A Shining Light in the Darkness for the Treatment of Pancreatic Neuroendocrine Tumors

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

MEDICAL MANAGEMENT OF METASTATIC GEP-NET

TRACTAMENT ONCOLÒGIC DELS TUMORS NEUROENDOCRINS METASTÀSICS

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

Pancreatic Neuroendocrine Tumors: Entering a New Era Highlights from the 2012 ASCO Gastrointestinal Cancers Symposium.

SUPPLEMENTARY INFORMATION

PRINCESS MARGARET CANCER CENTRE CLINICAL PRACTICE GUIDELINES

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

Neuroendocrine Tumors: Just the Basics. George Fisher, MD PhD

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

Systemic Therapy for Advanced Pancreatic Neuroendocrine Tumors: An Update

Development of New Treatment Modalities Oncolytic Viruses and Nanotechnique

Oberndofer 1907 Illeal Serotonin Secreting Tumor Carcinoid (Karzinoide)

I nuovi farmaci: associazione o superamento del trattamento con analoghi

Cutting Edge Treatment of Neuroendocrine Tumors

Cutting Edge Treatment of Neuroendocrine Tumors

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

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

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

The PI3K/AKT axis. Dr. Lucio Crinò Medical Oncology Division Azienda Ospedaliera-Perugia. Introduction

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

Systemic Therapy for Gastroenteropancreatic (GEP) Neuroendocrine Tumors and Lung Carcinoid

Novel Molecular Molecular Therapies In Hepatocarcinoma Prof Eric

Recent developments of oncology in neuroendocrine tumors (NETs)

Teresa Alonso Gordoa Servicio Oncología Médica Hospital Universitario Ramón y Cajal

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

7/6/2015. Cancer Related Deaths: United States. Management of NSCLC TODAY. Emerging mutations as predictive biomarkers in lung cancer: Overview

Targeted Therapies in Metastatic Colorectal Cancer: An Update

Targeted and immunotherapy in RCC

Breast Cancer: the interplay of biology, drugs, radiation. Prof. L. Livi Università degli Studi di Firenze. Brescia, October 3rd 4th, 2013

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

Bevacizumab in Advanced Adenocarcinoma of the Pancreas. Original Policy Date

Jon Trent, MD, PhD. Associate Professor Dept. of Sarcoma Medical Oncology The University of Texas, M. D. Anderson Cancer Center

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

2015: Year in Review Results of Recent Trials

UNIVERSITY OF MEDICINE AND PHARMACY CRAIOVA PhD SCHOOL. PhD THESIS

Mechanisms of hormone drug resistance

2015 EUROPEAN CANCER CONGRESS

Future outlooks in NET: The pathologists point of view. Aurel Perren, Institute of Pathology University of Bern

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

Systemic Therapy for Pheos/Paras: Somatostatin analogues, small molecules, immunotherapy and other novel approaches in the works.

NET ΠΝΕΥΜΟΝΑ: τι νεότερο / νέες μελέτες

pan-canadian Oncology Drug Review Final Clinical Guidance Report Axitinib (Inlyta) for metastatic Renal Cell Carcinoma March 7, 2013

Evidenze cliniche nel trattamento del RCC

A particular set of insults induces apoptosis (part 1), which, if inhibited, can switch to autophagy. At least in some cellular settings, autophagy se

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

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

Antiangiogenics are effective treatments in NETs

Horizon Scanning in Oncology

Key Words. mtor inhibition Insulinoma Everolimus 18 F-FDG-PET

David N. Robinson, MD

Backgrounder. 1. What are targeted therapies? 2. How do targeted therapies work?

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

Have Results of Recent Randomized Trials Changed the Role of mtor Inhibitors?

Update on the Management of HER2+ Breast Cancer. Christian Jackisch, MD, PhD Sana Klinikum Offenbach Offenbach, Germany

Ongoing and future clinical investigation in GEP NENs

Diabetes Mellitus and Breast Cancer

Review of Gastrointestinal Carcinoid Tumors: Latest Therapies

ESMO 2017, Madrid, Spain Dr. Loredana Vecchione Charite Comprehensive Cancer Center, Berlin HIGHLIGHTS ON CANCERS OF THE UPPER GI TRACT

LAPATINIB-Resistance to small Molecule ErbB2 Tyrosine Kinase Inhibitor (TKI)

The 2010 Gastrointestinal Cancers Symposium Oral Abstract Session: Cancers of the Pancreas, Small Bowel and Hepatobilliary Tract

Cancer Cell Research 14 (2017)

Targeted Therapy in Advanced Renal Cell Carcinoma

Multimedia Appendix 6 Educational Materials Table of Contents. Intervention Educational Materials Audio Script (version 1)

Conflicts of Interest GI Malignancies: An Update on Current Treatment Options

The following slides are provided as presented by the author during the live educa7onal ac7vity and are intended for reference purposes only.

Everolimus for Advanced Pancreatic Neuroendocrine Tumors

Study No Title : Rationale: Phase: Study Period: Study Design: Centres: Indication: Treatment:

Review Article Management Options for Advanced Low or Intermediate Grade Gastroenteropancreatic Neuroendocrine Tumors: Review of Recent Literature

Sequential Therapy in Renal Cell Carcinoma*

Phospho-AKT Sampler Kit

Angiogenesis and tumor growth

pan-canadian Oncology Drug Review Final Clinical Guidance Report Nivolumab (Opdivo) for Metastatic Renal Cell Carcinoma September 1, 2016

Chemotherapy for Advanced Gastric Cancer

Nature Medicine: doi: /nm.3559

Phase II Cancer Trials: When and How

Antiangiogenic Agents in NSCLC Where are we? Which biomarkers? VEGF Is the Only Angiogenic Factor Present Throughout the Tumor Life Cycle

EGFR: fundamenteel en klinisch

Management Guidelines and Targeted Therapies in Metastatic Non-Small Cell Lung Cancer: An Oncologist s Perspective

EGFR Antibody. Necitumumab, LY , IMC-11F8. Drug Discovery Platform: Cancer Cell Signaling

José Baselga, MD, PhD

The Current Champion: Angiogenesis inhibitors

Advanced HER2+ Breast Cancer: New Options and How to Deploy Them. José Baselga MD, PhD

Phase II Cancer Trials: When and How

Contents. Preface XV Acknowledgments XXI List of Abbreviations XXIII About the Companion Website XXIX

ADVANCED COLORECTAL CANCER: UNRESECTABLE OR BORDERLINE RESECTABLE (GROUP 1) CHEMOTHERAPY +/- TARGETED AGENTS. Andrés Cervantes. Professor of Medicine

A randomized phase 2 trial of CRLX101 in combination with bevacizumab in patients with metastatic renal cell carcinoma (mrcc) vs standard of care

Management of Neuroendocrine Tumors

Nintedanib in Oncology Backgrounder

Carcinoma de Tiroide: Teràpies Diana

My name is Dr. David Ilson, Professor of Medicine at Memorial Sloan Kettering Cancer Center and Weill Cornell Medical Center in New York, New York.

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

THE FUTURE OF IMMUNOTHERAPY IN COLORECTAL CANCER. Prof. Dr. Hans Prenen, MD, PhD Oncology Department University Hospital Antwerp, Belgium

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

Gastrinoma: Medical Management. Haley Gallup

Biologics Effects of Targeted Therapeutics

National Horizon Scanning Centre. Sunitinib (Sutent) for advanced and/or metastatic breast cancer. December 2007

Mechanisms of Resistance to Antiangiogenic. Martin J. Edelman, MD University of Maryland Greenebaum Cancer Center Dresden, 2012

Breast cancer update. Iryna Kuchuk, MD Oncology department Meir Medical Center

COMETS: COlorectal MEtastatic Two Sequences

Transcription:

MINI REVIEW A Shining Light in the Darkness for the Treatment of Pancreatic Neuroendocrine Tumors Jaume Capdevila and Josep Tabernero ABSTRACT Gastroenteropancreatic neuroendocrine tumors are rare neoplasms; past decades have seen limited research channeled into this area. Recently, 2 placebocontrolled phase III trials using 2 drugs everolimus and sunitinib with distinct molecular rationales achieved their principal objective of increasing survival in patients with advanced pancreatic neuroendocrine tumors (PNET). Nonetheless, several questions remain unanswered, notably defining the optimal schedule for integrating these targeted agents with conventional cytotoxics and other treatment options, and identifying appropriate biomarkers for patients with the potential to derive greater benefit. In this article, we analyze the results of the 2 largest studies ever completed in patients with PNETs and discuss the challenges for future drug development in this setting. Summary: Sunitinib and everolimus will become new treatment options for patients with PNETs and will be integrated into the complex therapeutic management of this disease. In this review, we summarize the evidence-based data of these drugs as well as the molecular-based science in this setting that will lay the groundwork for future studies. Cancer Discovery; 1(3); 213 21. 2011 AACR. INTRODUCTION Gastroenteropancreatic neuroendocrine tumors (GEPNET) constitute a heterogenic group of neoplasms derived from the Kultchitzky cells of the diffuse neuroendocrine system located in the gastrointestinal tract (origin of carcinoid tumors) and pancreatic islet cells (origin of pancreatic endocrine tumors). Although GEPNETs represent less than 2% of all gastrointestinal cancers, they constitute the second most prevalent advanced tumor of the gastrointestinal tract after colorectal cancer ( 1 ). The biological behavior of well-differentiated pancreatic neuroendocrine tumors (PNET) depends mainly on their histologic characteristics, such as differentiation grade, Ki67 expression, or angioinvasion, and the proportion of malignant tumors developing metastases varies widely between tumor types (from 10% of insulinomas to 90% of glucagonomas). Authors Affiliation: Medical Oncology Department, Vall d Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain Corresponding Author: Josep Tabernero, MD, Medical Oncology Department, Vall d Hebron Institute of Oncology (VHIO), Vall d Hebron University Hospital, Universitat Autònoma de Barcelona, Pg Vall d Hebron 119-129, Barcelona 08035, Spain. Phone: 34-93-2746085; Fax: 34-93- 2746059; E-mail: jtabernero@vhebron.net doi: 10.1158/2159-8290.CD-11-0151 2011 American Association for The medical treatment of advanced PNETs has included somatostatin analogs, interferon, and cytotoxic agents. This latter group of drugs has limited activity in PNETs for many reasons, including the fact that they usually have a low mitosis rate and also because of their particular biological properties, such as the presence of biologic markers related to chemoresistance (i.e., Akt expression) ( 2 ). The lack of effectiveness of conventional cytotoxic agents has prompted exploration of new targeted drugs exploiting phenotypical features of PNETs. Nevertheless, the development of targeted therapies in this field has been limited by a number of factors. First, PNETs are extremely heterogeneous with very different characteristics in highly and poorly differentiated tumors. Second, the preclinical development of drugs directed against these tumors is limited because very few in vitro and in vivo models exist. And last, but not least, the frequent shortage of resources has impacted strongly on rare neoplasms that require international efforts to design and implement scientifically valid studies. In-depth characterization of molecular features and pathways of cell growth, apoptosis, angiogenesis, and invasion are still lacking in this group of tumors, particularly concerning genetic mutations or epigenetic alterations that generate oncogenic dependency or even addiction. This is the concept behind an ideal target; unfortunately, however, the majority of new developments are based only on phenotypic overexpression of pathways with AUGUST 2011 CANCER DISCOVERY 213 Downloaded from cancerdiscovery.aacrjournals.org on October 21, 2018. 2011 American Association for

MINI SECTION REVIEW Capdevila et al. MANAGEMENT OF PNET Management of advanced PNETs is based on a multidisciplinary approach that includes surgery, liver-directed treatments, radionuclide and biological therapies, chemotherapy, and targeted agents. Strong molecular rationale supports antiangiogenic and mtor-directed therapies for the treatment of PNETs, with a favorable toxicity profile compared with standard chemotherapy. After decades of slight improvement, sunitinib and everolimus have demonstrated a significant benefit in progression-free survival and response rate in the largest ever completed phase III studies in PNETs. No predictive biomarkers have been characterized so far for better patient selection. PTEN and TSC1/2 downregulation have been suggested to be prognostic factors in PNETs and their potential predictive value in the treatment with PI3K pathway inhibitors is being evaluated. Gene expression profiling and tumor sequencing studies have confirmed the relevance of the PI3K- AKT-mTOR pathway in the pathogenesis of PNETs, and have highlighted other significant pathways less known in the PNET molecular profile, but with enough preclinical data to be assessed in the future, such as Wnt/β-catenin, Hedgehog, Notch, or TGF-β. The prolonged survival of patients with PNETs requires that they sequentially receive all the available treatment options. It is anticipated that ongoing and next-generation studies will clarify whether the best approach is to combine some active cytotoxic drugs with targeted agents or to sequentially administer all available treatment options. In this regard, predictive and prognostic biomarkers will likely be useful to better define the most appropriate sequence. The (over)expression of some factors has been associated with poor prognosis and decreased progression-free survival (PFS), as well as with tumor growth, aggressiveness, and disease extent in patients with PNETs ( 4 ). Currently, the medical management of functional PNETs almost invariably involves the use of somatostatin analogs to control the symptoms related to hormone secretion. Interferon-α is added when patients develop resistance. Although both treatments provide up to 70% to 80% of hormone syndrome control and relief of induced symptoms, antiproliferative effects are far from impressive, ranging from 5% to 10% at best. Cytotoxic chemotherapy is a treatment option in patients with PNETs, given that these tumors typically have intermediate cell proliferation rates (2% to 20% measured by Ki67 immunohistochemistry index). Combination regimens containing streptozocin, dacarbazine, epirubicin, lomustine, cisplatin, doxorubicin, etoposide, and 5-fluorouracil have shown the best efficacy, with overall response rates (ORR) of 20% to 60%, and a median overall survival (mos) ranging from 15 to 26 months ( 5 ). Surgery, the mainstay of treatment for localized tumors, can be considered in patients with metastatic disease, to reduce tumor mass, before or concomitantly with systemic therapy, and to palliate symptoms. Other approaches to reduce tumor burden or to contribute to hormone syndrome control include hepatic artery (chemo)embolization, radiofrequency ablation, and somatostatin receptor-targeted radionuclide therapy ( 6 ). Several targeted agents have been studied in the treatment of GEPNETs including antiangiogenic compounds (e.g., bevacizumab), inhibitors of multiple receptors with kinase activity (e.g, sunitinib), and inhibitors of intracellular downstream effector proteins such as mtor (e.g., everolimus). Two phase III studies with everolimus and sunitinib were recently conducted in patients with PNETs and demonstrated significant clinical activity increasing PFS, and results were published in the New England Journal of Medicine. The last publication of a chemotherapy combination in this disease in this prestigious journal was two decades ago, and that treatment became the standard of care of patients with advanced well-differentiated PNETs (7 ). moderate signaling repercussions in cell growth and survival, which limit their therapeutic potential. Emerging studies are providing insights into specific deregulation in proliferative and growth signaling pathways of various PNET subsets ( 3 ). PNETs are characterized by being remarkably vascular and expressing several growth factors, including vascular endothelial growth factor (VEGF), platelet-derived growth factor (PDGF), insulin-like growth factor 1 (IGF-1), basic fibroblast growth factor (bfgf), and transforming growth factor (TGF)-α and TGF-β. In addition, expression of several receptors of these growth factors and ligands has been described, including stem cell factor receptor (c-kit), epidermal growth factor receptors (EGFR), VEGF receptors (VEGFR)-2 and 3, IGF receptors (IGF-R), and PDGF receptors (PDGFR). ANTIANGIOGENESIS TREATMENT IN ADVANCED PNETs PNETs are some of the most vascularized tumors identified to date. Currently, a number of antiangiogenic compounds are being evaluated in the clinic, and can be divided into 3 groups: (i) drugs targeting VEGF, such as the VEGFdirected monoclonal antibody bevacizumab; (ii) small molecules that inhibit the receptor tyrosine kinase domains of VEGFR and other related receptors, such as sunitinib, sorafenib, and pazopanib; and (iii) other compounds with different antiangiogenic mechanisms, such as thalidomide or endostatin (Table 1 ). The first evidence of activity of bevacizumab in GEPNETs was reported by Yao and colleagues ( 8 ) in a phase II study with 44 patients randomly assigned to receive bevacizumab (15 mg/kg every 3 weeks) or weekly pegylated interferon α-2b 214 CANCER DISCOVERY AUGUST 2011 www.aacrjournals.org

Treatment of Pancreatic Neuroendocrine Tumors MINI REVIEW Table 1. Main antiangiogenic compounds in clinical development for advanced neuroendocrine tumors Agent(s) Target(s) Phase Tumor ORR PFS (m) Comments Reference Bevacizumab VEGF Phase II Carcinoid 18% 16.5 Ongoing phase III 8 BV + CHT FOLFOX GEPNET 20% 33% Ongoing 31 XELOX PNETs 31% Ongoing 32 Temozolomide GEPNET 0% 24% Ongoing 33 BV + everolimus VEGF + mtor GEPNET 21% 14 Ongoing 41 Sunitinib VEGFR1-3, PDGFR, Phase II Carcinoid 2% 10.5 10 RET, FLT3, KIT PNETs 17% 7.7 Phase III PNETs 9% 11.4 Positive phase III 11 Sorafenib VEGFR2-3, PDGF, Phase II Carcinoid 7% 7.8 Completed 34 Raf, KIT, RET PNETs 17% 11.9 Sorafenib + BV VEGFR2-3, PDGF, GETNET 10% 12.4 Completed 35 Raf, KIT, RET + VEGF Vatalanib VEGFR1-3, PDGFR, KIT Phase II GEPNET 0% NR Withdrawn Pazopanib VEGFR1-3, PDGFR, Phase II Carcinoid 0% 12 Ongoing 36 PNETs 19% 14.2 Motesanib VEGFR1-3, PDGFR, RET Phase II GEPNET Ongoing Atiprimod Inhibits VEGF secretion Phase II GEPNET 0% 76% at 6 m Ongoing 37 Deactivates Akt and STAT3 Thalidomide VEGF and bfgf Phase II GEPNET 0% 80% SD 38 Thalidomide + CHT Temozolomide + 7% 45% 39 VEGF and bfgf rh-endostatin Endogenous endothelial Phase II Carcinoids 0% 5.8 80% SD 40 inhibition PNETs 0% 7.6 Abbreviations: BV, bevacizumab; CHT, chemotherapy; NR, not reached; PFS, progression-free survival; STAT, signal transducers and activators of transcription; rh, recombinant human; m, months. (PIF; 0.5 μg/kg, for 18 weeks). After 18 weeks of treatment or in the event of disease progression, patients could receive the combination of the 2 drugs. The bevacizumab arm showed a higher ORR (18% vs. 0%), a lower progression rate (5% vs. 27%), and therefore a better PFS rate after 18 weeks of treatment (95% vs. 68%). Bevacizumab treatment was also associated with a significant tumor blood decrease measured by functional computed tomography (CT) scans compared with PIF ( P < 0.01). The toxicity profile was manageable with hypertension as the main adverse event. A larger phase III study (SWOG S0518) is currently recruiting patients to confirm these promising results. Bevacizumab is being evaluated in combination with cytotoxic chemotherapy and also with other targeted therapies, including sorafenib and everolimus (Table 1). Sunitinib malate (SU-11248) is a highly potent, ATPcompetitive binding small molecule capable of inhibiting several tyrosine kinase membrane receptor proteins involved in the main angiogenic and proliferative pathways, such as VEGFR-1 to 3, PDGFR, FLT-3, c-kit, and RET. The first evidence of activity of sunitinib in advanced GEPNETs was reported in a first-in-man phase I study ( 9 ), which was followed by a phase II trial with 107 patients (41 with carcinoid tumors and 66 with PNETs) with documented disease progression. The authors reported an ORR of 16.7% in PNETs with 68% of patients presenting stable disease (SD). In patients with carcinoid tumors, the ORR and SD rates were 2.4% and 83%, respectively. A pharmacodynamic study of potential soluble surrogate biomarkers was also done and identified svegfr-3 as a potential biomarker of the biological effect of sunitinib and interleukin-8 as a potential predictor of response ( 10 ). These results led to the design of a double-blind, placebo-controlled phase III study in advanced PNETs ( 11 ). The initial design planned to recruit 340 patients with disease progression documented by Response Evaluation Criteria in Solid Tumors (RECIST) in the 12 months prior to trial enrollment. The study was prematurely stopped following a recommendation of the Data Monitoring Committee due to significant differences in disease progression and deaths in the placebo arm. A total of 171 patients were enrolled in the trial, 86 receiving sunitinib (37.5 mg daily) and 85 receiving placebo. The results of the study regarding ORR, median PFS (mpfs), and estimated mos significantly favored the sunitinib arm. Patients treated with sunitinib achieved mpfs of 11.4 months compared with 5.5 months in the placebo arm AUGUST 2011 CANCER DISCOVERY 215

MINI SECTION REVIEW Capdevila et al. (HR: 0.42; 95% CI, 0.26 0.66; P = 0.0001), with a probability of being event free at 6 months of 71.3% versus 43.2%. In the subgroup analysis, all groups showed benefit with sunitinib regardless of age, Eastern Cooperative Oncology Group (ECOG) performance status (PS), previous therapies, tumor burden, time since diagnosis, concomitant treatment with somatostatin analogs, or Ki67 levels. Patients treated with sunitinib presented an ORR of 9% compared with no responses in the placebo arm (P = 0.007). The toxicity profile was as expected for sunitinib with diarrhea, fatigue, nausea, and vomiting being the main side effects observed. The most frequent grade 3-4 adverse events were neutropenia (12% vs. 0%), hypertension (10% vs. 1%), handfoot syndrome (6% vs. 0%), and leukopenia (6% vs. 0%). Benefit data in mos (HR: 0.41; 95% CI, 0.19 0.89; P = 0.02) should be considered with caution because no crossover was planned in the design of the protocol, and although 69% of patients received sunitinib after progression with placebo, not all of them received the drug immediately after tumor progression. The trial also included quality of life (QoL) assessment, and sunitinib provided clinical benefit while maintaining global QoL. In conclusion, sunitinib demonstrated a significant benefit in mpfs and response rate in patients with radiologically progressive well-differentiated PNETs compared to placebo. INHIBITION OF mtor IN PNETs mtor is an intracellular serine-threonine kinase downstream of the phosphatidyl inositol 3 kinase (PI3K)-AKT signaling pathway. Under physiologic conditions the PI3K- AKT-mTOR signaling pathway plays a major role in regulating cell growth, proliferation, motility, and survival, and cellular nutrient and energy levels, protein synthesis, autophagy, transcription, and redox status. mtor integrates multiple upstream signals, including growth factors (e.g., EGF and IGF-1/2), and mitogens. Interestingly, mtor is also involved in angiogenesis, regulating the translation and activity of hypoxia-inducible factor 1α (HIF1α), which is related to VEGF expression under hypoxic conditions. Hyperactive mtor signaling is a key participant in development, growth, and proliferation in about 50% of all human tumors. mtor acts as the catalytic subunit of 2 distinct complexes. mtor complex 1 (mtorc1) consists of mtor, the target of the rapamycin complex subunit LST8 (LST8, also known as GβL), the proline-rich Akt substrate 1 (AKT1S1, also named PRAS40), and its main partner, the regulatory associated protein of mtor (RAPTOR). mtor complex 2 (mtorc2) is composed of mtor, LST8, the target of rapamycin complex 2 subunit MAPKAP1 (SIN1), the proline-rich protein 5 (PRR5, or PROTOR1), and its most important partner, rapamycin-insensitive companion of mtor (RICTOR). mtorc1 is characterized by the classic features of mtor, functioning as a nutrient-energy-redox sensor and controlling protein synthesis. The activity of this complex is stimulated by several growth factors, hormones, nutrients (amino acids, glucose), cellular energy status, and stress conditions. mtorc2 functions as an important regulator of the cytoskeleton and also phosphorylates the serine-threonine protein kinase AKT when mtor is blocked, through the use of an mtor inhibitor. Activation of mtorc1 by positive regulators induces the phosphorylation of 2 of the main downstream effectors, the eukaryotic translation initiation factor 4E binding protein-1 (4E-BP1) and the ribosomal protein S6 kinase β-1 (S6K1). The activation of these effectors leads to the enhancing of the translation of essential mrnas involved in the cell cycle progression through G1 to S1 phase (12). Several targets of the PI3K-AKT-mTOR signaling pathway have been identified in PNETs, and several genetic disorders that have constitutive activation of this pathway along with increased incidence of neuroendocrine tumors have been well defined, such as the tuberous sclerosis complex (TSC), neurofibromatosis type I, Von Hipple-Lindau (VHL) disease, or multiple endocrine neoplasia type I (MEN1) (13). Two rapamycin inhibitors have been evaluated in neuroendocrine tumors: temsirolimus and everolimus. Temsirolimus was only studied in a multicenter phase II trial with 37 advanced GEPNETs; although the therapy was generally well tolerated, the drug showed modest antitumor activity with only 2 confirmed responses (5.6%). The activity was similar between carcinoid and PNETs: mpfs was 6 months and OS at 1 year was 71.5%. The authors considered the activity was not sufficiently clinically meaningful for further development. Everolimus has been intensively developed in GEPNETs. The first study in this disease, conducted at the MD Anderson Cancer Center, included 30 patients with carcinoid tumors and 30 patients with PNETs in 2 consecutive cohorts (14). The first cohort treated 30 patients with the combination of depot octreotide (30 mg intramuscularly every 28 days) plus everolimus (5 mg daily). The second cohort included 30 patients treated with octreotide at the same doses and everolimus (10 mg daily). The authors reported encouraging antitumor activity with an ORR of 17% in carcinoid tumors and 27% in islet cell carcinomas, with mpfs of 63 and 50 weeks, respectively. Moreover, patients included in the cohort receiving everolimus (10 mg) obtained a higher ORR (30% vs. 13%) and prolonged mpfs (72 vs. 50 weeks). Overall, everolimus showed an acceptable toxicity profile, the most frequent grade 3-4 adverse events being aphthous ulcers, fatigue, diarrhea, hyperglycemia, and hypophosphatemia. After these encouraging results, the RAD001 in Advanced Neuroendocrine Tumors (RADIANT) trials were designed to explore the efficacy of everolimus in neuroendocrine tumors of different origins. The RADIANT-1 study was a confirmatory international phase II trial in progressive chemotherapy-refractory islet cell carcinomas divided into 2 strata: the first included 115 patients with advanced PNETs treated with everolimus (10 mg daily) and the second had 45 patients with the combination of depot octreotide (30 mg intramuscularly every 28 days) plus everolimus (10 mg daily) (15). The ORR was 7.8% in stratum 1 and 4.4% in stratum 2, with SD rates of 68.7% and 77.8%, respectively. The mpfs was 9.3 months in stratum 1 and 12.9 months in stratum 2. Additionally, 49% and 56% of patients with 216 CANCER DISCOVERY AUGUST 2011 www.aacrjournals.org

Treatment of Pancreatic Neuroendocrine Tumors MINI REVIEW increased chromogranin A levels in stratum 1 and 2, respectively, achieved either normalization or a reduction greater than 50%. Moreover, the authors reported a statistically significant relationship between the decrease in chromogranin A levels and mpfs (decrease 30%, mpfs 13.3 months vs. nonresponders, 7.5 months). The treatment was generally well tolerated with few grade 3-4 adverse events, mainly fatigue, diarrhea, and stomatitis. RADIANT-2 and RADIANT-3 studies are 2 international, double-blind, placebo-controlled phase III trials in advanced clinically functional extrapancreatic neuroendocrine tumors and advanced PNETs, respectively. The initial report of the RADIANT-2 trial was presented at the 35th European Society for Medical Oncology (ESMO) Congress (Milan, Italy, October 8 12, 2010). In this trial, the efficacy and safety of everolimus (10 mg/d) plus octreotide LAR (30 mg every 28 days) was compared to that of placebo plus octreotide LAR (30 mg every 28 days) in 429 patients with advanced carcinoid tumors and a history of hormone-related symptoms. Although the study did not meet its primary endpoint based on central radiologic review of data, the results showed that everolimus plus octreotide LAR significantly improved mpfs by 5.1 months (HR: 0.77; 95% CI, 0.59 1.00; P = 0.026) when compared with placebo. After adjusting for imbalances in baseline characteristics between the 2 treatment arms, and inconsistencies between radiology scans to assess disease progression, the results also showed that everolimus plus octreotide LAR significantly reduced the risk of disease progression by 40% (HR: 0.60; 95% CI, 0.44 0.84; P = 0.0014) when compared to octreotide LAR alone (16). The results of the RADIANT-3 study were recently published, showing the efficacy of everolimus in advanced PNETs (17). A total of 410 patients with disease progression documented prior to inclusion were randomly assigned to receive either daily oral everolimus (10 mg/d) or placebo (1:1). Treatment with everolimus demonstrated a significant increase in mpfs by central radiological review, more than double that of placebo, from 4.6 to 11.0 months (HR: 0.35; 95% CI, 0.27 0.45; P < 0.0001) fulfilling the primary endpoint. At 18 months, PFS in the everolimus arm was 34%, with a subgroup of patients who obtained prolonged clinical benefit of everolimus treatment with an acceptable safety profile. The benefit in PFS was observed in all subgroups of patients regardless of previous therapies, ECOG PS, age, tumor burden, time since diagnosis, treatment with somatostatin analogs, and tumor grade. Although the response rate was relatively low but significantly higher in the everolimus arm (5% vs. 2%, P = 0.001) 64.4% of patients had decreases of between 1% and 29% in target lesion size. Thus, the main benefit of everolimus in PFS was seen due to minor responses and tumor stabilizations. No differences in OS were observed due to the preplanned crossover at disease progression defined in the protocol (73% of patients in the placebo arm received open-label everolimus at progression). The most common grade 3-4 adverse events were stomatitis (observed in 7% of patients), anemia (6%), and hyperglycemia (5%). Infections (23%), pneumonitis (12%), and interstitial lung disease (2%), which often represent the main clinical concerns with everolimus therapy, were typically grade 1-2 and generally managed with antibiotics and steroids. Results of further secondary analyses of the RADIANT-3 study are expected in the near future, such as possible predictive biomarkers that may guide clinicians in the selection of patients who are more likely to respond to everolimus therapy. In summary, the RADIANT trials are the largest studies in advanced neuroendocrine tumors ever conducted and, as a result, everolimus will become a new standard of care in advanced well-differentiated PNETs and probably a new therapy option in advanced carcinoid tumors in combination with somatostatin analogs. KEY UNMET NEEDS AND FUTURE DIRECTIONS Almost half a century has elapsed since the most widely used neuroendocrine tumor classification, which was developed according to embryologic origin, was proposed by Williams and Sandler. Since then, various systems of nomenclature based on site of origin, tumor stage, histologic, or functionality classifications have been introduced, causing much confusion. These terminology issues have jeopardized not only the creation of a worldwide prognostic classification of neuroendocrine tumors but also the development of well-designed clinical trials, where the inclusion of tumors with different behavior has compromised interpretation of the results, such as happened in the RADIANT-2 study. Fortunately, in recent years, gene expression profiling and surface sequencing have shown promising potential in the molecular features of neuroendocrine tumors. The key role of the PI3K-Akt-mTOR pathway in the pathogenesis of PNETs was known from phenotypes of hereditary syndromes, such as TSC, neurofibromatosis, VHL disease, or MEN1 syndromes, where constitutive activation of the PI3K-Akt-mTOR pathway leads to an increased incidence of PNETs (18 20). Data from tumor expression profiling in PNETs has also shown that the downregulation of PTEN and TSC2 correlates with poorer prognosis (3), but also microrna expression profiling has demonstrated that the expression level of mir-21 is strongly associated with liver metastases, and a high Ki67 index and mir21 levels are inversely proportional to PTEN levels (Fig. 1) (21, 22). The significance of HIF/p53 pathway in the development and progression of PNETs has also been highlighted and the protein encoded by the MEN1 gene, Menin, has altered distribution and intensity in 80% of PNETs, leading to the inhibition of AKT activation by regulating its cellular location (23, 24). Moreover, activation of the PI3K-Akt-mTOR pathway in PNETs is also led by the (over)expression of membrane tyrosine kinase receptors that trigger this pathway, such as IGF-1R, fibroblast growth factor receptor 3 (FGFR3), or in the companion vasculature with mutations in the FLT1 gene, which encodes VEGFR1 (25). Furthermore, expression of EGFR has been widely described in PNETs, but no meaningful activity has been observed with EGFR inhibitors. Preclinical data with the most used transgenic mouse model, RIP-Tag2, have demonstrated that AUGUST 2011 CANCER DISCOVERY 217

MINI SECTION REVIEW Capdevila et al. IGF-1R FGFR3 VEGFR EGFR NOTCH SSTR1-5 PTEN P13K RAS NICD Wnt mir-21 AKT BRAF SHP1 PLC MENIN mtor MAPK GSK-3 Hedgehog -catenin Caspase 8 p53 Bax GLi2/3A Smoothened DAXX/ATRX ATM HIF1 SMADs VHL MGMT TGF- Figure 1. Schematic of intracellular signaling pathways involved in pancreatic neuroendocrine tumor differentiation and proliferation, of intranuclear transcription factors, and DNA repair enzymes involved in tumor carcinogenesis and cytotoxic therapy resistance. ATM, ataxia telangiectasia mutated; ATRX, X-linked mental retardation and a-thalassemia syndrome protein; Bax, Bcl-2 associated X protein; DAXX, death domain-associated protein; EGFR, epidermal growth factor receptor; FGFR3, fibroblast growth factor receptor 3; GSK-3b, glycogen synthase kinase-3b; HIF1, hypoxia-inducible factor 1; IGF-1R, insulin-like growth factor receptor 1; MAPK, mitogen-activated protein kinase; MGMT, O 6 - methylguanine DNA methyltransferase; mir21, microrna-21; mtor, mammalian target of rapamycin; NICD, intracellular domain of Notch; PI3K, phosphatidylinositol-3-kinase; PLC, phospholipase C; PTEN, phosphatase and tensin homolog deleted; SHP1, Src homology region 2 domaincontaining phosphatase-1; SSTR1-5, somatostatin receptors 1-5; TGF-b, transforming growth factor-b; VEGFR, vascular endothelial growth factor receptor; VHL, Von Hippel-Lindau. EGFR inhibition with erlotinib is able to abrogate the potential resistance mediated by Akt upregulation after mtorc1 inhibition (26). Deep sequencing of the entire neuroendocrine tumor genome could be expected to provide not only instrumental knowledge for prognostic classification but also a biotype characterization that would allow assessment of potential drug pathways for individual patients. Currently, within the International Cancer Genome Consortium, there is a task force to perform deep sequencing of GEPNETs. Recent data based on deep sequencing have confirmed the key role of MEN1 and the PI3K-Akt-mTOR pathways, but has also highlighted the importance of several transcription factors, such as DAXX (death domain-associated protein), ATRX (X-linked mental retardation and α-thalassemia syndrome protein), or ATM (ataxia telangiectasia mutated), which may open new target opportunities for the treatment of PNETs (13). The next step after this characterization is the design of molecularly guided clinical studies that could help to determine the best treatment combinations for PNETs. In order to counter escape mechanisms along with primary 218 CANCER DISCOVERY AUGUST 2011 www.aacrjournals.org

Treatment of Pancreatic Neuroendocrine Tumors MINI REVIEW Table 2. Main planned/ongoing clinical trials with targeted therapies in advanced pancreatic neuroendocrine tumors Agent (s) Target(s) Phase Protocol ID Everolimus + erlotinib mtor + EGFR Phase II NCT00843531 AMG479 IGFR Phase II NCT01024387 Bevacizumab + pertuzumab + octreotide LAR VEGF + HER1/HER2 + SSRT Phase II NCT01121939 Everolimus +/ bevacizumab mtor + VEGF Phase II NCT01229943 LY2584702 + everolimus p70 S6 Kinase + mtor Phase Ib NCT01115803 Cixutumumab + everolimus + octreotide LAR IGFR + mtor + SSRT Phase Ib NCT01204476 Everolimus + pasireotide mtor + SSRT Phase Ib NCT01263353 Phase II SOM230I2201 Everolimus + sorafenib mtor + VEGFR2-3, PDGF, Raf, KIT, RET Phase Ib NCT00942682 FR901228 Histone deacetylase Phase II NCT00084461 MK-2206 Akt Phase II NCT01169649 Bortezomib Proteasome Phase II NCT00017199 Abbreviations: HER, human epithelial growth factor receptor; SSRT, somatostatin receptors. and secondary resistance to mtor inhibition, several combined approaches are being evaluated, including mtor and IGF-1R inhibition, mtor inhibition plus pan-analog of somatostatin receptors (SOM230), mtor and EGFR inhibition, and dual mtorc1 and mtorc2 and dual PI3K and mtorc1/2 inhibition. Some studies are already in early recruitment ( Table 2 ; also see clinicaltrials.gov ). Nonetheless, despite the strong molecular rationale for this approach, it seems very unlikely that combination of targeted agents will displace the use of cytotoxic agents. We should keep in mind that PNETs are not completely resistant to chemotherapy. Moreover, recent data suggest that temozolamide, a cytotoxic agent approved for the treatment of glioblastoma, may be active in patients with PNETs. Encouragingly, the patients most sensitive to this treatment may be characterized by O 6 -methylguanine DNA methyltransferase (MGMT) expression using immunohistochemistry ( 27 ). Another approach uses the abrogation of other signaling pathways such as Wnt/β-catenin, Hedgehog, Notch, or TGF-β, known to be critical in processes of epithelial-mesenchymal transition, and regulation of neuroendocrine differentiation and cancer stem cell proliferation and survival. Limited preclinical data support the evaluation of drugs targeting these pathways in patients with advanced PNETs ( 28 30 ). CONCLUSIONS The future management of advanced neuroendocrine tumors finally appears to be changing after decades of generally ineffective chemotherapy and limited usefulness of somatostatin analogs in hormone-related syndromes. Better knowledge of molecular mechanisms related to carcinogenesis, angiogenesis, and tumor progression have led to the development of specific targeted therapies that will change the armamentarium of treatment of neuroendocrine tumors. In April 2011, everolimus and sunitinib were presented to the Food and Drug Administration for approval in the treatment of advanced PNETs, and everolimus for tumors of gastrointestinal and lung origin, based on the RADIANT-2 results. Selection of one of these drugs for the treatment of advanced PNETs will be based mainly on their toxicity profiles because both drugs are able to double mpfs. Only the sunitinib study has assessed the QoL profile of these patients. Nonetheless, QoL is an important objective in this particular group of tumors because most are pauci- or asymptomatic and therefore drug side effects could jeopardize compliance with long-term treatment. Tumor response does not facilitate the choice between these new drugs because both give less than 10% ORR. Moreover, ORR with these drugs compares unfavorably with those of older studies of cytotoxic therapies in PNETs, which achieved rates of up to 60%. In this regard, it is suggested that these new targeted therapies are reserved for slow-growing tumors and that we should continue using traditional chemotherapy in those patients for whom reduction of tumor burden is the main objective. The vast population that achieves SD in these studies constitutes the main objective of prolonging mpfs, and the real benefit of these stabilizations can only be assessed in placebo-controlled phase III studies with documented disease progression before therapy begins. Furthermore, no relationship has been established between mpfs increase and prolongation of OS. The crossover design in placebo-controlled studies, although ethically mandatory, increases the difficulty of survival interpretation. Despite this, the sunitinib study showed OS gain reflecting the value of stabilizations in heavily pretreated PNET patients. With this scenario, sunitinib and everolimus will become new treatment options for PNETs but are far from becoming the new standard of care for all patients, in whom medical therapy, surgery, and locoregional ablations will continue to play the main role. The development of AUGUST 2011 CANCER DISCOVERY 219

MINI SECTION REVIEW Capdevila et al. predictive biomarkers, such as reduced expression of PTEN or TSC1/2 for mtor inhibition, should be run in parallel for better patient selection and consequently a better balance of clinical benefit, side effects, and resources limitation. New studies are combining everolimus and sunitinib with other targeted agents and cytotoxic drugs to evaluate if combination treatment translates into an increase in survival of patients with advanced PNETs. In this regard, a phase III study is being designed in association with the European Neuroendocrine Tumor Society (ENETS) to assess the importance of sequentiality of everolimus and chemotherapy in this setting. Disclosure of Potential Conflicts of Interest J. Tabernero disclosed consulting and participation in advisory boards for Novartis and Pfizer; J. Capdevila disclosed consulting and participation in advisory boards for Novartis and Pfizer. Received June 24, 2011; accepted July 5, 2011; published online August 16, 2011. REFERENCES 1. Yao JC, Hassan M, Phan A, Dagohoy C, Leary C, Mares JE, et al. 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 72. 2. O Toole D, Couvelard A, Rebours V, Zappa M, Hentic O, Hammel P, et al. Molecular markers associated with response to chemotherapy in gastro-entero-pancreatic neuroendocrine tumors. Endocr Relat Cancer 2010;17:847 56. 3. Missiaglia E, Dalai I, Barbi S, Beghelli S, Falconi M, della Peruta M, et al. Pancreatic endocrine tumors: expression profiling evidences a role for AKT-mTOR pathway. J Clin Oncol 2010;28:245 55. 4. Fjällskog ML, Lejonklou MH, Oberg KE, Eriksson BK, Janson ET. Expression of molecular targets for tyrosine kinase receptor antagonists in malignant endocrine pancreatic tumors. Clin Cancer Res 2003;9:1469 73. 5. Kouvaraki MA, Ajani JA, Hoff P, Wolff R, Evans DB, Lozano R, et al. Fluorouracil, doxorubicin, and streptozocin in the treatment of patients with locally advanced and metastatic pancreatic endocrine carcinomas. J Clin Oncol 2004;22:4762 71. 6. Modlin IM, Oberg K, Chung DC, Jensen RT, de Herder WW, Thakker RV, et al. Gastroenteropancreatic neuroendocrine tumours. Lancet Oncology 2008;9:61 72. 7. Moertel CG, Lefkopoulo M, Lipsitz S, Hahn RG, Klaassen D. Streptozocin-doxorubicin, streptozocin-fluorouracil or chlorozotocin in the treatment of advanced islet-cell carcinoma. N Engl J Med 1992;326:519 23. 8. Yao JC, Phan A, Hoff PM, Chen HX, Charnsangavej C, Yeung SC, et al. Targeting vascular endothelial growth factor in advanced carcinoid tumor: a random assignment phase II study of depot octreotide with bevacizumab and pegylated interferon alfa-2b. J Clin Oncol 2008;26:1316 23. 9. Faivre S, Delbaldo C, Vera K, Robert C, Lozahic S, Lassau N, et al. Safety, pharmacokinetic, and antitumor activity of SU11248, a novel oral multitarget tyrosine kinase inhibitor, in patients with cancer. J Clin Oncol 2006;24:25 35. 10. Kulke MH, Lenz HJ, Meropol NJ, Posey J, Ryan DP, Picus J, et al. Activity of sunitinib in patients with advanced neuroendocrine tumors. J Clin Oncol 2008;26:3403 10. 11. Raymond E, Dahan L, Raoul JL, Bang YJ, Borbath I, Lombard-Bohas C, et al. Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. N Engl J Med 2011;364:501 13. 12. Sabatini DM. MTOR and cancer: insights into a complex relationship. Nat Rev Cancer 2006;6:729 34. 13. Jiao Y, Shi C, Edil BH, et al. DAXX/ATRX, MEN1, and mtor pathway genes are frequently altered in pancreatic neuroendocrine tumors. Science 2011;331:1199 203. 14. Yao JC, Phan AT, Chang DZ, Wolff RA, Hess K, Gupta S, et al. Efficacy of RAD001 (everolimus) and octreotide LAR in advanced low- to intermediate-grade neuroendocrine tumors: results of a phase II study. J Clin Oncol 2008;26:4311 8. 15. Yao JC, Lombard-Bohas C, Baudin E, Kvols LK, Rougier P, Ruszniewski P, et al. 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. 16. Pavel M, Hainsworth J, Baudin E, Peeters M, Hoersch D, Anthony L, et al. A randomized, double-blind, placebo-controlled, multicenter phase III trial of everolimus + octeotride LAR vs placebo + octeotride LAR in patients with advanced neuroendocrine tumors (NET) (RADIANT-2). Ann Oncol 2010;21(Suppl 8):LBA8. 17. Yao JC, Shah MH, Ito T, Bohas CL, Wolin EM, Van Cutsem E, et al. Everolimus for advanced pancreatic neuroendocrine tumors. N Engl J Med 2011;364:514 23. 18. Tee AR, Fingar DC, Manning BD, Kwiatkowski DJ, Cantley LC, Blenis J. Tuberous sclerosis complex-1 and -2 gene products function together to inhibit mammalian target of rapamycin (mtor)-mediated downstream signaling. Proc Natl Acad Sci U S A 2002;99:13571 6. 19. Johannessen CM, Reczek EE, James MF, Brems H, Legius E, Cichowski K. The NF1 tumor suppressor critically regulates TSC2 and mtor. Proc Natl Acad Sci U S A 2005;102:8573 8. 20. Debelenko LV, Zhuang Z, Emmert-Buck MR, Chandrasekharappa SC, Manickam P, Guru SC, et al. Allelic deletions on chromosome 11q13 in multiple endocrine neoplasia type 1-associated and sporadic gastrinomas and pancreatic endocrine tumors. Cancer Res 1997;57:2238 43. 21. Volinia S, Calin GA, Liu CG, Ambs S, Cimmino A, Petrocca F, et al. A microrna expression signature of human solid tumors defines cancer gene targets. Proc Natl Acad Sci U S A 2006;103:2257 61. 22. Roldo C, Missiaglia E, Hagan JP, Falconi M, Capelli P, Bersani S, et al. MicroRNA expression abnormalities in pancreatic endocrine and acinar tumors are associated with distinctive pathologic features and clinical behavior. J Clin Oncol 2006;24:4677 84. 23. Corbo V, Dalai I, Scardoni M, Barbi S, Beghelli S, Bersani S, et al. MEN1 in pancreatic endocrine tumors: analysis of gene and protein status in 169 sporadic neoplasms reveals alterations in the vast majority of cases. Endocr Relat Cancer 2010;17:771 83. 24. Wang Y, Ozawa A, Zaman S, Prasad NB, Chandrasekharappa SC, et al. The tumor suppressor protein menin inhibits AKT activation by regulating its cellular localization. Cancer Res 2011;71:371 82. 25. Corbo V, Beghelli S, Bersani S, Antonello D, Talamini G, Brunelli M, et al. Pancreatic endocrine tumours: mutational and immunohistochemical survey of protein kinases reveals alterations in targetable kinases in cancer cell lines and rare primaries. Ann Oncol 2011 Mar 29. [Epub ahead of print]. 26. Chiu CW, Nozawa H, Hanahan D. Survival benefit with proapoptotic molecular and pathologic responses from dual targeting of mammalian target of rapamycin and epidermal growth factor receptor in a preclinical model of pancreatic neuroendocrine carcinogenesis. J Clin Oncol 2010;28:4425 33. 27. Kulke MH, Hornick JL, Frauenhoffer C, Hooshmand S, Ryan DP, Enzinger PC, et al. O6-Methylguanine DNA methyltransferase deficiency and response to temozolomide-based therapy in patients with neuroendocrine tumors. Clin Cancer Res 2009;15:338 45. 28. Takebe N, Harris PJ, Warren RQ, Ivy SP. Targeting cancer stem cells by inhibiting Wnt, Notch, and Hedgehog pathways. Nat Rev Clin Oncol 2011;8:97 106. 29. Gregorieff A, Clevers H. Wnt signaling in the intestinal epithelium: from endoderm to cancer. Genes Dev 2005;19:877 90. 220 CANCER DISCOVERY AUGUST 2011 www.aacrjournals.org

Treatment of Pancreatic Neuroendocrine Tumors MINI REVIEW 30. Shan L, Aster JC, Sklar J, Sunday ME. Notch-1 regulates pulmonary neuroendocrine cell differentiation in cell lines and in transgenic mice. Am J Physiol Lung Cell Mol Physiol 2007;292:500 9. 31. Venook AP, Ko AH, Tempero MA, Uy J, Weber T, Korn M, et al. Phase II trial of FOLFOX plus bevacizumab in advanced, progressive neuroendocrine tumors. Proc Am Soc Clin Oncol 2008;26:15545. 32. Kunz PL, Kuo T, Kaiser HL, Norton JA, Longacre TA, Ford JM, et al. A phase II study of capecitabine, oxaliplatin, and bevacizumab for metastatic or unresectable neuroendocrine tumors: preliminary results. Proc Am Soc Clin Oncol 2008;26:15502. 33. Kulke MH, Stuart K, Earle CC, Bhargava P, Clark JW, Enzinger PC, et al. A phase II study of temozolomide and bevacizumab in patients with advanced neuroendocrine tumors. Proc Am Soc Clin Oncol 2006;24:4044. 34. Hobday TJ, Rubin J, Holen K, Picus J, Donehower R, Marschke R, et al. MC044h, a phase II trial of sorafenib in patients (pts) with metastatic neuroendocrine tumors (NET): a phase II consortium (P2C) study. Proc Am Soc Clin Oncol 2007;25:4504. 35. Castellano D, Capdevila J, Salazar R, Sastre J, Alonso V, Llanos M, et al. Sorafenib and bevacizumab combination targeted therapy in advanced neuroendocrine tumor: a phase II study of the Spanish Neuroendocrine Tumor Group (GETNE0801). Proc Am Soc Clin Oncol 2011;29:4113. 36. Phan AT, Yao JC, Fogelman DR, Hess KR, Ng CS, Bullock SA, et al. A prospective, multi-institutional phase II study of GW786034 (pazopanib) and depot octreotide (sandostatin LAR) in advanced low-grade neuroendocrine carcinoma (LGNEC). Proc Am Soc Clin Oncol 2010;28:4001. 37. Sung MW, Kvols L, Wolin E, Jacob G, Talluto C, Torres JC, et al. Phase II proof-of-concept study of atiprimod in patients with advanced low- to intermediate-grade neuroendocrine carcinoma. Proc Am Soc Clin Oncol 2008;26:4611. 38. Xu Y, Ellison C, Martin E, Young D, Campbell J, Vinci L, et al. A phase II study of thalidomide in metastatic neuroendocrine tumors. Proc Am Soc Clin Oncol 2002;21:1931. 39. Kulke MH, Stuart K, Enzinger PC, Ryan DP, Clark JW, Muzikansky A, et al. Phase II study of temozolomide and thalidomide in patients with metastatic neuroendocrine tumors. J Clin Oncol 2006; 24:401 6. 40. Kulke MH, Bergsland EK, Ryan DP, Enzinger PC, Lynch TJ, Zhu AX, et al. Phase II study of recombinant human endostatin in patients with advanced neuroendocrine tumors. J Clin Oncol 2006;24:3555 61. 41. Yao JC, Phan AT, Fogleman D, Ng C, Jacobs C, Dagohoy CD, et al. Randomized run-in study of bevacizumab and everolimus in low- to intermediate-grade neuroendocrine tumors using perfusion CT as functional biomarker. Proc Am Soc Clin Oncol 2010;28:4002. AUGUST 2011 CANCER DISCOVERY 221

A Shining Light in the Darkness for the Treatment of Pancreatic Neuroendocrine Tumors Jaume Capdevila and Josep Tabernero Cancer Discovery 2011;1:213-221. Updated version Access the most recent version of this article at: http://cancerdiscovery.aacrjournals.org/content/1/3/213 Cited articles Citing articles This article cites 40 articles, 23 of which you can access for free at: http://cancerdiscovery.aacrjournals.org/content/1/3/213.full#ref-list-1 This article has been cited by 2 HighWire-hosted articles. Access the articles at: http://cancerdiscovery.aacrjournals.org/content/1/3/213.full#related-urls E-mail alerts Sign up to receive free email-alerts related to this article or journal. Reprints and Subscriptions Permissions To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at pubs@aacr.org. To request permission to re-use all or part of this article, use this link http://cancerdiscovery.aacrjournals.org/content/1/3/213. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.