Pancreatic NeuroEndocrine Tumors. Prof Eric Van Cutsem, MD, PhD Gastroenterology/Digestive Oncology Leuven, Belgium

The Antiproliferative Role of Lanreotide in Controlling Growth of Neuroendocrine Tumors: A Systematic Review

United States-based practice patterns and resource utilization in advanced neuroendocrine tumor treatment

Pembrolizumab for Patients With PD-L1 Positive Advanced Carcinoid or Pancreatic Neuroendocrine Tumors: Results From the KEYNOTE-028 Study

Neuroendocrine Tumors: Treatment Updates Highlights from the 2013 ASCO Annual Meeting. Chicago, IL, USA; May 30 - June 4, 2013

Health-Related Quality of Life in Patients With Progressive Midgut Neuroendocrine Tumors Treated With

PHASE II CLINICAL TRIAL OF PASIREOTIDE LAR IN PATIENTS WITH METASTATIC NEUROENDOCRINE TUMORS

Updates in Pancreatic Neuroendocrine Carcinoma Highlights from the 2010 ASCO Annual Meeting. Chicago, IL, USA. June 4-8, 2010

Guideline A Quality Initiative of the Program in Evidence-Based Care (PEBC), Cancer Care Ontario (CCO)

Long-term safety and efficacy of vismodegib in patients with advanced basal cell carcinoma (BCC): 24-month update of the pivotal ERIVANCE BCC study

Background. Capdevila J, et al. Ann Oncol. 2018;29(Suppl 8): Abstract 1307O. 1. Dasari A, et al. JAMA Oncol. 2017;3(10):

Evidenze cliniche nel trattamento del RCC

NET del pancreas ben differenziato: la terapia oncologica. Alfredo Berru: Università degli Studi di Brescia Azienda Ospedaliera Spedali Civili Brescia

Negative Trials in RCC: Where Did We Go Wrong? Can We Do Better?

GEP NET: algoritmo terapeutico. Dottor Nicola Fazio

Primary Endpoint The primary endpoint is overall survival, measured as the time in weeks from randomization to date of death due to any cause.

J Clin Oncol 26: by American Society of Clinical Oncology INTRODUCTION

sunitinib 12.5mg, 25mg, 37.5mg, 50mg hard capsules (Sutent ) SMC No. (698/11) Pfizer Limited

Articles. Funding Novartis Pharmaceuticals.

NeuroEndocrine Tumors Diagnostic and therapeutic challenges: introduction

PTAC meeting held on 5 & 6 May (minutes for web publishing)

Advanced typical and atypical carcinoid tumours of the lung: management recommendations

Recent Advances in Gastrointestinal Cancers

WHAT TO EXPECT IN 2015? - Renuka Iyer, MD Associate Professor of Medicine, University at Buffalo Associate Professor of Oncology, Roswell Park Cancer

Clinical Development in the Treatment of Carcinoid Syndrome. Pablo Lapuerta, MD Executive Vice President and Chief Medical Officer

Oberndofer 1907 Illeal Serotonin Secreting Tumor Carcinoid (Karzinoide)

Long Term Results in GIST Treatment

EXOCRINE: 93% Acinar Cells Duct Cells. ENDOCRINE: 5% Alpha Cells Beta Cells Delta Cells Others

Hot of the press. Γρηγόριος Καλτσάς MD FRCP Καθηγητής Παθολογίας Ενδοκρινολογίας ΕΚΠΑ

EVIDENCE IN BRIEF OVERALL CLINICAL BENEFIT

Disclosure of Relevant Financial Relationships

An Open-Label Phase Ib/II Study of Sulfatinib in Patients with Advanced Neuroendocrine Tumors (NCT )

Gastroenteropancreatic Neuroendocrine Tumors with Liver Metastases in Korea: A Clinicopathological Analysis of 72 Cases in a Single Institute

SIRT in the Management of Metastatic Neuroendocrine Tumors

Scottish Medicines Consortium

PNET 3/7/2015. GI and Pancreatic NETs. The Postgraduate Course in Breast and Endocrine Surgery. Decision Tree. GI and Pancreatic NETs.

An Immunotherapy Clinical Trial for NETs

Chibueze Onyemkpa 1, Alan Davis 1, Michael McLeod 1, Tolutope Oyasiji 1,2. Original Article

Phase 3 Trial of 177 Lu-Dotatate for Midgut Neuroendocrine Tumors

SOMATOSTATIN RECEPTORS IN HEPATOCELLULAR CARCINOMA. Marie LEQUOY Saint-Antoine Hospital, Department of Hepatology, Paris, France

Schweizer Register für Neuroendokrine Tumore Le Registre Suisse des tumeurs neuroendocrines Registro Svizzero per i Tumori Neuroendocrini

This clinical study synopsis is provided in line with Boehringer Ingelheim s Policy on Transparency and Publication of Clinical Study Data.

Chemotherapy and Immunotherapy in Combination Non-Small Cell Lung Cancer (NSCLC)

Real-World Treatment Patterns for Lung Neuroendocrine Tumors: A Claims Database Analysis

Recent advances in the management of metastatic breast cancer in older adults


Short Study Report for Health Authorities

SUPPLEMENTARY INFORMATION

Diagnosis and workup of 522 consecutive patients with neuroendocrine neoplasms in Switzerland

EVIDENCE IN BRIEF OVERALL CLINICAL BENEFIT

THERAPEUTIC RADIOPHARMACEUTICALS

Therapeutic Radiopharmaceuticals in Oncology

LONDON CANCER NEW DRUGS GROUP RAPID REVIEW. Erlotinib for the third or fourth-line treatment of NSCLC January 2012

Everolimus, lutetium-177 DOTATATE and sunitinib for treating unresectable or metastatic neuroendocrine tumours with disease progression MTA

TUMORES NEUROENDOCRINOS. Miguel Navarro. Salamanca

Synopsis (C0168T60) Associated with Module 5.3 of the Dossier

OUR EXPERIENCES WITH ERLOTINIB IN SECOND AND THIRD LINE TREATMENT PATIENTS WITH ADVANCED STAGE IIIB/ IV NON-SMALL CELL LUNG CANCER

Amreen Husain, 10 Eric P. Winer, 11 Sylvia Adams, 12 Peter Schmid 13

lutetium ( 177 Lu) oxodotreotide 370MBq/mL solution for infusion (Lutathera ) SMC No 1337/18 Advanced Accelerator Applications

Horizon Scanning in Oncology

Indication for- and timing of cytoreductive nephrectomy Kidney- and bladder cancer: Immunotherapy

symposium article Gastrointestinal neuroendocrine tumors K. E. Öberg* introduction and epidemiology diagnosis symposium article biochemical markers

This clinical study synopsis is provided in line with Boehringer Ingelheim s Policy on Transparency and Publication of Clinical Study Data.

Comprehensive treatment of a functional pancreatic neuroendocrine tumor with multifocal liver metastases

Evolving Paradigms in HER2+ MBC: Strategies for Individualizing Therapy with Available Agents

Pharmacy Prior Authorization Somatostatin Analogs Clinical Guideline

National Horizon Scanning Centre. Imatinib (Glivec) for adjuvant therapy in gastrointestinal stromal tumours. August 2008

Transcription:

Page 1 of 20 Accepted Preprint first posted on 15 September 2015 as Manuscript ERC-15-0314 1 2 Efficacy of Octreotide LAR in Neuroendocrine Tumors: RADIANT-2 Placebo Arm Post Hoc Analysis 3 4 Authors: 5 6 Jonathan R Strosberg 1, James C Yao 2, Emilio Bajetta 3, Mounir Aout 4, Bert Bakker 5*, John D Hainsworth 6, Philippe B Ruszniewski 7, Eric Van Cutsem 8, Kjell Öberg 9 and Marianne E Pavel 10 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 Affiliations: 1 Department of Medicine, Moffitt Cancer Center, Tampa, Florida, USA 2 Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA 3 Istituto di Oncologia, Policlinico di Monza, Monza, Italy 4 Novartis International AG, Basel, Switzerland 5 Novartis Pharmaceuticals Corporation, East Hanover, New Jersey, USA 6 Sarah Cannon Research Institute, Nashville, Tennessee, USA 7 University of Paris VII and Beaujon Hospital, Paris, France 8 Digestive Oncology, University Hospitals Gasthuisberg/Leuven and KULeuven, Leuven, Belgium 9 Uppsala University Hospital, Uppsala, Sweden 10 Department of Hepatology and Gastroenterology, Charité-Universitätsmedizin Berlin/Campus Virchow Klinikum, Berlin, Germany *Bert Bakker was an employee of Novartis Pharmaceuticals Corporation until June 06, 2014. 23 Page 1 of 13 Copyright 2015 by the Society for Endocrinology.

Page 2 of 20 24 25 26 Correspondence should be addressed to Jonathan R Strosberg, Department of Medicine, Moffitt Cancer Center, Tampa, Florida, USA. Phone: 813 745-3636; Fax: 813 745-7229 Email: Jonathan.Strosberg@moffitt.org 27 28 Short title: Efficacy of octreotide LAR in advanced NET 29 30 31 Keywords: neuroendocrine tumors, octreotide LAR, progression-free survival, somatostatin analogues 32 33 Word count: ~3000 (Maximum, 5000) 34 Figures: 2 (Maximum, 10) 35 Tables: 2 (No limit given by the journal) 36 37 Earlier presentations: 38 39 40 Presented at the Gastrointestinal Cancers Symposium (ASCO GI), January 16 to 18, 2014, San Francisco, California, and at the European Neuroendocrine Tumor Society (ENETS), March 5 to 7, 2014, Barcelona, Spain. Page 2 of 13

Page 3 of 20 41 Abstract (250; maximum allowed, 250) 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 Somatostatin analogues (SSA) have demonstrated antiproliferative activity in addition to efficacy for carcinoid symptom control in functional neuroendocrine tumors (NET). A post hoc analysis of the placebo arm of the RADIANT (RAD001 In Advanced Neuroendocrine Tumors)-2 study was conducted to assess the efficacy of octreotide long-acting repeatable (LAR) on progression-free survival (PFS) and overall survival (OS) estimated using the Kaplan-Meier method. Out of 213 patients randomized to placebo plus octreotide LAR in RADIANT-2, 196 patients with foregut, midgut, or hindgut NET were considered for present analysis. Of these, 41 patients were SSA-treatment naïve and 155 had received SSA therapy before study entry. For SSA-naïve patients, median PFS by adjudicated central review was 13.6 (95% confidence interval [CI], 8.2 22.7) months. For SSA-naïve patients with midgut NET (n = 24), median PFS was 22.2 (95% CI 8.3 29.5) months. For patients who had received SSA previously, the median PFS was 11.1 (95% CI, 8.4 14.3) months. Among the SSA-pretreated patients who had midgut NET (n = 119), the median PFS was 12.0 (95% CI, 8.4 19.3) months. Median OS was 35.8 (95% CI, 32.5 48.9) months for patients in the placebo plus octreotide LAR arm; 50.6 (36.4 not reached) months for SSA-naïve patients and 33.5 (95% CI, 27.5 44.7) months for those who had received prior SSA. This post hoc analysis of the placebo arm of the large phase 3 RADIANT-2 study provides data on PFS and OS among patients with progressive NET treated with octreotide therapy. 58 59 60 Trial registration: ClinicalTrials.gov number, NCT00412061 (http://clinicaltrials.gov/show/nct00412061) 61 Page 3 of 13

Page 4 of 20 62 63 64 65 66 67 68 69 70 71 72 73 74 75 76 77 78 79 80 81 82 83 84 85 86 87 Introduction Neuroendocrine tumors (NET) are a rare, heterogeneous group of tumors that arise from neuroendocrine cells throughout the body and are capable of producing various bioactive substances such as peptides and amines (Vinik, et al. 2010; Yao, et al. 2008). Somatostatin analogues (SSA) can effectively suppress the secretion of serotonin and other bioactive substances and relieve associated symptoms in patients with functional NET. In addition to symptom control, therapeutic management for patients with advanced, unresectable NET aim to induce tumor regression or stabilization and improve survival (Pavel, et al. 2012). Octreotide and lanreotide have demonstrated antiproliferative activity in pivotal phase 3 trials (Caplin, et al. 2014; Rinke, et al. 2009). In the landmark PROMID (Placebo controlled, double-blind, prospective, Randomized study of the effect of Octreotide LAR in the control of tumor growth in patients with metastatic neuroendocrine MIDgut tumors; N = 85) study, octreotide long-acting repeatable (LAR; 30 mg intramuscularly every 28 days) demonstrated antitumor activity in patients with newly diagnosed advanced metastatic NET of midgut origin. The median time to tumor progression was 14.3 months with octreotide LAR versus 6.0 months with placebo (hazard ratio [HR], 0.34; 95% confidence interval [CI], 0.20 0.59; P = 0.000072) using the World Health Organization (WHO) response criteria for tumor evaluation (Rinke et al. 2009). In the more recent phase 3 CLARINET study (N = 204), patients with advanced NET treated with lanreotide autogel (120 mg via deep subcutaneous injection every 28 days) also had improvement in progressionfree survival (PFS) when compared with placebo (PFS, not reached [NR] versus 18 months; HR, 0.47; 95% Cl, 0.30 0.73; P < 0.001) (Caplin et al. 2014). In the large phase 3 RADIANT (RAD001 In Advanced Neuroendocrine Tumors)-2 study, everolimus in combination with octreotide LAR showed a clinically meaningful 5.1-month increase in median PFS, compared with placebo plus octreotide LAR treatment in patients with advanced NET associated with carcinoid syndrome, although the study did not meet its prespecified endpoint (median PFS, 16.4 versus 11.3 months; HR, 0.77; 95% CI, 0.59 1.00; P = 0.026, prespecified P = 0.0246) (Pavel, et al. 2011). Page 4 of 13

Page 5 of 20 88 89 90 This exploratory post hoc analysis of the placebo plus octreotide LAR arm of the RADIANT-2 study was conducted to assess the effect of octreotide LAR on survival endpoints in a heterogeneous group of patients with advanced NET. 91 92 93 94 95 96 97 98 99 100 101 102 103 104 105 106 107 108 109 110 111 Patients and methods Study design RADIANT-2 was a multinational, multicenter, randomized, double-blind, placebo-controlled, phase 3 study (ClinicalTrials.gov number, NCT00412061); the detailed study design has been described earlier (Pavel et al. 2011). Patients were randomly assigned (1:1) to receive everolimus (10 mg/d) plus octreotide LAR (30 mg intramuscularly [IM] every 28 days) or placebo plus octreotide LAR (30 mg IM every 28 days). The study treatment continued until disease progression, development of unacceptable adverse events, withdrawal of patient consent, or primary analysis (cutoff date, April 2, 2010). During the double-blind phase, patients were permitted to crossover from the placebo plus octreotide LAR arm to the open-label everolimus plus octreotide LAR arm upon disease progression according to the Response Evaluation Criteria in Solid Tumors (RECIST, version 1.0) (Therasse, et al. 2000). A blind was broken for these patients. At the end of the double-blind phase, all ongoing patients were unblinded and were allowed to transition to open-label everolimus plus octreotide LAR at the discretion of treating physician. All patients who switched to the open-label everolimus plus octreotide LAR (during the double-blind phase or after unblinding) discontinued everolimus treatment upon disease progression. These patients were then followed-up monthly for survival information (until June 13, 2013, data cutoff date for the overall survival [OS] analysis). The patients enrolled in the placebo plus octreotide LAR arm were the focus of the present post hoc analysis (Fig. 1). 112 113 Ethics 114 115 The study protocol and all amendments were reviewed and approved by independent ethics committees or institutional review boards for all participating centers. The study conformed to the Good Page 5 of 13

Page 6 of 20 116 117 Clinical Practice guidelines and was conducted in the spirit of the principles set in the Declaration of Helsinki. Written informed consent was obtained from all patients prior to participation. 118 119 120 121 122 123 124 125 126 127 128 129 130 131 132 133 134 135 Patient population The patient population has been described earlier in detail (Pavel et al. 2011). Briefly, adult patients (age, 18 years) with low- or intermediate-grade advanced (unresectable locally advanced or distant metastatic) NET, a history of symptoms attributed to carcinoid syndrome (flushing, diarrhea, or both), and disease progression within the past 12 months were eligible. Other key inclusion criteria included the presence of measurable disease according to RECIST, version 1.0 (Therasse et al. 2000), and a WHO performance status (PS) score of 2 with adequate bone marrow, liver, and kidney function. Patients with poorly differentiated or high-grade NET were not eligible. Furthermore, patients were ineligible if they had received cytotoxic chemotherapy, immunotherapy, or radiotherapy within 4 weeks prior to randomization, or prior therapy with mammalian target of rapamycin (mtor) inhibitors (sirolimus, temsirolimus, or everolimus). Patients who had received SSA treatment within 2 weeks prior to randomization were ineligible. Information on the previous SSA dose was not available for all patients. Patients with foregut, midgut, or hindgut NET were considered for this subgroup analysis of placebo plus octreotide LAR arm of the RADIANT-2 study. Tumors originating in the lung, stomach, duodenum, or pancreas were categorized as foregut NET; tumors originating in the small intestine, appendix, proximal colon, or of unknown primary sites were considered to be midgut NET; and tumors with the colon (transverse or distal) or rectum as the primary site were classified as hindgut NET. 136 137 138 139 140 141 142 Statistical analysis In the primary analysis of the RADIANT-2 study, the primary endpoint was PFS, assessed by an adjudicated central radiology review according to RECIST, version 1.0 (Pavel et al. 2011). PFS (data cutoff date, April 2, 2010) was estimated using the Kaplan-Meier method and reported with corresponding 95% CIs. The median PFS was also assessed by local investigators. The secondary endpoint was overall survival (OS; data cutoff date, June 13, 2013). Page 6 of 13

Page 7 of 20 143 144 145 146 147 148 149 For this post hoc analysis, patients were stratified based on previous SSA use and tumor location (foregut, midgut, or hindgut), and the median PFS and OS were calculated in these subgroups. In a multivariate analysis, HRs and 95% CIs were calculated with a Cox proportional hazards model using specified variables selected by stepwise regression. The specified variables included age (<65 or 65 years), sex, race (Caucasian or non-caucasian), WHO PS score (0 or >0), previous octreotide use, baseline chromogranin A level ( 2 upper limit of normal [ULN] or >2 ULN), baseline 5-hydroxyindoleacetic acid level (elevated [>median] or normal [ median]), and primary tumor site. 150 151 152 153 154 155 156 157 158 159 160 161 162 163 164 165 166 Results Patient demographics Among 213 patients assigned to the placebo plus octreotide LAR arm, 196 patients had foregut, midgut, or hindgut NET and were included in the present analysis. Of these, 155 (79%) patients had received previous SSA therapy (previous SSA group), whereas 41 (21%) had not (SSA-naïve group; Table 1). The time since initial diagnosis was longer for patients who received previous SSA therapy; 7% of patients in the previous SSA group had a diagnosis of NET within 6 months prior to study entry compared with 24% of patients in the SSA-naïve group. All patients qualifying for enrollment in the RADIANT-2 study had progressive disease within the past 12 months and history of carcinoid symptoms (diarrhea or flushing). For the 155 patients who had received SSA therapy prior to study entry, the median exposure to prior SSA was 1.8 (range, 0 12.5) years. Of these 155 patients, 142 (92%) received octreotide LAR and the remaining 13 (8%) patients received lanreotide. Seventy-three (51%) of the 142 patients on previous octreotide received a dose of 30 mg every 4 weeks and 68 (48%) patients received a dose <30 mg every 4 weeks; for one patient, information on the prior octreotide dose was missing. 167 168 169 170 Progression-free survival Based on the findings of the adjudicated central assessment for the placebo plus octreotide LAR arm, the median PFS was 13.6 (95% CI, 8.2 22.7) months for SSA-naïve patients and 11.1 (95% CI, Page 7 of 13

Page 8 of 20 171 172 173 174 175 176 177 178 179 180 181 182 183 184 185 186 8.4 14.3) months for patients who had received SSA therapy previously. Among patients with midgut NET, the median PFS was 22.2 (95% CI, 8.3 29.5) months for SSA-naïve patients, and 12.0 (95% CI, 8.4 19.3) months for patients who had prior exposure to SSA. Figure 2 illustrates the median PFS in patients with midgut tumors by prior SSA use as per local investigator and per adjudicated central review. Multivariate analysis for PFS showed that baseline WHO PS of >0 (HR [95% CI], 0.65 [0.44 0.98], P = 0.038) was associated with decreased in PFS; and non-elevated baseline CgA level (HR [95% CI], 2.26 [1.45 3.52], P < 0.001) were associated with increased PFS. Overall survival For patients in the placebo plus octreotide LAR arm of RADIANT-2, the median OS was 35.8 (95% CI, 32.5 48.9) months. For SSA-naïve patients, the median OS was 50.6 (95% CI, lower limit, 36.4; upper limit, not reached) months, compared with 33.5 (95% CI, 27.5 44.7) months for patients who had received previous SSA therapy (Table 2). In multivariate analysis for survival, baseline WHO PS of >0 (HR [95% CI], 0.39 [0.26 0.58], P < 0.001) was associated with a decreased in OS; and non-elevated baseline CgA level (HR [95% CI], 2.08 [1.21, 3.60], P = 0.009), and non-elevated 5-hydroxyindoleacetic acid level (HR [95% CI], 1.69 [1.07, 2.69], P = 0.026) were found to be prognostic of increased OS. 187 188 189 190 191 192 193 194 195 196 197 198 Discussion In addition to antisecretory efficacy, SSA have demonstrated antitumor activity in the phase 3 PROMID and CLARINET studies. The PROMID study evaluated octreotide LAR versus placebo in treatment-naïve, low-grade midgut NET, whereas the CLARINET study evaluated lanreotide autogel versus placebo in a more heterogeneous population of enteropancreatic NET, nearly all of whom had stable disease at the outset (Caplin et al. 2014; Rinke et al. 2009). In contrast, the present post hoc analysis of the placebo plus octreotide LAR arm of the RADIANT-2 study provides the outcomes of patients with progressive NET who were started on long-acting SSA treatment. The CLARINET study allowed patients from placebo arm to receive lanreotide autogel in an open-label extension phase, upon disease progression during the double-blind core phase. Data from the open-label extension phase of the CLARINET study demonstrated a median PFS of 14.0 months after Page 8 of 13

Page 9 of 20 199 200 201 202 203 204 205 206 207 208 209 210 211 212 213 214 215 216 217 218 219 220 221 222 223 224 225 226 switch to lanreotide autogel (Pavel, et al. 2012). Of note, despite differences in the patient population between the RADIANT-2 and CLARINET studies, (Pavel, et al. 2011; Caplin, et al. 2014) the median PFS observed in open-label extension phase of the CLARINET study is similar to that reported in the present analysis The results of this analysis suggest an expected clinical benefit of octreotide treatment for SSA-naïve patients with midgut NET. The data indicated that the median PFS was 22.2 (95% CI 8.3 29.5) months among patients with midgut NET who were SSA-naïve and went on to receive octreotide LAR on the trial. For patients with midgut NET who had been treated with SSA previously, the median PFS was 13.6 (95% CI, 8.2 22.7) months. The findings, furthermore, provide a clearer picture regarding PFS by RECIST criteria in patients with midgut NET treated with SSA. In the PROMID study, the median time to tumor progression in the octreotide LAR arm was 14.3 months, whereas in the CLARINET study, the median PFS was not reached in the lanreotide autogel arm after a median follow-up of 27 months (Caplin et al. 2014; Rinke et al. 2009). A potential explanation for this large disparity in PFS is the differing criteria for progression in the two studies: WHO criteria in PROMID (25% increase in two dimensions) and RECIST criteria in CLARINET (20% increase in one dimension, which corresponds to a 44% increase in two dimensions) (James, et al. 1999; Oxnard, et al. 2012; Therasse, et al. 2005). In our present analysis which used RECIST criteria, the median PFS was 22 months among SSA-naïve patients with midgut NET, all of whom were progressive prior to study entry. This finding from the present subgroup analysis is also aligned with CLARINET data. It is, therefore, increasingly apparent that the median time to tumor progression of 14.3 months in the PROMID study is an underestimation of PFS by RECIST criteria. In other words, when patients with midgut NET are treated with SSA therapy, one can expect a median time to progression of approximately 2 years by RECIST criteria. The results for the final OS analysis of the RADIANT-2 study placebo arm demonstrate longer survival in SSA-naïve patients treated with octreotide LAR. The median OS was 50.6 months among SSA-naïve patients compared with 33.5 months for patients who had received prior SSA treatment. Moreover, among SSA-naïve patients with midgut NET, the median OS was not reached with a median follow-up of 64 months. Notably, in the SSA-naïve group, the proportion of patients with longer duration Page 9 of 13

Page 10 of 20 227 228 229 230 231 232 233 since initial diagnosis was lower than in the group with previous SSA therapy. It is likely that the shorter duration since diagnosis in the SSA-naïve group may have resulted in a longer OS. The complete results for the secondary endpoint of OS will be reported separately. A major limitation of this analysis is the fact that only 41 patients on the octreotide plus placebo arm of the RADIANT-2 study were SSA naïve. As our analysis focuses on this subset of patients, all median PFS and OS figures are associated with large CIs, limiting the reliability of these estimates. The findings warrant further confirmation from larger prospective studies. 234 235 236 237 238 239 240 241 242 243 Conclusions The post hoc analysis of the placebo arm of large phase 3 RADIANT-2 study, for the first time, provides data on PFS and OS from a prospective study among patients with progressive NET treated with octreotide therapy. This post hoc analysis showed that SSA-naïve patients with progressive midgut NET treated with octreotide LAR had a relatively long median PFS of 22.2 months, exceeding the time to tumor progression of 14.3 months observed in the PROMID study. The median PFS assessed by RECIST criteria may represent a more accurate reflection of the true PFS associated with SSA use in midgut NET. 244 Page 10 of 13

Page 11 of 20 245 246 247 248 249 250 251 252 253 254 255 256 Declaration of interest J R Strosberg: Grants and personal fees from Novartis, outside the submitted work. J C Yao: Grants and personal fees from Novartis, outside the submitted work. E Bajetta: Nothing to disclose. M Aout: Employee of Novartis during the conduct of the study. B Bakker: Was a full time employee of Novartis, the sponsor of the study and the post-hoc analysis. J D Hainsworth: Grants from Novartis, during the conduct of the study. P B Ruszniewski: Grants, personal fees and non-financial support from Novartis, Ipsen, Pfizer, outside the submitted work. E V Cutsem: Grants from Novartis, outside the submitted work. K Öberg: Member of Advisory Board for Novartis and received honorarium for lectures from Novartis. M E Pavel: Grants and personal fees from Novartis, outside the submitted work. 257 258 259 Funding This study was funded by Novartis Pharmaceuticals Corporation. 260 261 262 263 264 265 266 267 268 Author contribution statement E V Cutsem, J C Yao and K Öberg contributed in study design. E V Cutsem, J C Yao, J R Strosberg, M E Pavel and P B Ruszniewski were involved in the recruitment of patients. M Aout supported statistical analysis and provided results and data interpretation. J D Hainsworth also contributed in statistical analysis. E Bajetta, J C Yao, J R Strosberg and M E Pavel were involved in data interpretation. B Bakker, J C Yao, J D Hainsworth and J R Strosberg contributed in writing of the manuscript content. B Bakker, E Bajetta, E V Cutsem, J C Yao, J D Hainsworth, J R Strosberg, K Öberg, M Aout, P B Ruszniewski and M E Pavel critically revised the content and approved the final draft of the manuscript for publication. 269 270 271 272 Acknowledgments The authors thank Bhavik Shah, PhD of Novartis Healthcare Pvt. Ltd. for providing medical editorial assistance with this manuscript. Page 11 of 13

Page 12 of 20 273 References 274 275 276 Caplin ME, Pavel M, Cwikla JB, Phan AT, Raderer M, Sedlackova E, Cadiot G, Wolin EM, Capdevila J, Wall L, et al. 2014 Lanreotide in metastatic enteropancreatic neuroendocrine tumors. N Engl J Med 371 224-233. 277 278 279 280 James K, Eisenhauer E, Christian M, Terenziani M, Vena D, Muldal A & Therasse P 1999 Measuring response in solid tumors: unidimensional versus bidimensional measurement. J Natl Cancer Inst 91 523-528. 281 282 283 284 Oxnard GR, Morris MJ, Hodi FS, Baker LH, Kris MG, Venook AP & Schwartz LH 2012 When progressive disease does not mean treatment failure: reconsidering the criteria for progression. J Natl Cancer Inst 104 1534-1541. 285 286 287 288 289 Pavel ME, Hainsworth JD, Baudin E, Peeters M, Horsch D, Winkler RE, Klimovsky J, Lebwohl D, Jehl V, Wolin EM, et al. 2011 Everolimus plus octreotide long-acting repeatable for the treatment of advanced neuroendocrine tumours associated with carcinoid syndrome (RADIANT-2): a randomised, placebocontrolled, phase 3 study. Lancet 378 2005-2012. 290 291 292 293 294 Pavel M, Baudin E, Couvelard A, Krenning E, Öberg K, Steinmller T, Anlauf M, Wiedenmann B, Salazar R. 2012 ENETS consensus guidelines for the management of patients with liver and other distant metastases from neuroendocrine neoplasms of foregut, midgut, hindgut, and unknown primary.neuroendocrinology 95 157. 295 296 297 298 299 Rinke A, Muller HH, Schade-Brittinger C, Klose KJ, Barth P, Wied M, Mayer C, Aminossadati B, Pape UF, Blaker M, et al. 2009 Placebo-controlled, double-blind, prospective, randomized study on the effect of octreotide LAR in the control of tumor growth in patients with metastatic neuroendocrine midgut tumors: a report from the PROMID Study Group. J Clin Oncol 27 4656-4663. 300 Page 12 of 13

Page 13 of 20 301 302 303 304 Therasse P, Arbuck SG, Eisenhauer EA, Wanders J, Kaplan RS, Rubinstein L, Verweij J, Van Glabbeke M, van Oosterom AT, Christian MC, et al. 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. 305 306 307 308 Therasse P, Le Cesne A, Van Glabbeke M, Verweij J, Judson I, Tissue ES & Bone Sarcoma G 2005 RECIST vs. WHO: prospective comparison of response criteria in an EORTC phase II clinical trial investigating ET-743 in advanced soft tissue sarcoma. Eur J Cancer 41 1426-1430. 309 310 311 Vinik AI, Woltering EA, Warner RR, Caplin M, O'Dorisio TM, Wiseman GA, Coppola D & Go VL 2010 NANETS consensus guidelines for the diagnosis of neuroendocrine tumor. Pancreas 39 713-734. 312 313 314 315 Yao JC, Hassan M, Phan A, Dagohoy C, Leary C, Mares JE, Abdalla EK, Fleming JB, Vauthey JN, Rashid 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. 26 3063-3072. 316 317 Page 13 of 13

Page 14 of 20 Figure Legends Figure 1 RADIANT-2 study design and the focus of the present post hoc analysis. LAR, long-acting repeatable; q28d, every 28 days; SSA, somatostatin analogues. *A total of 196 patients (41 SSA-naïve and 155 who had received SSA previously) had foregut, midgut, or hindgut NET and were included in the present analysis. Figure 2 Kaplan-Meier plots of progression-free events in patients with midgut tumors. A. By adjudicated central review. B. By local assessment. SSA, somatostatin analogues Page 1 of 1

Page 15 of 20 Table 1 Baseline demographics and clinical characteristics (stratified by previous SSA use) SSA naïve n = 41 Previous SSA n = 155 All patients N = 196 Median age (range), years 59 (37 75) 60 (27 81) 60 (27 81) Sex, n (%) Male 22 (54) 92 (59) 114 (58) Female 19 (46) 63 (41) 82 (42) WHO PS, n (%) a 0 30 (73) 100 (65) 130 (66) 1 11 (27) 45 (29) 56 (29) 2 0 10 (6) 10 (5) Tumor location, n (%) b Foregut 15 (37) 17 (11) 32 (16) Midgut 24 (59) 119 (77) 143 (73) Hindgut 2 (5) 19 (12) 21 (11) Histology grade, n (%) c Well differentiated or low 32 (78) 128 (83) 160 (82) grade Moderately differentiated or 9 (22) 20 (13) 29 (15) intermediate grade Poorly differentiated or high 0 1 (<1) 1 (<1) grade Unknown 0 6 (4) 6 (3) Time since initial diagnosis, n (%) cd

Page 16 of 20 6 months 10 (24) 11 (7) 21 (11) >6 months to 2 years 9 (22) 37 (24) 46 (23) >2 to 5 years 14 (34) 35 (23) 49 (25) >5 to 10 years 6 (15) 51 (33) 57 (29) >10 years 2 (5) 20 (13) 22 (11) Systemic antitumor drugs, n (%) Chemotherapy 11 (27) 43 (28) 54 (28) Immunotherapy 1 (2) 18 (12) 19 (10) Targeted therapy 1 (2) 13 (8) 14 (7) Other 1 (2) 25 (16) 26 (13) Previous SSA therapy, n (%) Octreotide LAR 142 (92) 142 (72) de Lanreotide 13 (8) 13 (7) de WHO PS, World Health Organization performance status; SSA, somatostatin analogues; LAR, long-acting repeatable. a Data missing for one patient. b Tumors originating in the lung, stomach, duodenum, or pancreas were categorized as foregut NET; those originating in the small intestine, appendix, proximal colon, or of unknown primary sites were considered to be midgut NET; and those with the colon (transverse or distal) or rectum as the primary site were classified as hindgut NET. c The histology-based categories as reported in pathology reports. At the time of study conduct, the harmonized definition of grading for NET was not established and the World Health Organization classification was not in practice. Thus, the categories reported here may not translate in to strict numerical categories based on World Health Organization grading (based on mitotic count and/or Ki-67 labeling index) being used in current clinical practice. d Data missing for two patients. de Total is not 100%, as 41 (21%) patients did not receive prior SSA therapy.

Page 17 of 20 Table 2 Overall survival (data cutoff date, June 13, 2013) SSA naïve Previous SSA a n (%) Median OS, n (%) Median OS, months months (95% CI) (95% CI) Total patient population 41 (100) 50.6 (36.4 NR) 155 (100) 33.5 (27.5 44.7) Tumor location Foregut 15 (37) 36.4 (25.5 NR) 17 (11) 33.6 (16.7 NR) Midgut 24 (59) NR (42.4 NR) 119 (77) 33.5 (27.5 49.4) Hindgut 2 (5) NR (4.7 NR) 19 (12) 35.8 (15.4 48.9) SSA, somatostatin analogues; OS, overall survival; NR, not reached. a Includes patients who had received previous octreotide or lanreotide treatment.

All patients (N = 429) Randomization (1:1) Page 18 of 20 Focus of the present post hoc analysis Everolimus 10 mg/d + octreotide LAR 30 mg q28d (n = 216) Crossover to openlabel everolimus at disease progression (n = 124) Placebo + octreotide LAR 30 mg q28d (n = 213) SSA naïve* (n = 41) Previous SSA* (n = 155) Treatment until disease progression

A 100 Page 19 of 20 Percentage of event-free 80 60 40 20 0 Censoring Times Naïve to SSA (n/n = 16/24) Non-naïve to SSA (n/n = 61/119) Hazard Ratio: 1.20 95% CI [0.69 2.09] Kaplan-Meier medians Naïve to SSA : 22.21 months Non-naïve to SSA: 11.96 months Logrank P value = 0.5209 0 6 12 18 24 Time (months) No. of patients still at risk Naïve to SSA 24 21 14 12 5 Non-naïve to SSA 119 67 40 29 12

B Percentage of event-free 100 80 60 40 20 0 Censoring Times Naïve to SSA (n/n = 15/24) Non-naïve to SSA (n/n = 88/119) Hazard Ratio: 1.84 95% CI [1.06 3.19] Kaplan-Meier medians Page 20 of 20 Naïve to SSA : 21.85 months Non-naïve to SSA: 8.67 months Logrank P value = 0.0278 0 6 12 18 24 Time (months) No. of patients still at risk Naïve to SSA 24 21 15 13 5 Non-naïve to SSA 119 71 42 30 12