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

Similar documents
MEDICAL MANAGEMENT OF METASTATIC GEP-NET

TRACTAMENT ONCOLÒGIC DELS TUMORS NEUROENDOCRINS METASTÀSICS

Review of Gastrointestinal Carcinoid Tumors: Latest Therapies

Cutting Edge Treatment of Neuroendocrine Tumors

Cutting Edge Treatment of Neuroendocrine Tumors

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

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

SUPPLEMENTARY INFORMATION

2015: Year in Review Results of Recent Trials

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

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

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

Recent developments of oncology in neuroendocrine tumors (NETs)

NET ΠΝΕΥΜΟΝΑ: τι νεότερο / νέες μελέτες

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

Antiangiogenics are effective treatments in NETs

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

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

Nuevas alternativas en el manejo de TNE avanzados

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

Teresa Alonso Gordoa Servicio Oncología Médica Hospital Universitario Ramón y Cajal

NET und NEC. Endoscopic and oncologic therapy

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

Octreotide LAR in neuroendocrine tumours a summary of the experience

OPTIMISING OUTCOMES IN GASTROINTESTINAL NEUROENDOCRINE TUMOURS

GEP NET: algoritmo terapeutico. Dottor Nicola Fazio

Neuroendocrine Tumors Positron Emission Tomography (PET) Imaging and Peptide Receptor Radionuclide Therapy

Evaluation and Management of Neuroendocrine Tumors

Systematic reviews and meta-analyses of observational studies (MOOSE): Checklist.

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

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

Selection of Appropriate Treatment

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

Chair s presentation Lutetium (177lu) oxodotreotide for treating unresectable or metastatic neuroendocrine tumours in people with progressive disease

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

TUMORES NEUROENDOCRINOS. Miguel Navarro. Salamanca

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

Le target therapy nei Tumori Neuroendocrini

Recent Advances in Gastrointestinal Cancers

Targeted Therapies in Metastatic Colorectal Cancer: An Update

Lu 177-Dotatate (Lutathera) Therapy Information

Pancreatic Neuroendocrine Tumours

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

pan-canadian Oncology Drug Review Final Clinical Guidance Report Sunitinib malate (Sutent) for pancreatic neuroendocrine tumours May 3, 2012

Immunotherapy in the Adjuvant Setting for Melanoma: What You Need to Know

EVIDENCE IN BRIEF OVERALL CLINICAL BENEFIT

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

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

Cancer Cell Research 14 (2017)

An Immunotherapy Clinical Trial for NETs

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

EVIDENCE IN BRIEF OVERALL CLINICAL BENEFIT

Pancreatic neuroendocrine cancer with liver metastases and multiple peritoneal metastases: report of one case

Prior Authorization Review Panel MCO Policy Submission

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

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

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

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

PRINCESS MARGARET CANCER CENTRE CLINICAL PRACTICE GUIDELINES

Bevacizumab for the treatment of recurrent advanced ovarian cancer

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

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

A New Proposal for Metabolic Classification of NENs Stefano Severi IRST Meldola Italy

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

EVIDENCE IN BRIEF OVERALL CLINICAL BENEFIT

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

SIRT in the Management of Metastatic Neuroendocrine Tumors

Metastatic renal cancer (mrcc): Evidence-based treatment

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

Technology appraisal guidance Published: 25 February 2015 nice.org.uk/guidance/ta333

Ongoing and future clinical investigation in GEP NENs

2. The effectiveness of combined androgen blockade versus monotherapy.

Surveillance report Published: 17 March 2016 nice.org.uk

Development of New Treatment Modalities Oncolytic Viruses and Nanotechnique

Oberndofer 1907 Illeal Serotonin Secreting Tumor Carcinoid (Karzinoide)

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

The Use of Inhibitors of Angiogenesis in Patients with Inoperable Locally Advanced or Metastatic Renal Cell Cancer: Guideline Recommendations

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

PANCREATIC NEUROENDOCRINE TUMOURS A

New Developments in the Care and Management of Patients with Gastroenteropancreatic Neuroendocrine Tumors Dr. Tim Asmis The Ottawa Hospital Cancer

Bisphosphonates and other bone agents for breast cancer(review)

Collaborative Practice in the Management of Patients With Gastrointestinal and Pancreatic Neuroendocrine Tumors

Systematic Reviews and Meta- Analysis in Kidney Transplantation

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

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

PANCREATIC NEUROENDOCRINE TUMORS DECEMBER 12, 2017 IF YOU EXPERIENCE TECHNICAL DIFFICULTY DURING THE PRESENTATION:

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

David N. Robinson, MD

Appendices. Appendix A Search terms

ENETS Consensus Guidelines for the Standard of Care in

Gastrinoma: Medical Management. Haley Gallup

NEUROENDOCRINE TUMOURS Updated December 2015 by Dr. Doreen Ezeife (PGY-5 Medical Oncology Resident, University of Calgary)

CLINICAL POLICY Department: Medical Management Document Name: Inlyta Reference Number: NH.PHAR.100 Effective Date: 05/12

Case Report. Ameya D. Puranik, MD, FEBNM; Harshad R. Kulkarni, MD; Aviral Singh, MD; Richard P. Baum, MD, PhD ABSTRACT

Evidenze cliniche nel trattamento del RCC

Peptide Receptor Radionuclide Therapy (PRRT) of NET

Management of Neuroendocrine Tumors

LONDON CANCER NEW DRUGS GROUP RAPID REVIEW

CheckMate 025, as patients may derive a benefit, based on the opinion of the CGP and the mechanism of action of nivolumab.

Specialised Services Policy CP66: 68-gallium DOTA- peptide scanning for the Management of Neuroendocrine Tumours (NETs)

Transcription:

At the Cutting Edge Received: February 29, 2016 Accepted: April 12, 2016 Published online: April 16, 2016 Systematic Review of the Role of Targeted Therapy in Metastatic Neuroendocrine Tumors Adrian Lee a, b, e David L. Chan a, b Matthew H. Wong b, f Bob T. Li c, g Sumit Lumba a Stephen J. Clarke a, b, e Jaswinder Samra d Nick Pavlakis a, b, e a Department of Medical Oncology, Royal North Shore Hospital, b Bill Walsh Translational Research Laboratory, Kolling Institute of Medical Research, University of Sydney, c University of Sydney, d Department of Gastrointestinal Surgery, Royal North Shore Hospital and North Shore Private Hospital, University of Sydney, and e Northern Cancer Institute, Sydney, N.S.W., and f Department of Medical Oncology, Gosford Hospital, Gosford, N.S.W., Australia; g Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, N.Y., USA Key Words Neuroendocrine tumor Targeted therapy Everolimus Sunitinib Interferon Octreotide Lanreotide Systematic review Abstract Background: Targeted therapies [interferon (IFN), vascular endothelial growth factor (VEGF) inhibitors, and somatostatin analogs (SSA)] have become an integral part of the neuroendocrine tumor (NET) treatment paradigm. We systematically reviewed the available literature to assess the overall beneficial and negative effects of targeted therapy on progression-free survival (PFS), overall survival (OS), response rate (RR), and toxicity. Methods: Randomized controlled trials (RCT) were identified from MEDLINE, Embase, other major databases, and an electronic search of major conferences. Abstract review, quality assessment, and data abstraction were performed independently by 2 investigators. Metaanalyses were conducted using the generic inverse variance method with a random-effects model, with studies pooled according to drug class and/or control arm for clinical homogeneity. Results: Fifteen RCT [SSA, n = 2; mammalian target of rapamycin (mtor)/vegf inhibitors, n = 4; IFN, n = 3; targeted therapy added to everolimus, n = 2, and other, n = 4] investigating 2,790 patients were included. Overall, targeted agents improved PFS (HR 0.54; 95% CI 0.40 0.73) but not OS (HR 0.86; 95% CI 0.72 1.01). SSA improved PFS (HR 0.41; 95% CI 0.29 0.58) but not OS (HR 1.00; 95% CI 0.58 1.74). mtor/ VEGF inhibitors improved PFS (HR 0.48; 95% CI 0.32 0.72) but not OS (HR 0.82; 95% CI 0.58 1.17). Targeted therapies added to everolimus or IFN did not improve either PFS or OS. The RR overall was improved (OR 2.85; 95% CI 1.77 4.59) but toxicity was increased (meta-analysis not performed). Conclusions: The addition of targeted therapies improves PFS but not OS in NET. The evidence is strongest for VEGF inhibitors and SSA. There is an ongoing need for well-designed RCT to inform the optimal use of targeted therapies in NET. A.L. and D.L.C. contributed equally to this work. 2016 S. Karger AG, Basel E-Mail karger@karger.com www.karger.com/nen 2016 S. Karger AG, Basel Nick Pavlakis Department of Medical Oncology Royal North Shore Hospital St Leonards, NSW 2065 (Australia) E-Mail nick.pavlakis @ sydney.edu.au

Introduction Neuroendocrine tumors (NET) represent a heterogeneous group of tumors that may be difficult both to diagnose and to treat. NET are found in a variety of locations such as the gastroenteropancreatic (GEP) system, along the gastrointestinal tract, and in the respiratory system. The natural history of these tumors is extremely variable, with some being relatively indolent and others aggressive in nature. While some patients present relatively early in the disease trajectory as a result of hormonal symptoms from functioning tumors, delayed presentation and diagnosis with nonfunctional tumors is the norm. Their incidence in recent years has increased globally, likely due to improved detection with gallium-68 and fluorodeoxyglucose positron emission topography (PET) scans as well as increased clinician awareness and vigilance [1, 2]. Their prevalence is also likely to increase with improved management options and consequently an increased survival of patients [3]. From an initial paucity of therapeutic options, the scope has now expanded to include cytotoxic chemotherapy, targeted biological agents, radioactive labeled delivery, and intra-arterial liver-directed therapies. Despite increasing recognition that this tumor group is histologically and biologically different from the more common epithelial cell tumors, a separate staging system for NET has only recently been established by the American Joint Committee on Cancer [4]. There is increasing knowledge that poorly differentiated (G3) tumors behave differently from well-differentiated (G1/G2) ones, and markers of proliferation such as the mitotic count and Ki-67 are increasingly used to guide both prognostication and the selection of optimal treatments [5]. We undertook this systematic review of the current evidence on targeted therapy for NET, with the exclusion of peptide receptor radionuclide therapy (PRRT) as this has been reviewed recently [6]. We analyzed the evidence using the Cochrane method in order to evaluate the overall benefits and harms of treatment to establish a clearer path in the decision-making process for a patient with metastatic well-differentiated NET. Methods Criteria for Consideration of Studies for Review We searched for randomized clinical trials that included adult patients (aged 18 years and over) with a confirmed histological diagnosis of well-differentiated, advanced (locally advanced/unresectable or metastatic) GEP NET with an Eastern Cooperative Oncology Group (ECOG) performance status score of 0 2 and radiologically measurable disease. Eligible studies included patients treated with targeted therapy compared to no targeted therapy or placebo, targeted therapy with another nontargeted therapy, and targeted therapy and other therapy compared to targeted therapy. Targeted therapy was defined to include somatostatin analogs (SSA), monoclonal antibodies, tyrosine kinase inhibitors, mammalian target of rapamycin (mtor) inhibitors, and immunotherapy. PRRT-related studies were also excluded from this paper due to a recent comprehensive review on this area by Bodei et al. [6]. The primary endpoint was progression-free survival (PFS), and secondary endpoints included overall survival (OS), time to tumor failure or progression, objective response rates (RR), rates of toxicity, and quality of life. Search Methods for the Identification of Studies We searched for trials consistent with our inclusion criteria in MEDLINE from 1946 to December 1, 2015, and in Embase from 1996 to December 1, 2015. The searches for patient population, intervention, and systematic or narrative reviews were combined separately, and limits were applied to adults aged 18 years or older. The database search also included the Cochrane Database of Systematic Reviews, the Cochrane Central Register of Controlled Trials, the Cochrane Methodology Register, the ACP Journal Club, and databases of abstracts of reviews of effects and abstracts from major conferences, including the American Society of Clinical Oncology (ASCO), the European Society of Medical Oncology (ESMO), the North American Neuroendocrine Tumor Society, and the European Neuroendocrine Tumor Society. In addition, we searched through relevant journals including the Journal of Clinical Oncology, Annals of Oncology, Lancet, Lancet Oncology, the New England Journal of Medicine, the European Journal of Cancer, Neuroendocrinology, and Pancreas. All members of the author group were asked to identify additional trials. Only trial reports in the English language were included. Two review authors (A.L. and D.L.C.) performed searches independently to identify eligible trials for inclusion. Data Collection and Analysis Studies identified from the MEDLINE and Embase searches were screened for randomized controlled trials (RCT) investigating targeted therapy. Uncertainties were resolved via further investigation into full-text articles. All possibly relevant articles were retrieved in full text and assessed with reference to the inclusion criteria. Study quality and the risk of bias were assessed by at least 2 authors (A.L., D.L.C., and M.H.W.) independently using Cochrane criteria. Where the primary references did not provide sufficient details, we resorted to secondary references, abstracts, presentations, or protocols. Disagreements were resolved after discussion with a third reviewer (N.P.). A meta-analysis was performed on the endpoints listed above, appreciating that some statistical heterogeneity might occur from the pooling of trials investigating different therapies. The statistical analysis was performed using standard meta-analytical techniques, with calculations using a random-effects model given the heterogeneity of the patient characteristics and the treatments investigated. HR and OR were calculated where feasible with 95% CI. Heterogeneity, defined by I 2 >30% or p < 0.10, was further explored. 210 Lee/Chan/Wong/Li/Lumba/Clarke/ Samra/Pavlakis

558 records identified through the MEDLINE database 548 records identified through an Embase database search 146 records identified through an EBM review database** 6 additional records identified through hand searches* 976 records after removal of duplicates 708 records excluded not NET related 169 records excluded not RCT 99 records for abstract review Excluded: 4 chemotherapy-related articles 20 conference abstracts related to full-text articles describing the same trial 37 subgroup analysis reports 15 update reports on published trials 3 nonrelated Fig. 1. Selection of studies. * Abstracts from major oncology and NET conferences; major journals including the Journal of Clinical Oncology, Annals of Oncology, Lancet, Lancet Oncology, and the New England Journal of Medicine. * * Cochrane Database of Systematic Reviews, Cochrane Central Register of Controlled Trials, Cochrane Methodology Register, ACP Journal Club, Database of Abstracts of Reviews of Effects, health technology assessments, and NHS Economic Evaluation Database. EBM = Evidence-based medicine. 20 articles assessed for eligibility in full 15 studies included in the qualitative synthesis 11 studies included in the quantitative analysis (meta-analysis) 5 articles excluded: 2 were not randomized trials 2 compared different preparations of the same investigational product 1 was retracted Results Study Selection The study flow is summarized in a CONSORT diagram ( fig. 1 ). A total of 976 records were identified after de-duplication. Twenty articles were assessed for eligibility. Five articles were removed upon full evaluation (detailed below), leaving 15 studies for qualitative synthesis and 11 for meta-analysis. Included Studies Fifteen trials (2,790 patients; table 1 ) investigated the role of targeted therapy in the treatment of advanced well-differentiated NET. Eleven studies (2,172 patients) were included in the meta-analysis, as 4 studies were not included for reasons detailed below. Two studies investigated the impact of SSA compared to placebo. Rinke et al. [7] investigated octreotide longacting repeatable (LAR) against placebo in midgut NET. Caplin et al. [8] investigated lanreotide versus placebo in GEP NET. Four studies investigated agents directed against either the mtor or the vascular endothelial growth factor (VEGF) pathways. Pavel et al. [9] compared everolimus in combination with octreotide LAR against octreotide LAR alone in mixed NET, Yao et al. [10] compared everolimus with placebo in patients with pancreatic NET (pnet), and Yao [11] compared everolimus with placebo in nonpancreatic NET patients (including primary pulmonary NET). Raymond et al. [12] compared sunitinib against placebo in pnet. Three studies, i.e. those of Arnold et al. [13], Faiss et al. [14], and Kolby et al. [15], investigated the impact of the addition of interferon (IFN)-α in either midgut or mixed NET. Targeted Therapy in NET 211

Table 1. Included studies and baseline characteristics Trial Intervention Comparator Participants randomized, n Mean age, years Primary tumor sites Liver involvement, % Included in the quantitative analysis? Rinke et al. [7] octreotide LAR (30 mg) placebo 85 62 midgut 86 yes Caplin et al. [8] lanreotide autogel (120 mg) placebo 204 54 GEP 83 yes Yao et al. [10] everolimus placebo 410 58 pancreas 92 yes Pavel et al. [9] everolimus + octreotide LAR placebo + octreotide LAR 429 60 mixed 92 yes Yao [11] everolimus placebo 302 63 lung/ 84 yes gastrointestinal Raymond et al. [12] sunitinib placebo 171 57 pancreas 95 yes Arnold et al. [13] IFN-α + octreotide octreotide 109 58 mixed NR yes Kolby et al. [15] octreotide + IFN-α octreotide 68 62 midgut 100 (all yes had TACE) Faiss et al. [14] IFN-α +/ lanreotide lanreotide 84 58 mixed 91 yes Kulke et al. [16] everolimus + pasireotide everolimus 160 58 pancreas NR yes Kulke et al. [17] everolimus + bevacizumab everolimus 150 59 pancreas NR yes Wolin et al. [18] pasireotide octreotide 110 62 mixed NR no Yao et al. [19] bevacizumab + octreotide IFN-α-2b + octreotide 44 55 mixed 95 no Yao et al. [20] bevacizumab + octreotide IFN-α-2B + octreotide 402 NR mixed NR no Salazar et al. [21] BEZ235 everolimus 62 (part 1) NR pancreas NR no TACE = Transarterial chemoembolization; NR = not reported. Two studies from 2015 in abstract form investigated the addition of targeted therapy to an everolimus backbone. Kulke et al. [16] investigated the addition of a combination of an SSA (pasireotide) and everolimus compared to everolimus alone in advanced pnet. Kulke et al. [17] also enrolled patients with advanced pnet, but they investigated the impact of adding bevacizumab to everolimus. The results reported in the above studies are summarized in table 2. Studies Included in the Qualitative Synthesis but Not in the Meta-Analysis Wolin et al. [18] investigated pasireotide LAR against octreotide LAR in metastatic functional NET. Their primary outcome was symptom control, i.e. bowel movements and flushing episodes, and no significant differences were observed between the 2 groups. Pasireotide was shown to achieve symptom control in 20.9% of cases and octreotide LAR in 26.7%. The tumor control rate in the 6th month was higher in the pasireotide arm (62.7 vs. 46.2%). PFS, calculated in a post hoc analysis, favored pasireotide LAR (11.8 vs. 6.8 months). Given that that study compared 2 SSA rather than an SSA versus placebo, it was not included with the other studies in the meta-analysis. Yao et al. [19] compared bevacizumab and IFN-α as part of a phase II study. Given that this study investigated the use of one targeted agent compared to another (as opposed to the addition of a targeted agent in the other 8 studies), it was reported separately instead of being incorporated into the meta-analysis. Enrolled patients had stable octreotide doses before randomization to the addition of either bevacizumab or PEG IFN-α-2b. The primary endpoint was the RR. At progression or after 18 weeks, the patients entered phase II and were given both drugs. The RR was 4/22 in the bevacizumab arm and 0/22 in the IFN arm. In the IFN arm, 2/22 patients achieved a partial response after the addition of bevacizumab in phase II. The median PFS was 66 weeks in the bevacizumab arm and 56 weeks in the IFN arm (p = 0.34). The main differing toxicities between the 2 arms included increased granulocytopenia in the PEG IFN arm (14 vs. 0%, p = 0.02) and hypertension in the bevacizumab arm (18 vs. 0%, p = 0.01). The phase III RCT that continued to investigate the above combinations, i.e. SWOG S0518 [20], reported preliminary results at the 2015 ASCO Annual Meeting. PFS was not significantly different between the arms, being 16.6 months in the bevacizumab and 15.4 months in the IFN arm. Increased rates of hypertension and proteinuria were noted in the bevacizumab arm, and increased fatigue and neutropenia were seen in the IFN arm. That study was not included in the quantitative synthesis for the same reasons. The study of Salazar et al. [21] was a phase II randomized trial with a 2-stage design investigating the activity of BEZ235, a dual PI3K/mTOR inhibitor. Patients with advanced pnet were initially randomized to everolimus or BEZ235. Those who were randomized to the everolimus arm were eligible for a second stage in which patients 212 Lee/Chan/Wong/Li/Lumba/Clarke/ Samra/Pavlakis

Table 2. Included studies and outcome measures Trial PFS OS Time to tumor failure/progression, months Objective RR, % Rinke et al. [7] not reported HR 0.81, p = 0.77 14.3 vs. 6 2.4 vs. 2.3 b Caplin et al. [8] HR 0.47, p = 0.0002, no difference, p = 0.88 not reached vs. 18 not reached vs. 18 months not reached vs. 18 months Yao et al. [10] HR 0.35, p < 0.001, HR 1.05, p = 0.59 not reported 5 vs. 2 11 vs. 4.6 months Pavel et al. [9] HR 0.77, p = 0.026, HR 1.22 not reported 2.4 vs. 2 b 16.4 vs. 11.3 months Yao [11] HR 0.48, p < 0.00001, median not reached not reported 2 vs. 1 11.0 vs. 3.9 months Raymond et al. [12] HR 0.42, p < 0.001, HR 0.41, p = 0.02 not reported 9.3 vs. 0 11.4 vs. 5.5 months Arnold et al. [13] not clearly reported c HR 0.82, p = 0.38 not clearly reported 50 vs. 45 Kolby et al. [15] not reported HR 0.62, p = 0.132 not reported not reported Faiss et al. [14] not reported not reported not clearly reported c, no difference as reported by authors lanreotide, 4; IFN-α, 3.7; combination, 7.1 Kulke et al. [16] HR 0.99, p = 0.488, median not reached not reported 20.3 vs. 6.2 16.82 vs. 16.59 months Kulke et al. [17] HR 0.80, p = 0.12, HR 0.75, p = 0.16, not reported 31 vs. 12 16.7 vs. 14 months 36.7 vs. 35 months Wolin et al. [18] HR 0.46, p = 0.045, not reported not reported 2 vs. 3.8 11.8 vs. 6.8 months a Yao et al. [19] 16.5 vs. 14 months, no difference observed; not reported 18 vs. 0 b p = 0.34 exact figure not reported Yao et al. [20] HR 0.93, p = 0.55, not reported 9.9 vs. 5.6, HR 0.72, p = 0.003 12 vs. 4 16.6 vs. 15.4 months Salazar et al. [21] HR 1.53 (95% CI 0.72 3.25), 8.2 vs. 10.8 months not reported not reported 10 vs. 10 a Post hoc analysis. b Not reported but calculated from raw data presented in the report. c Actual number not reported. Kaplan-Meier curve presented. were randomized to either BEZ235 or best supportive care. The results were presented at the 2015 ASCO Annual Meeting. No statistically significant difference was recorded in terms of the median PFS in the first stage (8.2 months in the BEZ235 arm compared to 10.8 months in the everolimus arm). The median PFS has not been presented for stage 2 to date (the PFS at 16 weeks for BEZ235 was 52%). Although stage 2 would be eligible for inclusion at a future date, insufficient results have been reported to date; therefore, we excluded this study from the quantitative analysis until further data are available. Excluded Studies There were 5 excluded studies. The studies of Bajetta et al. [22] and Rubin et al. [23] were excluded as they focused on showing equivalence in response and symptom control of the same drug compound in different preparations. Bajetta et al. [22] tested lanreotide autogel versus lanreotide microparticles, and Rubin et al. [23] used octreotide LAR versus octreotide acetate. The report of Bajetta et al. [24] (investigating treatment with everolimus and octreotide in GEP and pulmonary NET populations) was not an RCT and was thus excluded. The report of Yao et al. [25] was a phase II study investigating everolimus versus placebo in patients with pnet. Although the results were fully published and the study was eligible for inclusion, this was subsequently retracted, and thus we have excluded it from the analysis for the time being. We note a recent study published by Phan et al. [26] investigating the combination of pazopanib long-acting octreotide in 52 patients with advanced well-differentiated NET, with an objective response in 7/32 patients with pancreatic primaries. Given that this was a single-arm trial, it was excluded from the meta-analysis. Ongoing Studies The ongoing studies identified by the literature search are summarized in table 3. Targeted Therapy in NET 213

Table 3. Ongoing (unreported) phase II/III randomized controlled studies involving targeted therapies (non-prrt, no chemotherapy) for well-differentiated GEP NET National clinical trial ID No. (study name) Phase Patients, n Intervention Comparator Population Study start Primary completion Status NCT02031536 II 150 everolimus placebo pancreatic NET with liver metastases previously treated with surgery January 2014 October 2016 NCT01841736 II 165 pazopanib placebo progressive carcinoid tumors June 2013 December 2016 NCT01744249 II 80 sandostatin LAR + placebo advanced well-differentiated November December axitinib nonpancreatic NET 2011 2013 NCT01731925 II 104 lanreotide + lanreotide + advanced midgut carcinoid December March (SUNLAND) sunitinib placebo tumors 2012 2016 NCT01755182 II 140 TAE octreotide metastatic NET to the liver: with July 2013 December (LOTUS) LAR or without upfront TAE 2017 NCT01678664 II 72 everolimus TACE digestive endocrine tumor October April 2017 (EVACEL) (doxorubicin) 2012 NCT00781911 II 43 IMC-A12 octreotide islet cell cancer February July 2011 depot 2009 NCT02288377 II/III 222 lanreotide placebo duodenopancreatic NET September June 2017 2014 NCT02231762 II 40 lanreotide + placebo GEP NET February October temozolomide 2015 2018 NCT02031536 II 150 everolimus placebo pancreatic NET: after liver January October metastasis resection 2014 2016 NCT02246127 III 180 everolimus STZ-5FU pancreatic NET October September 2014 2018 not yet active, not TAE = Transarterial embolization; TACE = transarterial chemoembolization. Quality of the Trials On the whole, the quality of the trials was good. Of the 15 trials included ( table 4 ), 8 were of good quality. While minor issues were noted with regard to blinding, these did not affect the assessment of the results. For instance, the report of Arnold et al. [13] was an open-label study with the primary endpoint of time to treatment failure, but radiological assessment was blinded. In the study of Wolin et al. [18], complete double blinding was not achievable due to different appearances of LAR formulations, but only the independent study nurse/coordinator was privy to the assignment, with the participant, investigator, and assessors remaining blinded throughout the study. Yao [11] has yet to report on all of his prespecified outcome measures (such as quality of life and biomarkers), but he has stated that he will do so in future publications. Four studies, i.e. those of Kulke et al. [16, 17], Yao et al. [20], and Salazar et al. [21], have only been reported in abstract form thus far, with insufficient information to fully assess the risk of bias. Three studies were considered to have a moderate to high risk of bias, i.e. those of Yao et al. [19], Kolby et al. [15], and Faiss et al. [14]. Yao et al. [19] randomized patients but did not specify blinding of patients/investigators to the treatment arms or to the tumor response. The study of Faiss et al. [14] was also an open-label RCT with assessment of cases by unblinded local investigators. Of the 80 cases, 13 were considered critical and were reviewed independently by a blinded radiologist, but the rest were not, resulting in assessment of a high risk of bias. The report of Kolby et al. [15] similarly was an open-label RCT without an independent blinded review of radiology and PFS as one of the stated endpoints. Publication Bias A funnel plot for PFS ( fig. 2 ) revealed a possible mild publication bias toward trials which favored a benefit from targeted therapy. Progression-Free Survival Overall, 10 RCT (investigating 2,088 patients) investigated the effect of targeted therapy for NET on PFS. The 214 Lee/Chan/Wong/Li/Lumba/Clarke/ Samra/Pavlakis

Table 4. Risk-of-bias assessment of the included studies Trial Random sequence generation Allocation concealment Blinding of participants and personnel Blinding of assessors Incomplete outcome data Selective reporting Other bias Rinke et al. [7] + + + + + + + Caplin et al. [8] + + + + + + + Yao et al. [10] + + + + + + + Pavel et al. [9] + + + + + + + Yao [11] + + + + +? + Raymond et al. [12] + + + + + + + Arnold et al. [13] + + + + + Kolby et al. [15] + + + + Faiss et al. [14] + + Kulke et al. [16]??????? Kulke et al. [17]??????? Wolin et al. [18] + + + +? + Yao et al. [19] + + + + Yao et al. [20]??????? Salazar et al. [21]??????? + = Low risk of bias;? = unclear risk of bias; = high risk of bias. 0 SE (log HR) 0.1 0.2 0.3 Subgroups SSA IFN mtor/vegf inhibitors Targeted therapies added to everolimus 0.4 Fig. 2. Funnel plot for the included trials PFS. 0.5 0.01 0.1 1 HR 10 100 addition of targeted therapy improved PFS with an HR of 0.54 (95% CI 0.40 0.73, p < 0.0001; fig. 3 ). Significant statistical heterogeneity was present (p < 0.00001, I 2 = 83%), likely attributable to pooling of studies investigating different treatment modalities (IFN, VEGF inhibitors, and SSA). Use of the leave-one-out strategy only resulted in a decrease in heterogeneity to I 2 = 79%, with the exclusion of Yao et al. [10]. Therefore, these results should be interpreted with caution. Considering the different classes of targeted therapy separately, the addition of SSA (2 studies and 289 patients), compared to placebo, improved PFS with an HR of 0.41 (95% CI 0.29 0.58, p < 0.0001). No significant statistical heterogeneity was present. The addition of mtor/vegf inhibitors (4 studies and 1,312 patients) improved PFS with an HR of 0.48 (95% CI 0.32 0.72, p = 0.0003). Significant statistical heterogeneity was present (p < 0.0001, I 2 = 84%), likely due to the pooling of studies investigating these agents as monotherapy and studies investigating them in combination with other active agents (e.g. Pavel et al. [9] ). Restriction to those trials investigating monotherapy versus best Targeted Therapy in NET 215

Study or subgroup log HR SE Weight (%) HR (IV, random, 95% CI) HR IV, random, 95% CI 1.1.1. SSA Caplin et al. [8] 0.7593 0.2269 9.7 0.47 (0.30 0.73) Rinke et al. [7] 1.0685 0.276 8.7 0.34 (0.20 0.59) Subtotal 18.4 0.41 (0.29 0.58) Heterogeneity: τ 2 = 0.00, χ 2 = 0.75, d.f. = 1, p = 0.39, I 2 = 0% Test for overall effect: Z = 5.04, p < 0.00001 1.1.2. mtor/vegf inhibitors Pavel et al. [9] 0.2638 0.1346 11.2 0.77 (0.59 1.00) Raymond et al. [12] 0.8813 0.2376 9.5 0.41 (0.26 0.66) Yao et al. [10] 1.084 0.1342 11.2 0.34 (0.26 0.44) Yao [11] 0.7251 0.1657 10.7 0.48 (0.35 0.67) Subtotal 42.7 0.48 (0.32 0.72) Heterogeneity: τ 2 = 0.14, χ 2 = 19.28, d.f. = 3, p = 0.0002, I 2 = 84% Test for overall effect: Z = 3.58, p = 0.0003 1.1.3. IFN Arnold et al. [13] 0.1118 0.2107 10.0 1.12 (0.74 1.69) Faiss et al. [14] 0 0 not estimable Kolby et al. [15] 1.3155 0.2638 9.0 0.27 (0.16 0.45) Subtotal 18.9 0.55 (0.14 2.24) Heterogeneity: τ 2 = 0.96, χ 2 = 17.87, d.f. = 1, p < 0.0001, I 2 = 94% Test for overall effect: Z = 0.83, p = 0.41 1.1.4. Targeted therapies added to everolimus Kulke et al. [16] 0.0073 0.224 9.7 0.99 (0.64 1.54) Kulke et al. [17] 0.2204 0.1926 10.3 0.80 (0.55 1.17) Subtotal 20.0 0.88 (0.66 1.17) Heterogeneity: τ 2 = 0.00, χ 2 = 0.52, d.f. = 1, p = 0.47, I 2 = 0% Test for overall effect: Z = 0.89, p = 0.37 Total 100.0 0.54 (0.40 0.73) Heterogeneity: τ 2 = 0.18, χ 2 = 54.25, d.f. = 9, p < 0.00001, I 2 = 83% Test for overall effect: Z = 4.04, p < 0.0001 Test for subgroup differences: χ 2 = 12.52, d.f. = 3, p = 0.006, I 2 = 76.0% 0.01 0.1 1 10 Favors (experimental) Favors (control) 100 Fig. 3. Forest plot PFS. supportive care resulted in no further heterogeneity being present (p = 0.24, I 2 = 31%) and persistence of a PFS benefit (HR 0.40, 95% CI 0.32 0.50, p < 0.00001). The addition of IFN (2 studies and 177 patients) did not significantly improve PFS with an HR of 0.55 (95% CI 0.14 2.24, p = 0.41). Significant statistical heterogeneity was also present (p < 0.0001, I 2 = 94%), possibly due to the selection of NET of mixed primaries in the study of Arnold et al. [13] compared to restriction to midgut NET in the study of Kolby et al. [15]. Because there were only 2 studies, exclusion of either study resulted in no further heterogeneity. This result should be interpreted with caution given the significant heterogeneity and the relatively low number of patients investigated. The addition of targeted therapies to an everolimus backbone (2 trials and 310 patients) did not significantly improve PFS with an HR of 0.88 (95% CI 0.66 1.17, p = 0.37). No significant statistical heterogeneity was present. Sensitivity analysis excluding studies considered to have a moderate-to-high risk of bias demonstrated persistence of improvements in PFS with an HR of 0.52 (95% CI 0.37 0.72, p < 0.0001). 216 Lee/Chan/Wong/Li/Lumba/Clarke/ Samra/Pavlakis

Study or subgroup log HR SE Weight, % HR (IV, random, 95% CI) HR IV, random, 95% CI 1.2.1. SSA Caplin et al. [8] 0.1017 0.3387 6.2 1.11 (0.57 2.15) Rinke et al. [7] 0.2123 0.506 2.9 0.81 (0.30 2.18) Subtotal 9.0 1.00 (0.58 1.74) Heterogeneity: τ 2 = 0.00, χ 2 = 0.27, d.f. = 1, p = 0.61, I 2 = 0% Test for overall effect: Z = 0.02, p = 0.99 1.2.2. mtor/vegf inhibitors Pavel et al. [9] 0.061 0.1514 25.3 1.06 (0.79 1.43) Raymond et al. [12] 0.8886 0.3939 4.6 0.41 (0.19 0.89) Yao et al. [10] 0.0479 0.1992 16.1 1.05 (0.71 1.55) Yao [11] 0.4338 0.2462 11.1 0.65 (0.40 1.05) Subtotal 57.2 0.82 (0.58 1.17) Heterogeneity: τ 2 = 0.07, χ 2 = 7.53, d.f. = 3, p = 0.06, I 2 = 60% Test for overall effect: Z = 1.09, p = 0.28 1.2.3. IFN Arnold et al. [13] 0.1996 0.2318 12.4 0.82 (0.52 1.29) Faiss et al. [14] 0 0 not estimable Kolby et al. [15] 0.4801 0.3207 6.8 0.62 (0.33 1.16) Subtotal 19.2 0.74 (0.51 1.08) Heterogeneity: τ 2 = 0.00, χ 2 = 0.50, d.f. = 1, p = 0.48, I 2 = 0% Test for overall effect: Z = 1.57, p = 0.12 1.2.4. Targeted therapies added to everolimus Kulke et al. [16] 0.0741 0.3251 6.7 0.93 (0.49 1.76) Kulke et al. [17] 0.3347 0.2967 7.9 0.72 (0.40 1.28) Subtotal 14.6 0.81 (0.52 1.24) Heterogeneity: τ 2 = 0.00, χ 2 = 0.35, d.f. = 1, p = 0.55, I 2 = 0% Test for overall effect: Z = 0.99, p = 0.32 Total 100.0 0.86 (0.72 1.01) Heterogeneity: τ 2 = 0.01, χ 2 = 9.88, d.f. = 9, p = 0.36, I 2 = 9% Test for overall effect: Z = 1.79, p = 0.07 Test for subgroup differences: χ 2 = 0.79, d.f. = 3, p = 0.85, I 2 = 0% 0.01 0.1 1 10 Favors (experimental) Favors (control) 100 Fig. 4. Forest plot OS. Overall Survival Overall, 10 RCT (investigating 2,088 patients) investigated the effect of targeted therapy for NET on OS. The addition of targeted therapy did not improve OS with an HR of 0.86 (95% CI 0.72 1.01, p = 0.07; fig. 4 ). No significant statistical heterogeneity was present. Considering the different classes of targeted therapy separately, the addition of SSA (2 studies and 289 patients), compared to placebo, did not improve OS with an HR of 1.00 (95% CI 0.58 1.74, p = 0.99). No significant statistical heterogeneity was present. The addition of VEGF inhibitors (5 studies and 1,462 patients) did not improve OS with an HR of 0.81 (95% CI 0.60 1.09, p = 0.17). Mild statistical heterogeneity (I 2 = 51%, p = 0.09) was present, likely due to the combination of studies investigating agents with different targets of action. The addition of IFN (2 studies and 177 patients) did not improve OS with an HR of 0.74 (95% CI 0.51 1.08, p = 0.12). No significant statistical heterogeneity was present. Targeted Therapy in NET 217

Study or subgroup Experimental, Control, n n ev ents total events total Weight, % OR (M-H, random, 95% CI) OR M-H, Random, 95% CI 1.3.1. SSA Caplin et al. [8] 0 0 0 0 not estimable Rinke et al. [7] 1 42 1 43 2.9 1.02 (0.06 16.93) Subtotal 42 43 2.9 1.02 (0.06 16.93) Total events 1 1 Heterogeneity: not applicable Test for overall effect: Z = 0.02, p = 0.99 1.3.2. mtor/vegf inhibitors Pavel et al. [9] 5 216 4 213 12.9 1.24 (0.33 4.68) Raymond [12] 9 86 0 85 2.8 20.96 (1.20 366.16) Yao et al. [10] 10 191 4 189 16.4 2.56 (0.79 8.29) Yao [11] 4 205 1 97 4.7 1.91 (0.21 17.32) Subtotal 698 584 36.8 2.30 (0.96 5.52) Total events 28 9 Heterogeneity: τ 2 = 0.12, χ 2 = 3.48, d.f. = 3, p = 0.32, I 2 = 14% Test for overall effect: Z = 1.86, p = 0.06 1.3.3. IFN Arnold et al. [13] 5 54 1 51 4.8 5.10 (0.58 45.26) Faiss et al. [14] 2 28 1 27 3.8 2.00 (0.17 23.44) Kolby et al. [15] 0 0 0 0 not estimable Subtotal 82 78 8.5 3.38 (0.66 17.30) Total events 7 2 Heterogeneity: τ 2 = 0.00, χ 2 = 0.31, d.f. = 1, p = 0.57, I 2 = 0% Test for overall effect: Z = 1.46, p = 0.14 1.3.4. Targeted therapies added to everolimus Kulke et al. [16] 16 79 5 81 20.3 3.86 (1.34 11.12) Kulke et al. [17] 23 75 9 75 31.4 3.24 (1.38 7.60) Subtotal 154 156 51.7 3.47 (1.79 6.74) Total events 39 14 Heterogeneity: τ 2 = 0.00, χ 2 = 0.06, d.f. = 1, p = 0.80, I 2 = 0% Test for overall effect: Z = 3.68, p = 0.0002 Total 976 861 100.0 2.85 (1.77 4.59) Total events 75 26 Heterogeneity: τ 2 = 0.00, χ 2 = 4.92, d.f. = 8, p = 0.77, I 2 = 0% Test for overall effect: Z = 4.29, p < 0.0001 Test for subgroup differences: χ 2 = 1.12, d.f. = 3, p = 0.77, I 2 = 0% 0.01 0.1 1 10 Favors (experimental) Favors (control) 100 Fig. 5. Forest plot RR. The addition of targeted therapies to an everolimus backbone (2 trials and 310 patients) did not significantly improve OS with an HR of 0.81 (95% CI 0.52 1.24, p = 0.32). No significant statistical heterogeneity was present. Sensitivity analysis excluding studies considered to have a moderate-to-high risk of bias demonstrated no improvement in OS with an HR of 0.87 (95% CI 0.70 1.09, p = 0.23). Response Rate An RR was reported in 9 studies (1,837 patients). The odds of a radiological tumor response were increased with an OR of 2.85 (95% CI 1.77 4.59, p < 0.0001; fig. 5 ) and pooled rates of 75/976 (7.7%) and 26/861 (3.0%), respectively, resulting in a net RR increase of 4.7%. No significant statistical heterogeneity was present. 218 Lee/Chan/Wong/Li/Lumba/Clarke/ Samra/Pavlakis

Table 5. CgA response rates of the included studies Trial Definition of CgA response RR (experimental arm) RR (control arm) p value Arnold et al. [13] decrease to >50% from baseline 8/16 4/12 0.46 Faiss et al. [14] not reported NS Pavel et al. [9] decrease to normal or >50% from baseline 75/164 53/146 0.0041 Caplin et al. [8] decrease to >50% from baseline 27/64 3/64 <0.0001 Rinke et al. [7] normalization of CgA 9/26 4/30 0.11 NS = Not significant. Sensitivity analysis excluding studies considered to have a moderate-to-high risk of bias confirmed persistent RR improvement with an OR of 2.33 (95% CI 1.14 4.76, p = 0.02). Biochemical Response The chromogranin A (CgA) response criteria and rates, where reported, are summarized in table 5. Given the different definitions of CgA response, a meta-analysis was not performed. Decreases in 5-hydroxyindoleacetic acid (5-HIAA) were seldom reported, but in studies where they were reported they mirrored CgA RR (data not shown). Toxicity Given the variable reporting criteria and measures of toxicity, the decision was made to report toxicity outcomes in descriptive terms rather than quantitatively in a meta-analysis. Caplin et al. [8] recorded similar rates of diarrhea, which was the most common adverse event, in both arms. Both groups had 3 patients each who dropped out of the trials due to adverse events. Rinke et al. [7] reported higher discontinuation rates in the octreotide LAR arm compared to placebo. Serious adverse events (most commonly gastrointestinal symptoms) were otherwise balanced in both arms. Pavel et al. [9] reported higher rates of severe adverse events (57%) and therapy discontinuation (19%) in the experimental arm. The most common severe adverse events included stomatitis, fatigue, diarrhea, hyperglycemia, and thrombocytopenia. Yao et al. [10] reported a 13% discontinuation rate in the everolimus arm, with the most common grade 3 or 4 toxicities in the everolimus arm being stomatitis (7%), anemia (6%), and hyperglycemia (5%). Yao [11] demonstrated toxicity profiles similar to those found in previous everolimus trials. Patients in the everolimus arm reported more toxicity events (i.e. stomatitis, diarrhea, fatigue, and infection) compared to those in the placebo group. All individual grade 3 or 4 events had a frequency of less than 10%, and the discontinuation rate in the everolimus arm was also comparable to previous data at 12%. Raymond et al. [12] reported an increase in grade 3 or 4 events in the sunitinib arm compared to placebo, with the most common events being neutropenia (12%), hypertension (10%), and palmar-plantar erythrodysesthesia (6%). Arnold et al. [13] reported discontinuation of treatment due to side effects in 20% of the patients in the experimental arm. Faiss et al. [14] reported side effects leading to interruption of therapy in 25% of the patients in the IFN + lanreotide arm, 15% of the patients in the IFN arm, and 12% of the patients in the lanreotide arm. Kolby et al. [15] reported 1 patient in the experimental arm with a severe side effect without further clarification. In the study of Kulke et al. [16], both groups had a significant incidence of overall grade 3 4 toxicity, i.e. 77% in the combination arm compared to 69% in everolimus alone group. In another study of Kulke et al. [17] (CALGB80701), the combination of everolimus and bevacizumab resulted in a higher frequency of grade 3 or 4 adverse events, including diarrhea, hyponatremia, hypophosphatemia, proteinuria, and hypertension. In the other bevacizumab trials, i.e. those by Yao et al. [19, 20], adverse events were as expected, with the bevacizumab group recording more hypertension, proteinuria, and headaches and the IFN group having more neutropenia, leukopenia, and nausea. Fatigue occurred in both groups with more events recorded against IFN. As expected, fatigue increased in frequency in the study of Yao et al. [20] when the 2 cohorts merged (octreotide + bevacizumab + IFN). Targeted Therapy in NET 219

Table 6. Quality-of-life results of the included studies Trial Scale used Global score change Other subscales Arnold et al. [13] EORTC QLQ-C30 octreotide, +11.4 ; octreotide + IFN, 6.4 (p = 0.003) Raymond et al. [12] EORTC QLQ-C30 no overall difference in global score worsening of diarrhea (21.4 points, p < 0.001) and clinically insignificant worsening of insomnia (7.8 points, p = 0.04) in the sunitinib arm Caplin et al. [8] EORTC QLQ-C30 0.31 (95% CI 5.73 to 5.10), p = NS in the SSA group Rinke et al. [7] EORTC QLQ-C30 2.1 (95% CI 7.8 to 12.0), p = 0.67 in the SSA group NS = Not significant. In the study of Salazar et al. [21], more adverse events were noted with BEZ235 than with everolimus, including diarrhea, stomatitis, nausea, and vomiting. These adverse events are usually strongly associated with everolimus, and its increased frequency in BEZ235 further limited its potential for future developments. Wolin et al. [18] recorded more grade 3 or 4 severe adverse events in the pasireotide group compared to the octreotide group, with the most common events being hyperglycemia, diabetes mellitus, diarrhea, liver function derangements, and abdominal pain. Quality of Life The reported results regarding quality of life are listed in table 6. Discussion The treatment of well-differentiated (grade 1/2) NET has changed significantly in recent years, with newer trials supporting the use of targeted therapeutic agents that have demonstrated clear improvements in PFS based on robust randomized clinical trials. While multiple chemotherapy regimens were initially used and formed the basis of the treatment paradigm for a number of years, these were found to have limited benefits, and the trials were susceptible to bias. No single chemotherapy combination has been demonstrated to be superior to another. In addition, no chemotherapy protocol has been tested against placebo or best supportive care. The strength of this review lies in its broad investigation of the role of targeted therapies for NET and rigorous evaluation of the trial evidence according to study quality. Considered overall, the addition of targeted agents improved PFS with a clinically significant HR of 0.54 (p < 0.00001). This supports the ongoing investigation of such agents in these trials, and the adoption of agents such as SSA and everolimus/sunitinib in routine clinical practice. Significant heterogeneity was found, which was somewhat expected given the pooling of results from studies of differing agents with quite distinct modes of action. Benefits were observed in the SSA and mtor/vegf inhibitor subgroups but not in the IFN subgroup. The subgroup of trials investigating the addition of targeted agents to an everolimus backbone also did not show a PFS benefit, which could be attributable to either the efficacy of everolimus as a single agent (thus diluting any clinical benefit from the addition of a second agent) or the alternatively decreased efficacy of targeted agents such as pasireotide and bevacizumab when coadministered with everolimus. Sensitivity analyses excluding trials with a moderate-to-high risk of bias did not alter the significance of the above results. In contrast, no OS benefit was observed, either overall or in any of the subgroups. This is somewhat surprising given the magnitude of the PFS benefit. The significant number of trials which allowed crossover to the experimental arm after progression may explain this disparity between PFS and OS efficacy, as may the relatively long survival of some NET patients, resulting in increased access to later-line treatments. The toxicity was manageable, but the quality of life was mostly unchanged and worsened in one trial (i.e. Arnold et al. [13] ). Significant heterogeneity was noted in the mtor/vegf inhibitor subgroup, with a significant OS benefit reported by Raymond et al. [12] (HR 0.41) which was not replicated in the everolimus trials. Interestingly, Pavel et al. [9], Yao et al. [10], Yao [11], and Kulke et al. [16] (CALGB 80701) demonstrated significant PFS benefits with everolimus but no OS benefit. Early trial termination may have resulted in overestimation of the true benefit of sunitinib in the study of Raymond et al. [12], and significant crossover in the RADI- ANT-2 and RADIANT-3 trials (58% of the placebo arm 220 Lee/Chan/Wong/Li/Lumba/Clarke/ Samra/Pavlakis

in the study of Pavel et al. [9] and 73% in the study of Yao et al. [10] ) may have diluted any OS benefit. This is supported by the trend towards improved OS in RADI- ANT-4, which did not allow crossover after progression. The role of everolimus in the treatment of NET has been confirmed by the above trials, and even more so recently, with confirmation of its efficacy in bronchial and gastrointestinal carcinoids in addition to pnet. The differential OS outcomes in the interventional arms of RADIANT-2 (16.4 months on everolimus and octreotide) and RADIANT-3 (11 months on everolimus alone) are intriguing, especially given the significant benefit of mtor inhibition as monotherapy. However, it would be difficult to attribute the differential OS to the addition of octreotide given the investigation of different primary sites (midgut NET in RADIANT-2 and pnet in RADIANT-3) and the known prognostic implications of these sites. The role of bevacizumab also deserves further investigation. The 2 trials of Yao et al. [19] would indicate some activity against NET, although the comparator employed, i.e. IFN-α, is not widely used currently because of its significant toxicity profile. This was further supported by the data from Kulke et al. [16] where the combination of everolimus and bevacizumab led to superior PFS compared to everolimus alone. Bevacizumab is certainly associated with increased adverse events, particularly hypertension and proteinuria, but these can be managed easily with either temporary cessation of therapy or introduction of antihypertensives. Further trials, particularly investigating the combination of bevacizumab with other targeted therapies, are warranted. To our knowledge, this is the first review to summarize all currently published RCT investigating targeted agents in NET. A rigorous literature search was performed with an evaluation of trial quality according to Cochrane criteria. The key strength of this review is its accurate assessment of the risk of bias and the quality of evidence. In addition, the demonstration of a PFS benefit overall strongly argues for ongoing research into the best way to sequence these agents in the treatment paradigm for NET. The main limitation of this study is the significant statistical heterogeneity observed in the meta-analysis when pooling studies of differing agents in different clinical settings. Individual high-quality trials (such as PROMID and CLARINET) should be considered as stand-alone pieces of evidence. This is mainly due to the inclusion of mixed GEP NET in multiple trials and the use of different molecular targeted therapies. The different biology and prognosis of pnet versus other GEP NET have already been established. Inclusion of all GEP NET can certainly confound the results in a systematic review of trial level data. The use of individual patient data metaanalyses would decrease the risk of bias and provide greater statistical certainty regarding the benefit of specific targeted agents, and it would allow further subgroup analyses. There are a number of unanswered questions regarding the use of targeted therapies in metastatic NET. Firstly, the best targeted therapy has not been demonstrated, since there are no head-to-head trials comparing different SSA, sunitinib, and everolimus. Secondly, whether targeted therapy can replace chemotherapy, particularly in high-grade NET, is not known, again due to the lack of randomized data comparing 2 groups of therapy. Thirdly, the optimal timing and sequence for starting molecular targeted therapy are unknown. PROMID and CLARI- NET suggest a very long PFS with the use of SSA (14.3 months and not reached) compared to placebo used as the initial therapy. RADIANT-2 indicates a 23% improvement in PFS with the combination of everolimus and octreotide, but the PFS in the control arm is still fairly long at 11 months. Of the patients on placebo and octreotide, 58% crossed over on progression. After adjusting for prognostic factors, there were no difference between the experimental and placebo groups. This suggests that early versus delayed use of everolimus may not alter long-term outcomes in the setting of early use of SSA. However, this will need to be assessed in an RCT. Fourthly, the sequence of surgery of the primary and debulking in the setting of molecular targeted therapy is unknown. Removal of the primary and debulking of the tumor burden have been well described in international guidelines largely based on expert consensus and prospective nonrandomized studies. In an era in which somatatostatin analogs have been proven to be largely effective in symptom control and progression delay with tolerable side effects, the role of surgery, particularly in widespread metastatic disease, has now become less certain. We anticipate that future trials will look at determining the best strategy for sequencing an SSA agent, everolimus, sunitinib, and possibly even bevacizumab in the context of PRRT and liver-directed therapy. The use of PRRT instead of/together with targeted therapies will be the subject of ongoing investigations given the impressive results of NETTER-1 reported recently [27]. Alternating the administration of everolimus and sunitinib, as has been done in some metastatic renal cell carcinoma trials, may be a possible strategy to derive benefits from both Targeted Therapy in NET 221

agents while minimizing the impact of their adverse events. Traditional chemotherapy does not appear to have the same benefit and is generally less tolerable than newer biologics, and hence their role in the treatment paradigm remains uncertain. Acknowledgements We thank Drs. S. Faiss, B. Wiedenmann, and J. Yao for provision of information regarding the conduction of the studies of Faiss et al. [14] and Yao et al. [19], respectively. References 1 Mocellin S, Nitti D: Gastrointestinal carcinoid: epidemiological and survival evidence from a large population-based study (n = 25,531). Ann Oncol 2013; 24: 3040 3044. 2 Lepage C, Bouvier AM, Faivre J: Endocrine tumours: epidemiology of malignant digestive neuroendocrine tumours. Eur J Endocrinol 2013; 168:R77 R83. 3 Yao JC, 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 3072. 4 Exocrine and endocrine pancreas; in Edge SB, Compton CC, et al (eds): AJCC Cancer Staging Manual. New York, Springer, 2010, pp 241 249. 5 Jamali M, Chetty R: Predicting prognosis in gastroentero-pancreatic neuroendocrine tumors: an overview and the value of Ki-67 immunostaining. Endocr Pathol 2008; 19: 282 288. 6 Bodei L, et al: Yttrium-labelled peptides for therapy of NET. Eur J Nucl Med Mol Imaging 2012; 39(suppl 1):S93 S102. 7 Rinke A, et al: Placebo-controlled, doubleblind, 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 2009; 27: 4656 4663. 8 Caplin ME, et al: Lanreotide in metastatic enteropancreatic neuroendocrine tumors. N Engl J Med 2014; 371: 224 233. 9 Pavel ME, et al: Everolimus plus octreotide long-acting repeatable for the treatment of advanced neuroendocrine tumours associated with carcinoid syndrome (RADIANT-2): a randomised, placebo-controlled, phase 3 study. Lancet 2011; 378: 2005 2012. 10 Yao JC, et al: Everolimus for advanced pancreatic neuroendocrine tumors. N Engl J Med 2011; 364: 514 523. 11 Yao J: Everolimus plus best supportive care vs placebo plus best supportive care in the treatment of patients with advanced neuroendocrine tumors (GI or lung origin) (RADI- ANT-4). Eur Soc Med Oncol/Eur Cancer Congress, Vienna, 2015. 12 Raymond E, et al: Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. N Engl J Med 2011; 364: 501 513, erratum in N Engl J Med 2011; 364: 1082. 13 Arnold R, et al: Octreotide versus octreotide plus interferon-alpha in endocrine gastroenteropancreatic tumors: a randomized trial. Clin Gastroenterol Hepatol 2005; 3: 761 771. 14 Faiss S, et al: Prospective, randomized, multicenter trial on the antiproliferative effect of lanreotide, interferon alfa, and their combination for therapy of metastatic neuroendocrine gastroenteropancreatic tumors the International Lanreotide and Interferon Alfa Study Group. J Clin Oncol 2003; 21: 2689 2696. 15 Kolby L, et al: Randomized clinical trial of the effect of interferon alpha on survival in patients with disseminated midgut carcinoid tumours. Br J Surg 2003; 90: 687 693. 16 Kulke M, et al: A randomized open-label phase II study of everolimus alone or in combination with pasireotide LAR in advanced, progressive pancreatic neuroendocrine tumors (pnet): COOPERATE-2 trial. 12th Ann Eur Neuroendocr Tumor Soc Conf Diagn Treat Neuroendocr Tumor Dis, Barcelona, 2015. 17 Kulke M, et al: Randomized phase II study of everolimus (E) versus everolimus plus bevacizumab (E+B) in patients (Pts) with locally advanced or metastatic pancreatic neuroendocrine tumors (pnet), CALGB 80701 (Alliance). Annu Am Soc Clin Oncol Meet, Chicago, 2015. 18 Wolin EM, et al: Phase III study of pasireotide long-acting release in patients with metastatic neuroendocrine tumors and carcinoid symptoms refractory to available somatostatin analogues. Drug Des Dev Ther 2015; 9: 5075 5086. 19 Yao JC, 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 alpha-2b. J Clin Oncol 2008; 26: 1316 1323. 20 Yao JC, Guthrie K, Moran C, Strosberg JR, Kulke MH, Chan JA: SWOG S0518: phase III prospective randomized comparison of depot octreotide plus interferon alpha-2b versus depot octreotide plus bevacizumab (NSC #704865) in advanced, poor prognosis carcinoid patients (NCT00569127). J Clin Oncol 2015; 33; 4004. 21 Salazar R, et al: Phase II studies of BEZ235 in patients with advanced pancreatic neuroendocrine tumors (pnet). J Clin Oncol 2015; 33: 4102. 22 Bajetta E, et al: Lanreotide autogel every 6 weeks compared with lanreotide microparticles every 3 weeks in patients with well differentiated neuroendocrine tumors: a phase III study. Cancer 2006; 107: 2474 2481. 23 Rubin J, et al: Octreotide acetate long-acting formulation versus open-label subcutaneous octreotide acetate in malignant carcinoid syndrome. J Clin Oncol 1999; 17: 600 606. 24 Bajetta E, et al: Everolimus in combination with octreotide LAR as the first-line treatment for advanced neuroendocrine tumors: a phase II trial of the I.T.M.O. (Italian Trials in Medical Oncology) group. J Clin Oncol 2013; 31: 4136. 25 Yao J, et al: A randomized phase II study of everolimus for advanced pancreatic neuroendocrine tumors in Chinese patients. Med Oncol 2014; 31: 251. 26 Phan AT, et al: Pazopanib and depot octreotide in advanced, well-differentiated neuroendocrine tumours: a multicentre, singlegroup, phase 2 study. Lancet Oncol 2015; 16: 695 703. 27 Ruszniewski P, et al: 177 Lu-DOTATATE significantly improves progression-free survival in patients with mid-gut neuroendocrine tumours: results of the phase III NETTER-1 trial. Eur Cancer Congress, Vienna, 2015. 222 Lee/Chan/Wong/Li/Lumba/Clarke/ Samra/Pavlakis