Title: High Risk of Myelodysplastic Syndrome (MDS) and Acute Myeloid. in Neuroendocrine Tumor Patients Heavily Pre-treated with Alkylating

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

CLICK TO GO BACK TO KIOSK MENU

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

Advanced Thoracic NET Clinical Cases: PRRT in Lung NEN. Ulrike Garske-Román MD, PhD ESMO Preceptorship Prague 2017

Review of Gastrointestinal Carcinoid Tumors: Latest Therapies

Lutetium-DOTA TATE Treatment of inoperable GEP NETs

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

Cutting Edge Treatment of Neuroendocrine Tumors

Cutting Edge Treatment of Neuroendocrine Tumors

Peptide Receptor Radionuclide Therapy using 177 Lu octreotate

MEDICAL MANAGEMENT OF METASTATIC GEP-NET

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

ENETS Consensus Guidelines for the Standard of Care in

MEDICAL POLICY SUBJECT: PEPTIDE RECEPTOR RADIONUCLIDE THERAPY (PRRT)

Influence of the amount of co-infused amino acids on post-therapeutic potassium levels in peptide receptor radionuclide therapy

Peptide receptor radionuclide therapy (PRRT) is a highly efficient

Gastroenteropancreatic and thoracic neuroendocrine tumors

THERAPEUTIC RADIOPHARMACEUTICALS

Original Article Bone metastases in GEP-NET: response and long-term outcome after PRRT from a follow-up analysis

Renal function affects absorbed dose to the kidneys and haematological toxicity during 177 Lu-DOTATATE treatment

Patient information file

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

Neuroendocrine Tumour Theranostics

TRACTAMENT ONCOLÒGIC DELS TUMORS NEUROENDOCRINS METASTÀSICS

The role of peptide receptor radionuclide therapy in advanced/ metastatic thoracic neuroendocrine tumors

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

NET und NEC. Endoscopic and oncologic therapy

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

MEDICAL POLICY EFFECTIVE DATE: 06/21/07 REVISED DATE: 05/14/08, 04/16/09, 03/18/10, 03/17/11, 03/15/12, 02/21/13, 02/20/14, 02/19/15

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

Recent developments of oncology in neuroendocrine tumors (NETs)

Diabetes mellitus and its effects on all cause mortality after radiopeptide therapy for neuroendocrine tumors

Peptide Receptor Radionuclide Therapy (PRRT) of NET

Session 6 NEW TECHNIQUES IN RADIATION TREATMENT. Chairman : Françoise MORNEX

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

MEDICAL POLICY SUBJECT: PEPTIDE RECEPTOR RADIONUCLIDE THERAPY (PRRT)

Targeted radionuclide therapy for neuroendocrine tumours

Targeted Radionuclide Therapy with 90 Y-DOTATOC in Patients with Neuroendocrine Tumors

Prior Authorization Review Panel MCO Policy Submission

Nuclear oncology using SPECT and PET is able to show

PRINCESS MARGARET CANCER CENTRE CLINICAL PRACTICE GUIDELINES

Treatment algorithm Neuroendocrine tumours. Gregory Kaltsas Endocrine Unit, Department of Pathophysiology, University of Athens, Greece

Therapeutic Radiopharmaceuticals in Oncology

Amir Sabet 1, James Nagarajah 2, Ahmet Semih Dogan 1, Hans-Jürgen Biersack 1, Amin Sabet 1, Stefan Guhlke 1 and Samer Ezziddin 1*

original article introduction original article

THERANOSTICS MOLEKULARE BILDGEBUNG MITTELS PET/CT

Theragnostics Neuroendocrine and Prostate Cancer

Addison's disease Neuroendocrine tumors Paraneoplastic syndromes

Neuroendocrine Tumor Therapy:

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

NOVEL APPROACHES IN THE CLASSIFICATION AND RISK ASSESSMENT OF PATIENTS WITH MYELODYSPLASTIC SYNDROMES-CLINICAL IMPLICATION

PRRT in Management of NETs. Ioannis Karfis, MD PhD Assistant Head of Clinic Nuclear Medicine Dept IJB, Brussels

What is MDS? Epidemiology, Diagnosis, Classification & Risk Stratification

THERANOSTICS clinical aimshots in surgical warfare against well-differentiated neuroendocrine neoplasms

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

Prolonged Survival in a Patient with Neuroendocrine Tumor of the Cecum and Diffuse Peritoneal Carcinomatosis

Patient information and consent form concerning: Radiotherapy with lutetium-octreotate as treatment for patients with neuroendocrine tumors

OPTIMISING OUTCOMES IN GASTROINTESTINAL NEUROENDOCRINE TUMOURS

Martin H. Cherk 1,3,4, Grace Kong 1, Rodney J. Hicks 1,2 and Michael S. Hofman 1,2*

Dosimetry and radiobiology for Peptide Receptor Radionuclide Therapy

PRESS RELEASE. Advanced Accelerator Applications Receives US FDA Approval for LUTATHERA for Treatment of Gastroenteropancreatic Neuroendocrine Tumors

Color Codes Pathology and Genetics Medicine and Clinical Pathology Surgery Imaging

Pancreatic Neuroendocrine Tumours

Teresa Alonso Gordoa Servicio Oncología Médica Hospital Universitario Ramón y Cajal

Somatostatin-based radiotherapy with [ 90 Y-DOTA]-TOC in neuroendocrine tumors: long-term outcome of a phase I dose escalation study. Marincek et al.

NET ΠΝΕΥΜΟΝΑ: τι νεότερο / νέες μελέτες

Lu 177-Dotatate (Lutathera) Therapy Information

Amir Sabet, Feras Khalaf, Soha Mahjoob, Abdullah Al-Zreiqat, Hans-Jürgen Biersack, Samer Ezziddin

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

SPECIAL AUTHORIZATION REQUEST FOR COVERAGE OF HIGH COST CANCER DRUGS

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

SUPPLEMENTARY INFORMATION

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

Somatostatin-based radiopeptide therapy with [ 177 Lu-DOTA]-TOC versus [ 90 Y-DOTA]-TOC in neuroendocrine tumours

Recent Advances in Gastrointestinal Cancers

Digestive and Liver Disease

SIRT in the Management of Metastatic Neuroendocrine Tumors

Nephrotoxicity after PRRT with 177 Lu-DOTA-octreotate

A prospective, multicenter European Registry for newly diagnosed patients with Myelodysplastic Syndromes of IPSS low and intermediate-1 subtypes.

Disclosure of Relevant Financial Relationships

Prognostic factors and treatment of gastroenteropancreatic G3 neuroendocrine carcinomas.

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

Nuevas alternativas en el manejo de TNE avanzados

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

ENETS Consensus Guidelines. Neuroendocrinology 2016;103: DOI: /

Treatment failure in cancer is often attributed to insufficient

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

Peking University School of Oncology, Beijing Cancer Hospital and Institute, Beijing, China

The PET-NET Study 2016 CNETS Grant Award

Rodney J Hicks, MD, FRACP, FAAHMS, the Peter MaCallum Cancer Centre, Melbourne, Australia

PRRT genomic signature in blood for prediction of 177 Lu-octreotate efficacy

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

Prediction of clinically relevant hyperkalemia in patients treated with peptide receptor radionuclide therapy

Case Report Metastatic Insulinoma Managed with Radiolabeled Somatostatin Analog

Commissioning policies agreed by PCTs in Yorkshire and the Humber at Board meeting of YH SCG on December

Small-cell lung cancer (SCLC) accounts for 15% to 18% of

MDS - Diagnosis and Treatments. Dr Helen Enright, Adelaide and Meath Hospital Dr Catherine Flynn, St James Hospital

The College of Nuclear Physicians of South Africa Practice Guidelines on Peptide Receptor Radionuclide Therapy in Neuroendocrine Tumours

TUMORES NEUROENDOCRINOS. Miguel Navarro. Salamanca

A Phase II Study of the Combination of Oral Rigosertib and Azacitidine in Patients with Myelodysplastic Syndromes (MDS)

Transcription:

Page 1 of 12 Accepted Preprint first posted on 1 March 2016 as Manuscript ERC-15-0543 1 2 3 4 Title: High Risk of Myelodysplastic Syndrome (MDS) and Acute Myeloid Leukemia (AML) after 177 Lu-octreotate Peptide Receptor Radionuclide Therapy in Neuroendocrine Tumor Patients Heavily Pre-treated with Alkylating Chemotherapy 5 6 7 Running title: Myelodysplastic syndrome and acute myeloid leukemia after chemotherapy and PRRT 8 9 Research Letter: 10 Dear Editor, 11 12 13 14 Gastro-entero-pancreatic neuroendocrine tumors (GEP-NETs) are rare but seen with increasing incidence. Current medical options for the management of nonresectable GEP-NETs include somatostatin analogues, targeted therapies, chemotherapy, radiological and radionuclide therapies 15 16 17 18 19 20 21 22 23 Peptide receptor radionuclide therapy (PRRT) is a modern therapeutic approach using radionuclide combined with somatostatin analogue peptide whose affinity with somatostatin receptors (SSR) allows targeting disseminated tumor disease. According to the ENETS guidelines, PRRT is indicated for patients with nonresectable, progressive, grade 1 or 2 GEP-NETs with high uptake on somatostatin receptor scintigraphy (Pavel, et al. 2012). In a large retrospective study of 310 GEP- NETs, 46% had tumor response with 177 Lu-octreotate therapy (Kwekkeboom, et al. 2008) with good progression-free and overall survival. In addition, first results of the prospective randomized NETTER-1 trial comparing 177 Lu-octreotate PRRT and 1 Copyright 2016 by the Society for Endocrinology.

Page 2 of 12 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 octreotate LAR 60 mg recently, showed a median PFS not reached at 25 months and 8.4 months, respectively (Strosberg, et al. 2015). PRRT is generally well tolerated, short-term side effects including mild fatigue, hematological and renal toxicities. Regarding longer term hematological side effects, MDS or AML were reported in 0.2 to 5.4% of the patients in large series (Bodei et al. 2015; Imhof, et al. 2011; Kesavan, et al. 2014; Kwekkeboom et al. 2008; Sabet, et al. 2013) (Bodei, et al. 2015). Here, we report a much higher occurrence of MDS or AML in a single center experience in patients treated with 177 Lutetium-octreotate at late disease stage, after alkylatingbased chemotherapy. Our study included all 20 consecutive patients treated with 177 Lutetium octreotate PRRT between January 2004 and January 2011 at our center. All patients had progressive metastatic GEP-NETs. PRRT was performed in the Nuclear Medicine department of the Erasmus Medical Center of Rotterdam (Netherlands) due to the unavailability of PRRT in France. This unavailability explained why relatively few patients received this treatment in our center, and generally after first line chemotherapy. As defined in previous reports from Rotterdam s team (Kwekkeboom, et al. 2001), the radiochemical purity of 177 Luoctreotate was 88%, and the reached yield after addition of DTPA (diethylene triamine pentaacetic acid) approached 99.99%. Patient medical records were reviewed to collect relevant data on demographics, tumor characteristics, surgery, treatments.and tolerability. A Student s test was used to evaluate prognostic variables for the occurrence of t-mds/aml. 46 47 48 Baseline clinical characteristics of the 20 patients are described in Table 1. Median follow-up from PRRT was 3.1 years (range 0.3-8.9). Treatment with 177 Lu- PRRT consisted of four cycles of 7.5 GBq; 16 patients received the four full dose 2

Page 3 of 12 49 50 cycles one of them received two additional cycles - and four received lower dose due to early hematological toxicity - one of them one cycle -. 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65 PPRT induced short-term thrombocytopenia, neutropenia and anemia in 25%, 15% and 10%, respectively. However, 4 (20%) patients developed delayed hematological toxicity with MDS or AML, diagnosed 30, 31, 54 and 70 months, respectively, after PRRT. All patients, before PRRT, had received prior chemotherapy containing an alkylating agent, for 20, 12, 6 and 12 cycles, respectively. Three of the four patients (75%) had developed early hematological toxicity (grade 3 to 4 thrombocytopenia) during PRRT. At the time of MDS/AML diagnosis, the underlying tumor was active in 3 patients, and 3 had bone metastases. Three patients had MDS according to WHO criteria (Della Porta, et al. 2015), including refractory cytopenia with multilineage dysplasia (RCMD) in 2 cases, and refractory anemia with excess blasts type 2 (RAEB 2) in 1 case; the last patient had WHO defined AML (with, however, only 21 % marrow blasts, i.e. partial blast infiltration very close to MDS diagnostic criteria). Karyotype showed monosomy 7 in 3 cases, with or without other chromosomal deletions, especially monosomy 5, and was not performed in the last patient. 66 67 68 69 70 71 72 Compared to other patients, the 4 patients who developed MDS or AML had received, prior to PPRT, more cycles of chemotherapy (mean: 13.8 versus 4.7, p=0.001), more cycles of alkylating agents (mean: 12.5 versus 3.75, p=0.001), and had more frequently experienced early hematological toxicity (75% versus 13%, p=0.03) (Table 2). Two of the patients received supportive care only and 2 received azacitidine. Survival was 6, 2, 2 and 54 months, respectively. During the study period (2004-2011), 95 additional patients with GEP-NETs were treated with alkylating- 3

Page 4 of 12 73 74 based chemotherapy without subsequent PPRT at our center. Only one (1%) developed MDS. 75 76 77 78 79 80 81 82 83 84 85 86 87 88 89 The high rate of MDS or AML (20%) we report in this limited series of 20 nonresectable NET treated with 177 Lu PPRT after heavy pretreatment with chemotherapy is therefore much higher than in large published series of PRRT (Bodei et al. 2015; Imhof, et al. 2011; Kesavan, et al. 2014; Kwekkeboom et al. 2008; Sabet, et al. 2013). This higher rate may be due to the fact that most of our patients had received chemotherapy and alkylating agents prior to PPRT, compared to previously published series of PPRT in metastatic NET, where less than one third of the patients had received chemotherapy before PRRT. Indeed, in our series, patients who developed MDS or AML had received more chemotherapy and more alkylating agents than other patients (p=0.001). Reasons why patients had received more chemotherapy prior to PRRT in our series probably include the fact that peptide receptor radionuclide therapy was until recently unavailable in France, requiring a partnership with centers abroad. Thus, PRRT was not a frontline therapy for metastatic GEP-NETs in France, where patients first received conventional systemic chemotherapy, targeted therapies, and transarterial chemoembolization. 90 91 92 93 94 95 96 In another study about 65 patients who received 177 Lu-octreoate combined with capecitabine (n=28) or capecitabine and temozolomide chemotherapy (n=37) reported no MDS/AML in the former group, but two (5.4%) cases in the latter group (Kesavan, et al. 2014). Temozolomide is an alkylating agent, further supporting our assumption that the risk of MDS/AML, very low with 177 Lu-octreotate PRRT alone and possibly 177 Lu-octreotate PRRT combined with non-alkylating chemotherapy, may be higher when 177 Lu-octreotate PRRT is combined with alkylating agents. 4

Page 5 of 12 97 98 99 100 101 102 103 104 105 106 107 108 109 110 The duration of follow-up in our series (3.1 years) cannot explain differences of MDS/AML rates compared with other studies: indeed, while three studies had shorter follow-up (1.6, 1.9 and 2.6 years) (Kwekkeboom, et al. 2008; Imhof, et al. 2011; Sabet, et al. 2013), two other studies had similar or longer follow-up (3.0 and 4.2 years) (Kesavan, et al. 2014; Bodei, et al. 2015). Because of the known leukemogenic role of alkylating agents, we cannot exclude that MDS/AML in our series was a direct consequence of prior chemotherapy, the latency before the first cycle of chemotherapy and the diagnosis of the hematological malignancies being of 12.6, 9.8, 3.8 and 10.4 years respectively. Indeed, the maximum risk of MDS/AML ranges from 3 to 10 years after the myelotoxic treatment. On the other hand, the risk of MDS/AML in GEP-NETs treated with alkylating based chemotherapy alone may be relatively small: during the study period (2004-2011), 95 additional GEP-NETs patients were treated with this approach without subsequent PPRT at our center, and only one (1%) developed MDS. 111 112 113 114 115 116 117 118 119 120 Other risk factors for MDS/AML, in our study, included early hematological toxicity after PPRT, observed in 3 of the 4 patients who subsequently developed MDS/AML, compared to only two of the 16 patients who did not develop MDS/AML. Platelet toxicity was also significantly related to development of secondary MDS or AML in a previous study (Bodei et al. 2015) : close hematological monitoring therefore seems recommended in patients experiencing early hematological toxicity after PRRT. MDS and AML cases observed in the present series were typical of those observed after alkylating agents or purine analogs. Indeed, they all had partial marrow blast infiltration (maximum 21%) and the 3 karyotyped patients had monosomy 7. 121 The impact of bone metastases in the occurrence of MDS/AML remained 5

Page 6 of 12 122 123 124 125 126 127 128 129 unclear and it had no significant impact in our study (p=0.39). In a pilot study in 11 patients with GEP-NETs and florid bone metastases (with advanced widespread metastatic bone disease) receiving PRRT (Sabet, et al. 2014), a higher than usual rate of hematological toxicity was reported, with grade 3-4 reversible hematotoxicity of in 35% of the patients, but delayed hematological toxicity was not reported. In other studies, no statistical correlation between bone metastases after PRRT and the recovery of acute hematotoxicity or the subsequent occurrence MDS/AML were seen (Bergsma, et al. 2015) (Sabet et al. 2013). 130 131 132 133 134 135 136 137 138 In conclusion, our study, although based on a limited number of patients, suggests a high occurrence of MDS/AML in patients with GEP-NETs treated with PRRT after previous chemotherapy with alkylating agents. The indication of PRRT should take into consideration the importance of previous chemotherapy. Exposure to alkylating agents should be avoided in patients with low-grade NET, who have a long survival expectancy and a significant likelihood of benefiting from PRRT, since it may compromise the safety and future applicability of this more effective therapy. Regular and prolonged monitoring of blood counts is mandatory, especially in patients experiencing early hematological toxicity after PRRT. 139 140 6

Page 7 of 12 141 References: 142 143 144 145 146 147 148 149 150 151 152 153 154 155 156 157 158 159 160 161 162 163 164 165 166 167 168 169 170 171 172 173 Bergsma H, Konijnenberg MW, Kam BL, Teunissen JJ, Kooij PP, de Herder WW, Franssen GJ, van Eijck CH, Krenning EP & Kwekkeboom DJ 2015 Subacute haematotoxicity after PRRT with Lu-DOTA-octreotate: prognostic factors, incidence and course. Eur J Nucl Med Mol Imaging. Bodei L, Kidd M, Paganelli G, Grana CM, Drozdov I, Cremonesi M, Lepensky C, Kwekkeboom DJ, Baum RP, Krenning EP, et al. 2015 Long-term tolerability of PRRT in 807 patients with neuroendocrine tumours: the value and limitations of clinical factors. Eur J Nucl Med Mol Imaging 42 5-19. Della Porta MG, Tuechler H, Malcovati L, Schanz J, Sanz G, Garcia-Manero G, Sole F, Bennett JM, Bowen D, Fenaux P, et al. 2015 Validation of WHO classificationbased Prognostic Scoring System (WPSS) for myelodysplastic syndromes and comparison with the revised International Prognostic Scoring System (IPSS-R). A study of the International Working Group for Prognosis in Myelodysplasia (IWG-PM). Leukemia. Imhof A, Brunner P, Marincek N, Briel M, Schindler C, Rasch H, Macke HR, Rochlitz C, Muller-Brand J & Walter MA 2011 Response, survival, and long-term toxicity after therapy with the radiolabeled somatostatin analogue [90Y-DOTA]-TOC in metastasized neuroendocrine cancers. J Clin Oncol 29 2416-2423. J. Strosberg EW, B. Chasen, M. Kulke, D. Bushnell, M. Caplin, R.P. Baum, E. Mittra, T. Hobday, A. Hendifar, K. Oberg, M. Lopera Sierra, P. Ruszniewski, D. Kwekkeboom LATE BREAKING ABSTRACT: 177-Lu-Dotatate significantly improves progression-free survival in patients with midgut neuroendocrine tumours: Results of the phase III NETTER-1 trial. European Cancer Congress 2015 Abstract 6LBA. Kesavan M, Claringbold PG & Turner JH 2014 Hematological toxicity of combined 177Lu-octreotate radiopeptide chemotherapy of gastroenteropancreatic neuroendocrine tumors in long-term follow-up. Neuroendocrinology 99 108-117. Kwekkeboom DJ, Bakker WH, Kooij PP, Konijnenberg MW, Srinivasan A, Erion JL, Schmidt MA, Bugaj JL, de Jong M & Krenning EP 2001 [177Lu- DOTAOTyr3]octreotate: comparison with [111In-DTPAo]octreotide in patients. Eur J Nucl Med 28 1319-1325. Kwekkeboom DJ, de Herder WW, Kam BL, van Eijck CH, van Essen M, Kooij PP, Feelders RA, van Aken MO & Krenning EP 2008 Treatment with the radiolabeled 7

Page 8 of 12 174 175 176 177 178 179 180 181 182 183 184 185 186 187 somatostatin analog [177 Lu-DOTA 0,Tyr3]octreotate: toxicity, efficacy, and survival. J Clin Oncol 26 2124-2130. Pavel M, Baudin E, Couvelard A, Krenning E, Oberg K, Steinmuller T, Anlauf M, Wiedenmann B, Salazar R & Barcelona Consensus Conference p 2012 ENETS Consensus Guidelines for the management of patients with liver and other distant metastases from neuroendocrine neoplasms of foregut, midgut, hindgut, and unknown primary. Neuroendocrinology 95 157-176. Sabet A, Ezziddin K, Pape UF, Ahmadzadehfar H, Mayer K, Poppel T, Guhlke S, Biersack HJ & Ezziddin S 2013 Long-term hematotoxicity after peptide receptor radionuclide therapy with 177Lu-octreotate. J Nucl Med 54 1857-1861. Sabet A, Khalaf F, Yong-Hing CJ, Sabet A, Haslerud T, Ahmadzadehfar H, Guhlke S, Grunwald F, Biersack HJ & Ezziddin S 2014 Can peptide receptor radionuclide therapy be safely applied in florid bone metastases? A pilot analysis of late stage osseous involvement. Nuklearmedizin 53 54-59. 188 189 190 191 8

Page 9 of 12 192 193 194 195 196 197 198 199 200 201 202 203 204 205 206 Authors: Bertrand Brieau 1, MD, Olivia Hentic 1, MD, Rachida Lebtahi 2, Maxime Palazzo 1, MD, Makrem Ben Reguiga 3, MD, Vinciane Rebours 1, MD, PhD, Frederique Maire 1, MD, Pascal Hammel 1, MD, PhD, Philippe Ruszniewski 1, MD, PhD, Pierre Fenaux 4, MD, PhD. Affiliations: 1 : Service de gastro-entérologie et pancréatologie, Hôpital Beaujon, AP-HP, Université Paris Diderot, Clichy, France 2 : Service de médecine nucléaire, Hôpital Beaujon, AP-HP, Université Paris Diderot Clichy, France. 3 : Département de pharmacie, Hôpital Beaujon, AP-HP, Université Paris Diderot Clichy, France. 4 : Service d hématologie, Hôpital Saint Louis, AP-HP, Université Paris Diderot, Paris, France. Word Count: 1463 References: 11 Pages: 10 Tables: 2 207 208 209 210 211 212 213 Correspondence to: Pierre Fenaux, MD, PhD Service d hématologie, Hôpital Saint Louis, 1 avenue Claude Vellefaux, Paris, France. E-mail: pierre.fenaux@aphp.fr Phone: +33 1 71 20 70 18 Fax: +33 1 71 20 70 20 214 215 Conflicts of interest: none. 216 9

Page 10 of 12 217 218 219 220 221 222 223 224 225 226 Declaration of interest: Bertrand Brieau, Olivia Hentic, Rachida Lebtahi, Maxime Palazzo, Hela Boudabous, Makrem Ben Reguiga, Vinciane Rebours, Frederique Maire, Pascal Hammel, Philippe Ruszniewski and Pierre Fenaux have non conflict of interest to declare with the subject. Funding: This research did not receive any specific grant from any funding agency in the public, commercial or not-for-profit sector. 10

Page 11 of 12 Table 1 Patients characteristics Patient, age Type of tumor, Treatments before PRRT: time interval from diagnosis, type of at diagnosis ENETS grade treatment (number of cycles) and response (g1/g2/g3) Hematological toxicites of each treatment N of CT cycles and time from the first cycle to PRRT (months) Bone metastases (localization) Bone metastases (metabolic burden)* PRRT: N of cycles, total dose (Gbq), and tumor response (CR/PR/SD/PD) AHT of PRRT, durationdht (months) N 1, 45 Pancreatic NET, S at 2 months: left pancreatectomy with complete resection 25, 121 mo Left ischial tuberosity and occipital G2 4, 29.9, PR Yes g1 part of the skull CT at 54 months: streptozotocin + adriamycin + interferon (9), PR G2 NE, G1 TH TACE at 75 months: streptozotocin (3), SD S at 115 months: liver transplantation CT at 116 months (adjuvant): 5-fluorouracile + streptozotocin (4), SD RT at 160 months (hip and skull bones): clinical response CT at 163 months: 5-fluorouracil + streptozotocin (4), PD CT at 170 months: FOLFIRI regimen (5), SD N 2, 54 Pancreatic NET, CT at 4 months: 5-fluorouracil + streptozotocin (8), SD 12, 47 mo G0 4, 29.9, CR G3 TH, G1 AN, 12 mo Yes g1 CT at 54 months: adriamycin + dacarbazine (4), SD N 3, 66 Metastatic NET with unknown primary, g1 CT at 6 months: 5-fluorouracil + streptozotocin (6), SD 6, 15 mo Disseminated metastases of the spine, sternum, ribs, and bony pelvis G4 4, 26.5, PR G3 NE, G4 TH, G1 AN, 12 mo Yes N 4, 50 Gastric NET, g2 TACE at 4 months: streptozotocin (4), PR 12, 71 mo thoracic vertebrae (2nd and 7th) G0 4, 30, CR G3 NE, G3 AN, G3 TH, 54 mo CT at 34 months: 5-fluorouracil + dacarbazine (4), SD S at 48 months: liver transplantation CT at 50 months (adjuvant): 5-fluorouracil + streptozotocin (4), SD N 5, 37 Rectal NET, g2 TACE at 2 months: streptozotocin (1), SD 1, 6 mo Cervical vertebrae, left iliac wing G1 4, 25.75, PR G2 AN, G3 TH, G1 NE, 18 mo Yes No N 6, 49 Pancreatic NET, CT at 3 months: FOLFIRI regimen (10), PR 19, 32 mo Two lumbar vertebrae, sacrum G2 1, 7.5, SD G4 AN, G4 TH, G4 NE, g2 5 mo CT at 22 months: etoposid + cisplatin (4), PD CT at 25 months: sunitinib (5), SD G2 NE N 7, 68 Rectal NET, g2 TACE at 3 months: streptozotocin (3), SD 3, 11 mo Disseminated metastases of the spine, ribs N 8, 69 Rectal NET, g2 S at 1 month: rectal resection with coloanal anastomosis 9, 21 mo Disseminated metastases of the spine and ribs CT at 70 months: 5-fluorouracil + dacarbazine (8), SD TACE at 83 months: Streptozotocin (1), PR G3 6, 45, PR No G3 4, 30, PR No N 9, 59 Ileum NET, g1 S at 1 month: right ileo-colectomy 4, 21 mo G0 4, 30, SD No CT at 43 months: 5-fluorouracil + streptozotocin (4), PD N 10, 64 Ileum NET, g1 S at 1 month: Segmentary resection of the ileum tumor 0 G0 4, 30, PD No N 11, 61 Pancreatic NET, 0 G0 4, 30, CR No g1 N 12, 71 Pancreatic NET 0 G0 4, 30, PR No N 13, 46 Pancreatic NET, S at 2 months: cephalic duodenopancreatectomy 0 G0 4, 30, PR No g2 N 14, 53 Ileum NET, g2 S at 1 month: Segmentary resection of the ileum tumor 0 Left scapula, right hip G2 4, 30, PR No N 15, 47 Ileum NET, g1 CT at 12 months: 5-fluorouracil + streptozotocin (2), SD 2, 9 mo G0 4, 30, PR No N 16, 55 Colon NET, g1 S at 1 month: resection of the primary tumor 0 One thoracic vertebrae G1 4, 30, CR No No N 17, 26 Duodenal NET, g2 S at 1 month: cephalic duodenopancreatectomy 8, 12 mo Disseminated metastases of the spine G3 4, 30, SD No CT at 8 months: adriamycin + streptozotocin (4), PD CT at 15 months: FOLFIRI regimen + bevacizumab (4), PD N 18, 16 Pancreatic NET, S at 1 month: left pancreatectomy 6, 131 mo Sacrum and right hip G1 4, 30, PR No g1 S at 8 months: right hepatectomy CT at 10 months (adjuvant): adriamycin + streptozotocin (6), SD S at 51 months: liver resection N 19, 31 Anus NET, g2 S at 1 month: abdominoperineal resection 4, 12 mo G0 4, 30, PR No CT at 121 months: 5-fluorouracil + dacarbazine (4), PD N 20, 64 Pancreatic NET, CT at 2 months: etoposid + cisplatin (6), SD G3 NE 19, 18 mo G0 3, 22.5, PD No g2 CT at 10 months: FOLFIRI regimen (13), PR G2 NE NET: neuroendocrine tumor; PRRT: peptide receptor radionuclide therapy; S: surgery; CT: chemotherapy; RT: radiotherapy; TACE: tranzarterial chemoembolisation; G1: grade 1; G2: grade 2; G3: grade 3; CR: complete response; PR: partial response; SD: stable disease; PD: progressive disease; AHT: acute hematological toxicity; DHT: delayed hematological toxicity; TH: thrombopenia; NE: neutropenia; AN: anemia *: Bone involvement is graded according to the results of the OctreoScan. G0: no metastases; G1: 1 to 5 bone uptakes; G2: 6 to 10 bone uptakes; G3: more than 10 bone uptakes with disseminated spine involvement; G4: G3 and bone marrow involvement Tumor aggressiveness was evaluated according to the ENETS grade (Ki67 proliferative index) : g1 = tumor with Ki-67 from 0 to 2%; g2 : Ki-67 from 3 to 20%; g3: Ki67 superior than 20%.

Page 12 of 12 Table 2 prognostic factors of occurrence of MDS and AML in patients treated with PRRT Patients who developed MDS/AML: n (%) Other patients: n (%) P value Total 4 (20) 16 (80) - Gender F/M 3 (75) 4 (25) 0.16 Age at diagnosis (years) Median (range) 53.8 (45-66) 51 (16-71) 0.63 Mean number of cycles of previous chemotherapy (range) 13.8 (6-25) 4.7 (0-19) 0.001 Alkylating-based chemotherapy mean number of cycles (range) 12.5 (6-20) 3.75 (0-9) 0.001 Bone metastases before PRRT 3 (75) 8 (50) 0.39 Immunosuppressive treatment 2 (50) 0 (0) 0.006 Mean dose of PRRT (GBq) 29 30.5 0.94 Mean number of cycles of PRRT 4 4 0.97 Early hematological toxicity grade 3-4 3 (75) 2 (13) 0.03 Number of deaths 4 (100) 4 (29) - Cause of deaths: Underlying tumor MDS/AML 0 (0) 4(100) 4 (29) 0 (0) - - MDS: myelodysplastic syndrome; AML: acute myeloid leukemia; PRRT: peptide receptor radionuclide therapy