Streptozocin chemotherapy for advanced/metastatic well-differentiated neuroendocrine tumors: an analysis of a multi-center survey in Japan

Similar documents
TRACTAMENT ONCOLÒGIC DELS TUMORS NEUROENDOCRINS METASTÀSICS

PRINCESS MARGARET CANCER CENTRE CLINICAL PRACTICE GUIDELINES

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

Effectiveness of Chemotherapy for Advanced Adenocarcinoma of the Pancreas in Combined Modality Therapy

MEDICAL MANAGEMENT OF METASTATIC GEP-NET

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

NET und NEC. Endoscopic and oncologic therapy

Gastrinoma: Medical Management. Haley Gallup

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

Population-Based Study of Islet Cell Carcinoma

Factors Affecting Survival in Neuroendocrine Tumors: A 15-Year Single Center Experience

Cutting Edge Treatment of Neuroendocrine Tumors

Cutting Edge Treatment of Neuroendocrine Tumors

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

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

Index. Surg Oncol Clin N Am 15 (2006) Note: Page numbers of article titles are in boldface type.

Irinotecan (CPT-11) in Patients with Advanced Colon Carcinoma Relapsing after 5-Fluorouracil-Leucovorin Combination

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

Cancer Cell Research 14 (2017)

Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo, Japan

The clinical trial information provided in this public disclosure synopsis is supplied for informational purposes only.

Neuroendocrine Tumors

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

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

Evaluation of efficacy and toxicity of systemic chemotherapy of combined epirubicin, cisplatin and bolus 5- fluorouracil for hepatobiliary tumors

Oberndofer 1907 Illeal Serotonin Secreting Tumor Carcinoid (Karzinoide)

Supplementary Online Content

Cisplatin plus Gemcitabine versus Gemcitabine for Biliary Tract Cancer. Valle J et al. N Engl J Med 2010;362(14):

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

Chromogranin A as a Marker for Diagnosis, Treatment, and Survival in Patients With Gastroenteropancreatic Neuroendocrine Neoplasm

Effects of S-1 as a second-line chemotherapy for patients with relapsed pancreatic cancer

Combination chemotherapy with cisplatin and irinotecan in patients with adenocarcinoma of the small intestine

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

NEUROENDOCRINE CARCINOID TUMORS PANCREATIC NEUROENDOCRINE TUMORS

Embolotherapy for Cholangiocarcinoma: 2016 Update

Recent developments of oncology in neuroendocrine tumors (NETs)

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

Everolimus for Advanced Pancreatic Neuroendocrine Tumors

Clinical Trials for Liver and Pancreatic Cancer in Taiwan

Surgical Therapy of GEP-NET: An Overview

Disclosure of Relevant Financial Relationships

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

Imaging of Neuroendocrine Metastases

SIRT in the Management of Metastatic Neuroendocrine Tumors

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

A Phase II Trial of Uracil Tegafur (UFT) in Patients with Advanced Biliary Tract Carcinoma

Clinical Study Small Bowel Tumors: Clinical Presentation, Prognosis, and Outcomein33PatientsinaTertiaryCareCenter

Pancreas Quizzes c. Both A and B a. Directly into the blood stream (not using ducts)

Phase II trial of capecitabine plus nab-paclitaxel in patients with metastatic pancreatic adenocarcinoma

Horizon Scanning in Oncology

Review of Gastrointestinal Carcinoid Tumors: Latest Therapies

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

Protocol. This trial protocol has been provided by the authors to give readers additional information about their work.

General Information, efficacy and safety data

Fluorouracil/Leucovorin/Interferon α-2a in patients with advanced colorectal cancer. Effects of maintenance therapy on remission duration.

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

Color Codes Pathology and Genetics Medicine and Clinical Pathology Surgery Imaging

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

Systemic Therapy for Advanced Pancreatic Neuroendocrine Tumors: An Update

Low-dose capecitabine (Xeloda) for treatment for gastrointestinal cancer

Surgical Metabolism Section, Surgery Branch, NCI, Bethesda, MD Division of Surgical Oncology, University of Maryland, Baltimore, MD

Endocrine pancreatic tumors: factors correlated with survival

ONCOLOGY LETTERS 2: , 2011

Index. Note: Page numbers of article titles are in boldface type.

Update on the Management of Neuroendocrine Hepatic Metastases

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

Sponsor / Company: Sanofi Drug substance(s): Docetaxel (Taxotere )

CRITICAL ANALYSIS OF NEN GUIDELINES. G Pentheroudakis Associate Professsor of Oncology Medical School, University of Ioannina Chair, ESMO Guidelines

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

EBM-based Clinical Guidelines for Pancreatic Cancer (2013) Issued by the Japan Pancreas Society: A Synopsis

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

Clinical Study Synopsis

The impact of extrahepatic disease among patients undergoing liver-directed therapy for neuroendocrine liver metastasis

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

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

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

The clinical trial information provided in this public disclosure synopsis is supplied for informational purposes only.

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

The Role of the Selective Arterial Secretagogue Injection Test for Non- Functional Pancreatic Neuroendocrine Tumor

A Proposed Strategy for Treatment of Superficial Carcinoma. in the Thoracic Esophagus Based on an Analysis. of Lymph Node Metastasis

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

Twenty-six Cases of Advanced Ampullary Adenocarcinoma Treated with Systemic Chemotherapy

Characteristics of intramural metastasis in gastric cancer. Tatsuya Hashimoto Kuniyoshi Arai Yuichi Yamashita Yoshiaki Iwasaki Tsunekazu

Diagnosis abnormal morphology and /or abnormal biochemistry

Low Dose Docetaxel Combined With Low Dose Capecitabine in Treatment of Metastatic Breast Cancer Previously Treated With Anthracycline

Type 2 gastric neuroendocrine tumor: report of one case

Chemotherapy for Advanced Gastric Cancer

Targeted Therapies in Metastatic Colorectal Cancer: An Update

FOLFOX-4 as second-line therapy after failure of gemcitabine and platinum combination in advanced gall bladder cancer patients

NET ΠΝΕΥΜΟΝΑ: τι νεότερο / νέες μελέτες

An Advanced Well-differentiated Pancreatic Neuroendocrine Carcinoma (NET-G3) Associated with Von Hippel-Lindau Disease

Treatment outcomes and prognostic factors of gallbladder cancer patients after postoperative radiation therapy

Pancreatic polypeptide secreting tumors an institutional experience and review of the literature

Sponsor / Company: Sanofi Drug substance(s): SAR (iniparib)

Gallbladder Cancer. GI Practice Guideline. Michael Sanatani, MD, FRCPC (Medical Oncologist) Barbara Fisher, MD, FRCPC (Radiation Oncologist)

GEP NEN. Personalised approach. Curative and Palliative Surgery. ESMO Preceptorship Programme Neuroendocrine Neoplasms Lugano April 2018

Risk factors for 6-month continuation of S-1 adjuvant chemotherapy for gastric cancer

Edith A. Perez, Ahmad Awada, Joyce O Shaughnessy, Hope Rugo, Chris Twelves, Seock-Ah Im, Carol Zhao, Ute Hoch, Alison L. Hannah, Javier Cortes

NEWS RELEASE Media Contact: Megan Pace Investor Contact: Kathee Littrell Patient Inquiries: Ajanta Horan

Transcription:

J Gastroenterol (2015) 50:769 775 DOI 10.1007/s00535-014-1006-3 ORIGINAL ARTICLE LIVER, PANCREAS, AND BILIARY TRACT Streptozocin chemotherapy for advanced/metastatic well-differentiated neuroendocrine tumors: an analysis of a multi-center survey in Japan Taku Aoki Norihiro Kokudo Izumi Komoto Kyoichi Takaori Wataru Kimura Keiji Sano Takeshi Takamoto Takuya Hashimoto Takuji Okusaka Chigusa Morizane Tetsuhide Ito Masayuki Imamura Received: 31 May 2014 / Accepted: 5 October 2014 / Published online: 28 October 2014 The Author(s) 2014. This article is published with open access at Springerlink.com Abstract Background Neuroendocrine tumors (NETs) are believed to be relatively rare and to follow a generally indolent course. However, liver metastases are common in NET patients and the outcome of NET liver metastasis is poor. In Western countries, streptozocin (STZ) has been established as a first-line anticancer drug for unresectable NET; however, STZ cannot be used in daily practice in Japan. The aim of the present study was to determine the status of STZ usage in Japan and to evaluate the effectiveness and safety of STZ chemotherapy in Japanese NET patients. Electronic supplementary material The online version of this article (doi:10.1007/s00535-014-1006-3) contains supplementary material, which is available to authorized users. T. Aoki (&) N. Kokudo Hepato-Biliary-Pancreatic Surgery Division, Department of Surgery, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, Japan 113-8655 e-mail: aoki-2su@h.u-tokyo.ac.jp T. Aoki N. Kokudo Artificial Organ and Transplantation Division, Department of Surgery, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, Japan 113-8655 I. Komoto M. Imamura Department of Surgery, Kansai Electric Power Company Hospital, Osaka, Japan K. Takaori Department of Hepatobiliary-Pancreatic Surgery and Transplantation, Graduate School of Medicine, Kyoto University, Kyoto, Japan Methods A retrospective multi-center survey was conducted. Five institutions with experience performing STZ chemotherapy participated in the study. The patient demographics, tumor characteristics, context of STZ chemotherapy, and patient outcome were collected and assessed. Results Fifty-four patients were enrolled. The main recipients of STZ chemotherapy were middle-aged patients with pancreatic NET and unresectable liver metastases. The predominant regimen was the weekly/bi-weekly intravenous administration of STZ combined with other oral anticancer agents. STZ monotherapy was used in onefourth of the patients. The median progression-free and overall survival periods were 11.8 and 38.7 months, respectively, and sustained stable disease was obtained in K. Sano Department of Surgery, Teikyo University School of Medicine, Tokyo, Japan T. Takamoto T. Hashimoto Division of Hepato-Biliary-Pancreatic Surgery, Japanese Red Cross Medical Center, Tokyo, Japan T. Okusaka C. Morizane Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo, Japan T. Ito Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan W. Kimura Department of Gastroenterological, Breast, Thyroid, and General Surgery, Yamagata University Faculty of Medicine, Yamagata, Japan

770 J Gastroenterol (2015) 50:769 775 some selected patients. The adverse events profile was mild and tolerable. Conclusions Our survey showed the clinical benefit and safety of STZ therapy for Japanese patients with unresectable NET. Therefore, we recommend that STZ, which is the only cytotoxic agent available against NET, should be used in daily practice in Japan. Keywords Neuroendocrine tumors Streptozocin Multi-center survey Tumor response Progression-free survival rate Introduction Neuroendocrine tumors (NETs) have been regarded as relatively rare neoplasms, but the number of patients with NET is increasing in the US [1], Europe [2], and Japan [3, 4]. The epidemiological pattern of NET is highly heterogeneous; for example, the tumor location, biological behavior (functioning NET or non-functioning NET), and percentage of distant metastases differ extensively among databases. Therefore, the clinical outcomes of the various treatment modalities also differ according to the characteristics of the study cohort. The clinical course of well-differentiated NET (NET G1 or NET G2) is believed to be generally indolent, but some previous studies have documented that 40 95 % of NET patients are metastatic at presentation, and the 5-year survival rate is 56 83 % for metastatic intestinal NETs and 40 60 % for metastatic pancreatic NETs. Thus, optimal management of metastatic lesions, especially of liver metastases, is key to improving the outcomes of NET patients [5]. Streptozocin (STZ) was first discovered as an antibiotic derived from Streptomyces achromogenes, and was approved in the US as a cytotoxic antitumor drug for symptomatic or advanced pancreatic NET in 1982. In Western countries, STZ combined with doxorubicin (DOX) or fluorouracil (5-FU) has been established as a first-line chemotherapy for both pancreatic and gastrointestinal NETs based on several clinical trails including randomized clinical trials [6 12]. However, STZ has not been covered by the Japanese insurance system, and Japanese oncologists/gastroenterologists cannot choose this powerful option for the treatment of advanced or metastatic NETs. Because of these specific circumstances, STZ chemotherapy has only been used in clinical trials in Japan. Therefore, the aim of the study was to investigate the actual situations in which STZ is used in Japan and to evaluate the effectiveness and safety of STZ chemotherapy among Japanese NET patients. Methods This study was conducted as a retrospective multi-center survey. Five institutions (The University of Tokyo Hospital, Tokyo; Osaka Saiseikai Noe Hospital, Osaka; Kyoto University Hospital, Kyoto; Japanese Red Cross Medical Center, Tokyo; and Yamagata University Hospital, Yamagata) with experience performing STZ chemotherapy participated in the present study. Patients who were treated with STZ between September 1995 and November 2011 were included as the study subjects. The following clinicopathological factors were investigated: (1) sex, age at the start of STZ chemotherapy, date at the start of STZ chemotherapy, and performance status at the start of STZ chemotherapy; (2) clinical diagnosis, site of the primary tumor, age of tumor presentation, behavior of the tumor (functioning or non-functioning), presence or absence of metastasis, and metastatic site(s); (3) STZ treatment regimen, period of treatment, total dose of administered STZ, antitumor drugs used in combination with STZ; and (4) efficacy of STZ chemotherapy, adverse events, progression-free survival period, and overall survival period. The tumor response to STZ therapy was evaluated using RECIST criteria [13], and adverse events were assessed according to the National Cancer Institute Common Terminology Criteria for Adverse Events (CTCAE version 4.0). The survival curves were generated using Kaplan Meier methods [14], and the differences among the curves were evaluated using a log-rank test [15]. Differences were considered significant when P \ 0.05. The protocol was approved by the local ethical committee of each institution that participated in the study. The clinical data were summarized in a blinded manner. Results Patients The data of 54 patients were collected. The patient cohort consisted of 24 male and 30 female patients, and the median age was 54.0 years (range 24 76 years) at the onset of the disease and 56.0 years (range 31 77 years) at the start of STZ administration (Table 1). Regarding the distribution of age at the onset of the disease, a peak occurred at between 50 and 59 years, followed by a second peak at 60 69 years (as shown in the Electronic supplementary material). The performance status of most of the patients was 0 or 1 (Table 1). Tumor characteristics The characteristics of the tumors are summarized in Table 1. Forty-two patients had pancreatic NET (P-NET),

J Gastroenterol (2015) 50:769 775 771 Table 1 Patient demographics and tumor characteristics Parameters No. of patients Percent (%) Sex Male 24 44.4 Female 30 55.6 Age at onset Mean 52.5 Median 54.0 Range 24 76 Age at the beginning of STZ administration Mean 56.0 Median 56.0 Range 31 77 Performance status 0 34 63.0 1 17 31.5 2 3 5.5 3 4 0 0.0 Primary site Pancreaticoduodenal NET (46) (85.2) Pancreas head 12 26.1 Pancreas body 10 21.7 Pancreas tail 19 41.3 Head, body and tail 1 2.2 Duodenal 4 8.7 Gastrointestinal NET (8) (14.8) Stomach 2 25.0 Small Intestine 1 12.5 Rectum 4 50.0 Others 1 12.5 Pathological diagnosis (WHO 2000) Well-differentiated endocrine tumor 0 0.0 Well-differentiated endocrine carcinoma 52 96.4 Poorly-differentiated endocrine carcinoma/ 1 1.8 small cell carcinoma Others 1 1.8 Functioning NET/non-functioning NET Functioning (18) (33.3) Gastrinoma 9 16.7 Insulinoma 7 13.0 Glucagonoma 4 7.4 Somatostatinoma 1 1.9 Serotonin, tachykinins producing tumor 1 1.9 Non-functioning (36) (66.7) Metastatic site(s) Liver 53 98.1 Lymph nodes 26 48.1 Peritoneum 3 5.6 Lung 2 3.7 Others 10 18.5 and the duodenum and gastrointestinal tract were the original sites in 4 and 8 patients, respectively. The pathological diagnosis based on the WHO Classification 2000 was well-differentiated endocrine carcinoma in 52 patients (96.4 %). One-third of the tumors (n = 18) were functioning, with 9 gastrinomas and 7 insulinomas; the other two-thirds were non-functioning NETs. All the patients had metastatic sites: all but one patient had liver metastasis, with lymph node metastasis being the second most common site (n = 26, 48.1 %). STZ therapy STZ chemotherapy was used as a first-line therapy in 39 patients, as a second-line therapy in 11 patients, and as a third-line therapy in 4 patients. The treatments used prior to STZ chemotherapy included transcatheter arterial chemoembolization (TACE), octreotide, 5FU, and gemcitabine. STZ was administered intravenously in 35 patients (64.8 %) and intra-arterially in 3 patients. Both routes were used in 15 patients. The dosing regimen was daily [350 500 mg/m 2 of STZ administered for 5 consecutive days (days 1 5) every 6 weeks] in 14 patients and weekly or bi-weekly in 31 patients (350 1,000 mg/m 2 of STZ administered at each treatment). Both regimens were used in 3 patients. Interestingly, the participating institutions in Eastern Japan applied a weekly or bi-weekly regimen, while the institutions in Western Japan applied a daily regimen. Thirteen patients received STZ monotherapy, while a combination therapy was used in the other 41 patients. The combined antitumor agents included tegafur-uracil (UFT, n = 26), octreotide (n = 20), fluorouracil (5-FU, n = 15), and oral fluoropyrimidine (S-1, n = 6) (Table 2). The dosing period ranged from 0 to 105 months, with a median of 12.4 months. The dosing period was within 20 months in most patients (Fig. 1a). The total amount of STZ administered ranged from 1.0 to 128.0 g (median 18.8 g) (Fig. 1b). The tumor response as evaluated according to the RE- CIST criteria is shown in Table 3. The tumor response was CR in 2 patients, PR in 11 patients, SD in 9 patients, PD in 25 patients, and unknown in 7 patients, with a response rate of 27.7 %. The response to STZ monotherapy was CR in 1 patient, PR in 4 patients, SD in 1 patient, PD in 8 patients, and unknown in 4 patients, with a response rate of 35.7 %. Documented adverse events included nausea (n = 12, 22.2 %), vomiting (n = 7, 13.0 %), and lethargy (n = 4, 7.4 %). Other adverse hematological, hepatobiliary, or nervous system events were observed in a few patients. Grade 3 adverse events were observed in 6 patients (3 nausea and 3 vomiting), but no grade 4 adverse events were documented (Table 4).New-onset diabetes mellitus was not

772 J Gastroenterol (2015) 50:769 775 Table 2 STZ therapy Parameters No. of patients Percent (%) Dosing route Intravenous (IV) 35 64.8 Intra-arterial (IA) 3 5.6 IV/IA 15 27.8 Unknown 1 1.9 Dosing regimen Daily 14 25.9 Weekly/bi-weekly 31 57.4 Daily/weekly 3 5.6 Others 6 11.1 Antitumor agents combined with STZ Doxorubicin 1 1.9 Fluorouracil (5-FU) 15 27.8 Oral fluoropyrimidine (S-1) 6 11.1 Tegafur-uracil (UFT) 26 48.1 Octreotide 20 37.0 Mitomycin C 3 5.6 Interferon 1 1.9 Sunitinib 1 1.9 None (STZ monotherapy) 13 24.1 documented, but the control of the disease was impaired during STZ therapy in one patient who had been treated for diabetes mellitus. STZ therapy was discontinued in 46 patients. The reasons for the discontinuation were tumor progression in 43 patients, conversion to other treatments in 2 patients, and a severe adverse event in 1 patient. Patient outcome Data regarding patient outcome were available for 38 patients. The progression-free and overall survival curves are shown in Figs. 2 and 3. The median progression-free period was 11.8 months in all of the patients (mean 23.0 ± 3.5 months), 7.6 months in the functioning NET patients, and 16.8 months in the non-functioning NET patients (P = 0.14). Meanwhile, the median survival period was 38.7 months in all of the patients (mean 28.7 ± 2.6 months), 23.6 months in the functioning NET patients, and 38.7 months in the non-functioning NET patients (P = 0.32). The median amount of STZ administered was 18.8 g. When the patients were stratified according to the amount of STZ (C18.8 or \18.8 g), both the overall survival rate and the progression-free survival rate were better in the patients who received C18.8 g STZ (see the Electronic supplementary material 2). The overall and progression-free survival outcomes were similar among the patients who received a daily Fig. 1 Distribution of the dosing period (a) and the total amount of STZ administered (b) (n = 54) regimen and those receiving a weekly/bi-weekly regimen (data not shown). In addition, the outcomes did not differ between patients with pancreaticoduodenal NET (n = 46) and those with gastrointestinal NET (n = 8) (see the Electronic supplementary material 3). Discussion The present study was a retrospective multi-center cohort study in patients with unresectable NET receiving STZ chemotherapy. This is the first attempt to determine the circumstances surrounding chemotherapy for NET patients in Japan. The five participating centers were high-volume centers treating NET patients in Japan, and most of the patients who received STZ therapy before 2011 were thought to have been included in the study. During the study period (from 1995 to 2011), octreotide was the only antitumor agent against NET available in Japan until everolimus and sunitinib began to be covered by the Japanese insurance system. STZ is not yet covered by the Japanese insurance system:, so STZ therapy had only been conducted on a clinical trial basis using imported

J Gastroenterol (2015) 50:769 775 773 Table 3 Tumor response, evaluated according to the RECIST criteria Tumor response according to RECIST criteria All cases Pancreaticoduodenal NET Gastrointestinal NET Subtotal STZ monotherapy Combination therapy Subtotal STZ monotherapy Combination therapy n % n % n % n % n % n % n % 54 46 14 32 8 4 4 CR 2 4.3 2 5.3 1 11.1 1 3.4 0 0.0 0 0.0 0 0.0 PR 11 23.9 9 23.7 3 33.3 6 20.7 2 25.0 1 25.0 1 25.0 SD 9 19.6 8 21.1 1 11.1 7 24.1 1 12.5 0 0.0 1 25.0 PD 25 54.3 20 52.6 5 55.6 15 51.7 5 62.5 3 75.0 2 50.0 UK 7 7 4 3 0 0 0 UK unknown Table 4 Adverse events Adverse events n % CTCAE grade G1 G2 G3 G4 Unknown Gastointestinal disorder Abdominal pain 1 1.9 1 Diarrhea 2 3.7 1 1 Epigastric pain 1 1.9 1 Nausea 12 22.2 5 4 3 Acute pancreatitis 1 2.9 1 Vomiting 7 13.0 1 3 3 Hematolymphoid system disorder Leukopenia 1 1.9 1 Neutropenia 2 3.7 1 1 Thrombocytopenia 1 1.9 1 Ocular lesion Abnormal ocular sensation 1 1.9 1 Hepatobiliary system disorder Liver function 1 1.9 1 abnormality Nerve system disorder Syncope 1 1.9 1 Headache 1 1.9 1 Others Lethargy 4 7.4 3 1 Back pain 1 1.9 1 STZ at all of the institutions that participated in the present study. One of the aims of our study was to encourage the approval of STZ use in a daily clinical setting in Japan. The results of the present study revealed that the main recipients of STZ chemotherapy were patients with P-NET (well-differentiated endocrine carcinoma based on the WHO Classification 2000) with liver metastases. The dosing routes and dosing regimens varied among the regions and institutions, but an intravenous weekly/biweekly regimen was popularly applied. The original regimen proposed by Moertel et al. [7] was a combination therapy of STZ with doxorubicin or STZ with fluorouracil (5-FU); however, in the present study, various antitumor agents were combined with STZ, and STZ monotherapy was applied in one-fourth of the patients. The reasons for this were likely twofold: first, the use of oral anticancer drugs, such as S-1 and UFT, is popular in Japan; second, the use of other cytotoxic anticancer drugs has not been approved. Our results showed that the response rate was 27.7 % for all of the enrolled patients, and a subgroup analysis showed that the response rate was 28.2 % for pancreaticoduodenal NET patients and 25.0 % for gastrointestinal NET patients, respectively. In addition, STZ monotherapy was associated with a response rate of 35.7 % (40.0 % for pancreaticoduodenal NET and 25.0 % for gastrointestinal NET). These figures were comparable with those obtained in Western series in which radiological measurements were used to evaluate tumor response [10 12]. The dosing period was less than 10 months in 45 % of the patients, and 10 20 months in 22 % of the patients. As a result, the total amount of STZ adminstered was less than 20 g in over 50 % of the patients (Fig. 1b). These results corresponded to a median progression-free period of 11.8 months (Fig. 2). The figure of 11.8 months was similar to that obtained in studies examining everolimus and sunitinib [16, 17]. However, the progression-free survival curve in STZ therapy patients reached a plateau about 2 years after the start of the therapy (Fig. 2), showing a difference from the everolimus and sunitinib studies. This finding suggested that sustained stable disease can be expected in some selected patients receiving STZ, and that these patients can undergo STZ chemotherapy for a long period because of the mild adverse event profile. Actually, some patients in our study received STZ therapy for over 5 years. As expected, the

774 J Gastroenterol (2015) 50:769 775 Fig. 2 Progression-free survival curves for all of the patients (n = 38) (a) and stratified according to functioning (n = 12) and nonfunctioning (n = 26) tumors (b) Fig. 3 Overall survival curves for all the patients (n = 38) (a) and curves stratified according to functioning (n = 12) and non-functioning (n = 26) tumors (b) outcomes were better among the patients who received a larger dose of STZ (see the Electronic supplementary material 2). These results also support the idea that longterm STZ chemotherapy is associated with long-term SD maintenance. In our analyses, the progressions and overall survivals were comparable between the patients with functioning NET and those with non-functioning NET, suggesting that STZ is applicable to all NET patients with the same dosing regimen. Our survey showed that the adverse events associated with STZ chemotherapy were acceptable. Studies using animal models showed that high-dose STZ administration induced impaired glucose tolerance, leading to diabetes mellitus. In the present survey, new-onset diabetes mellitus induced by STZ was not documented. In addition, STZ therapy was discontinued because of a severe adverse event in only one patient. This mild adverse event profile can likely be attributed to the relatively low-dose regimens performed in our series (350 500 mg/m 2 in the daily regimen, and 350 1,000 mg/m 2 regimen in the weekly/biweekly regimen). In conclusion, our survey showed the clinical benefit and safety of STZ therapy for pancreaticoduodenal and gastrointestinal NET. Therefore, we recommend that STZ, the only cytotoxic agent available for NET, should be used in daily practice in Japan. Acknowledgments This study was conducted by the NET Chemotherapy Investigation and Research Group in cooperation with NobelPharma Co. Ltd. The context of the study was presented at the 49th Annual Meeting of the Japan Society of Clinical Oncology, November 2011, Nagoya. Conflict of interest of interest. The authors declare that they have no conflict Open Access This article is distributed under the terms of the Creative Commons Attribution Noncommercial License which permits any noncommercial use, distribution, and reproduction in any medium, provided the original author(s) and the source are credited. References 1. Yao JC, Hassan M, Phan A, et al. One hundred years after carcinoid : epidemiology of and prognostic factors for neuroendocrine tumors in 35,825 cases in the US. J Clin Oncol. 2008;26:3063 72. 2. Hauso O, Gustafsson BI, Kidd M, et al. Neuroendocrine tumor epidemiology: contrasting Norway and North America. Cancer. 2008;113:2655 64. 3. Ito T, Sasano H, Tanaka M, et al. Epidemiological study of gastroenteropancreatic neuroendocrine tumors in Japan. J Gastroenterol. 2010;45:234 43. 4. Ito T, Igarashi H, Nakamura K, et al. Epidemiological trends of pancreatic and gastrointestinal neuroendocrine tumors in Japan: a nationwide survey analysis. J Gastroenterol. 2014;6.

J Gastroenterol (2015) 50:769 775 775 5. Frilling A, Modlin IM, Kidd M, et al. Recommendations for management of patients with neuroendocrine liver metastases. Lancet Oncol. 2014;15:e8 21. 6. Moertel CG, Hanley JA, Johnson LA. Streptozocin alone compared with streptozocin plus fluorouracil in the treatment of advanced islet-cell carcinoma. N Engl J Med. 1980;303:1189 94. 7. Moertel CG, Lefkopoulo M, Lipsitz S, et al. Streptozocin-doxorubicin, streptozocin-fluorouracil or chlorozotocin in the treatment of advanced islet-cell carcinoma. N Engl J Med. 1992;326:519 23. 8. Rivera E, Ajani JA. Doxorubicin, streptozocin, and 5-fluorouracil chemotherapy for patients with metastatic islet-cell carcinoma. Am J Clin Oncol. 1998;21:36 8. 9. von Schrenck T, Howard JM, Doppman JL, et al. Prospective study of chemotherapy in patients with metstatic gastrinoma. Gastroenterology. 1988;94:1326 34. 10. Eriksson B, Skogseid B, Lundqvist G, et al. Medical treatment and long-term survival in a prospective study of 84 patients with endocrine pancreatic tumors. Cancer. 1990;65:1883 90. 11. Cheng PN, Saitz LB. Failure to confirm major objective antitumor activity for streptozocin and doxorubicin in the treatment of patients with advanced islet cell carcinoma. Cancer. 1999;86: 944 8. 12. Kouvaraki MA, Ajani JA, Hoff P, 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. 13. Eisenhauer EA, Therasse P, Boqaerts J, et al. New response evaluation criteria in solid tumors: revised RECIST guideline (version 1.1). Eur J Cancer. 2009;45:228 47. 14. Kaplan E, Meier P. Non-parametric estimation from incomplete observations. J Am Stat Assoc. 1958;53:257 481. 15. Mantel N. Evaluation of survival data and two new rank order statistics arising in its consideration. Cancer Chemother Rep. 1966;50:163 70. 16. Yao JC, Shah MH, Ito T, et al. Everolimus for advanced pancreatic neuroendocrine tumors. N Engl J Med. 2011;364:514 23. 17. Raymond E, Dahan L, Raoul JL, et al. Sunitinib malate for the treatment of oancreatic neuroendocrine tumors. N Engl J Med. 2011;364:501 13.