The joint IAEA, EANM, and SNMMI practical guidance on peptide receptor radionuclide therapy (PRRNT) in neuroendocrine tumours

L Bodei, J Mueller-Brand, R P Baum, M E Pavel, D Hörsch, M S O'Dorisio, T M O'Dorisio, J R Howe, M Cremonesi, D J Kwekkeboom, John J Zaknun, L Bodei, J Mueller-Brand, R P Baum, M E Pavel, D Hörsch, M S O'Dorisio, T M O'Dorisio, J R Howe, M Cremonesi, D J Kwekkeboom, John J Zaknun

Abstract

Peptide receptor radionuclide therapy (PRRNT) is a molecularly targeted radiation therapy involving the systemic administration of a radiolabelled peptide designed to target with high affinity and specificity receptors overexpressed on tumours. PRRNT employing the radiotagged somatostatin receptor agonists (90)Y-DOTATOC ([(90)Y-DOTA(0),Tyr(3)]-octreotide) or (177)Lu-DOTATATE ([(177)Lu-DOTA(0),Tyr(3),Thr(8)]-octreotide or [(177)Lu-DOTA(0),Tyr(3)]-octreotate) have been successfully used for the past 15 years to target metastatic or inoperable neuroendocrine tumours expressing the somatostatin receptor subtype 2. Accumulated evidence from clinical experience indicates that these tumours can be subjected to a high absorbed dose which leads to partial or complete objective responses in up to 30 % of treated patients. Survival analyses indicate that patients presenting with high tumour receptor expression at study entry and receiving (177)Lu-DOTATATE or (90)Y-DOTATOC treatment show significantly higher objective responses, leading to longer survival and improved quality of life. Side effects of PRRNT are typically seen in the kidneys and bone marrow. These, however, are usually mild provided adequate protective measures are undertaken. Despite the large body of evidence regarding efficacy and clinical safety, PRRNT is still considered an investigational treatment and its implementation must comply with national legislation, and ethical guidelines concerning human therapeutic investigations. This guidance was formulated based on recent literature and leading experts' opinions. It covers the rationale, indications and contraindications for PRRNT, assessment of treatment response and patient follow-up. This document is aimed at guiding nuclear medicine specialists in selecting likely candidates to receive PRRNT and to deliver the treatment in a safe and effective manner. This document is largely based on the book published through a joint international effort under the auspices of the Nuclear Medicine Section of the International Atomic Energy Agency.

References

    1. Patel YC. Somatostatin and its receptor family. Front Neuroendocrinol. 1999;20:157–198. doi: 10.1006/frne.1999.0183.
    1. Waser B, Tamma ML, Cescato R, Maecke HR, Reubi JC. Highly efficient in vivo agonist-induced internalization of sst2 receptors in somatostatin target tissues. J Nucl Med. 2009;50:936–941. doi: 10.2967/jnumed.108.061457.
    1. Reubi JC, Schär JC, Waser B, Wenger S, Heppeler A, Schmitt JS, et al. Affinity profiles for human somatostatin receptor subtypes SST1-SST5 of somatostatin radiotracers selected for scintigraphic and radiotherapeutic use. Eur J Nucl Med. 2000;27:273–282. doi: 10.1007/s002590050034.
    1. Maecke HR, Reubi JC. Somatostatin receptors as targets for nuclear medicine imaging and radionuclide treatment. J Nucl Med. 2011;52:841–844. doi: 10.2967/jnumed.110.084236.
    1. Rindi G. The ENETS guidelines: the new TNM classification system. Tumori. 2010;96:806–809.
    1. Modlin IM, Oberg K, Chung DC, Jensen RT, de Herder WW, Thakker RV, et al. Gastroenteropancreatic neuroendocrine tumors. Lancet Oncol. 2008;9:61–72. doi: 10.1016/S1470-2045(07)70410-2.
    1. Sundin A, Vullierme MP, Kaltsas G, Plöckinger U, Mallorca Consensus Conference participants; European Neuroendocrine Tumor Society ENETS Consensus Guidelines for the Standards of Care in Neuroendocrine Tumors: radiological examinations. Neuroendocrinology. 2009;90:167–183. doi: 10.1159/000184855.
    1. Bombardieri E, Ambrosini V, Aktolun C, Baum RP, Bishof-Delaloye A, Del Vecchio S, et al. Oncology Committee of the EANM. 111In-pentetreotide scintigraphy: procedure guidelines for tumour imaging. Eur J Nucl Med Mol Imaging. 2010;37:1441–1448. doi: 10.1007/s00259-010-1473-6.
    1. Virgolini I, Ambrosini V, Bomanji JB, Baum RP, Fanti S, Gabriel M, et al. Procedure guidelines for PET/CT tumor imaging with 68Ga-DOTA-conjugated peptides: 68Ga-DOTA-TOC, 68Ga-DOTA-NOC, 68Ga-DOTA-TATE. Eur J Nucl Med Mol Imaging. 2010;37:2004–2010. doi: 10.1007/s00259-010-1512-3.
    1. Binderup T, Knigge U, Loft A, Federspiel B, Kjaer A. 18F-fluorodeoxyglucose positron emission tomography predicts survival of patients with neuroendocrine tumors. Clin Cancer Res. 2010;16:978–985. doi: 10.1158/1078-0432.CCR-09-1759.
    1. Kerström G, Hellman P, Hessman O. Midgut carcinoid tumors: surgical treatment and prognosis. Best Pract Res Clin Gastroenterol. 2005;19:717–728. doi: 10.1016/j.bpg.2005.05.005.
    1. Plöckinger U, Couvelard A, Falconi M, Sundin A, Salazar R, Christ E, et al. Consensus guidelines for the management of patients with digestive neuroendocrine tumors: well-differentiated tumor/carcinoma of the appendix and goblet cell carcinoma. Neuroendocrinology. 2008;87:20–30. doi: 10.1159/000109876.
    1. Ramage JK, Goretzki PE, Manfredi R, Komminoth P, Ferone D, Hyrdel R, et al. Consensus guidelines for the management of patients with digestive neuroendocrine tumors: well-differentiated colon and rectum tumor/carcinoma. Neuroendocrinology. 2008;87:31–39. doi: 10.1159/000111036.
    1. Givi B, Pommier SJ, Thompson AK, Diggs BS, Pommier RF. Operative resection of primary carcinoid neoplasms in patients with liver metastases yields significantly better survival. Surgery. 2006;140:891–897. doi: 10.1016/j.surg.2006.07.033.
    1. Steinmüller T, Kianmanesh R, Falconi M, Scarpa A, Taal B, Kwekkeboom DJ, et al. Consensus guidelines for the management of patients with liver metastases from digestive (neuro)endocrine tumors: foregut, midgut, hindgut, and unknown primary. Neuroendocrinology. 2008;87:47–62. doi: 10.1159/000111037.
    1. Vogl TJ, Naguib NN, Zangos S, Eichler K, Hedayati A, Nour-Eldin NE. Liver metastases of neuroendocrine carcinomas: interventional treatment via transarterial embolization, chemoembolization and thermal ablation. Eur J Radiol. 2009;72:517–528. doi: 10.1016/j.ejrad.2008.08.008.
    1. Kamat PP, Gupta S, Ensor JE, Murthy R, Ahrar K, Madoff DC, et al. Hepatic arterial embolization and chemoembolization in the management of patients with large-volume liver metastases. Cardiovasc Intervent Radiol. 2008;31:299–307. doi: 10.1007/s00270-007-9186-3.
    1. Ruszniewski P, Malka D. Hepatic arterial chemoembolization in the management of advanced digestive endocrine tumors. Digestion. 2000;62(Suppl 1):79–83. doi: 10.1159/000051860.
    1. King J, Quinn R, Glenn DM, Janssen J, Tong D, Liaw W, et al. Radioembolization with selective internal radiation microspheres for neuroendocrine liver metastases. Cancer. 2008;113:921–929. doi: 10.1002/cncr.23685.
    1. Kennedy AS, Dezarn WA, McNeillie P, Coldwell D, Nutting C, Carter D, et al. Radioembolization for unresectable neuroendocrine hepatic metastases using resin 90Y-microspheres: early results in 148 patients. Am J Clin Oncol. 2008;31:271–279. doi: 10.1097/COC.0b013e31815e4557.
    1. Ruszniewski P, Ish-Shalom S, Wymenga M, O’Toole D, Arnold R, Tomassetti P, et al. Rapid and sustained relief from the symptoms of carcinoid syndrome: results from an open 6-month study of the 28-day prolonged-release formulation of lanreotide. Neuroendocrinology. 2004;80:244–251. doi: 10.1159/000082875.
    1. Modlin IM, Pavel M, Kidd M, Gustafsson BI. Review article: somatostatin analogues in the treatment of gastroenteropancreatic neuroendocrine (carcinoid) tumors. Aliment Pharmacol Ther. 2010;31:169–188.
    1. Eriksson B, Klöppel G, Krenning E, Ahlman H, Plöckinger U, Wiedenmann B, et al. Consensus guidelines for the management of patients with digestive neuroendocrine tumors – well-differentiated jejunal-ileal tumor/carcinoma. Neuroendocrinology. 2008;87:8–19. doi: 10.1159/000111034.
    1. Rinke A, Müller HH, Schade-Brittinger C, Klose KJ, Barth P, Wied M, et al. Placebo-controlled, double-blind, prospective, randomized study on the effect of octreotide LAR in the control of tumor growth in patients with metastatic neuroendocrine midgut tumors: a report from the PROMID Study Group. J Clin Oncol. 2009;27:4656–4663. doi: 10.1200/JCO.2009.22.8510.
    1. Öberg K. Interferon in the management of neuroendocrine GEP tumors. Digestion. 2000;62(Suppl 1):92–97.
    1. Eriksson B, Öberg K. An update of the medical treatment of malignant endocrine pancreatic tumors. Acta Oncol. 1993;32:203–208. doi: 10.3109/02841869309083913.
    1. Moertel CG, Kvols LK, O’Connell MJ, Rubin J. Treatment of neuroendocrine carcinomas with combined etoposide and cisplatin. Evidence of major therapeutic activity in the anaplastic variants of these neoplasms. Cancer. 1991;68:227–232. doi: 10.1002/1097-0142(19910715)68:2<227::AID-CNCR2820680202>;2-I.
    1. Moertel CG, Johnson CM, McKusick MA, Martin JK, Jr, Nagorney DM, Kvols LK, et al. The management of patients with advanced carcinoid tumors and islet cell carcinomas. Ann Intern Med. 1994;120:302–309. doi: 10.7326/0003-4819-120-4-199402150-00008.
    1. Mani MA, Shroff RT, Jacobs C, Wolff RA, Ajani JA, Yao JC, et al. A phase II study of irinotecan and cisplatin for metastatic or unresectable high grade neuroendocrine carcinoma [abstract]. J Clin Oncol 2008;26 Suppl; abstr 15550
    1. Bajetta E, Catena L, Procopio G, De Dosso S, Bichisao E, Ferrari L, et al. Are capecitabine and oxaliplatin (XELOX) suitable treatments for progressing low-grade and high-grade neuroendocrine tumors? Cancer Chemother Pharmacol. 2007;59:637–642. doi: 10.1007/s00280-006-0306-6.
    1. Kouvaraki MA, Ajani JA, Hoff P, Wolff R, Evans DB, Lozano R, et al. Fluorouracil, doxorubicin, and streptozocin in the treatment of patients with locally advanced and metastatic pancreatic endocrine carcinomas. J Clin Oncol. 2004;22:4762–4771. doi: 10.1200/JCO.2004.04.024.
    1. Fjallskog ML, Janson ET, Falkmer UG, Vatn MH, Oberg KE, Eriksson BK. Treatment with combined streptozotocin and liposomal doxorubicin in metastatic endocrine pancreatic tumors. Neuroendocrinology. 2008;88:53–58. doi: 10.1159/000117575.
    1. Moertel CG, Lefkopoulo M, Lipsitz S, Hahn RG, Klaassen D. Streptozocin-doxorubicin, streptozocin-fluorouracil or chlorozotocin in the treatment of advanced islet-cell carcinoma. N Engl J Med. 1992;326:519–523. doi: 10.1056/NEJM199202203260804.
    1. Strosberg JR, Fine RL, Choi J, Nasir A, Coppola D, Chen DT, et al. First-line chemotherapy with capecitabine and temozolomide in patients with metastatic pancreatic endocrine carcinomas. Cancer. 2011;117:268–275. doi: 10.1002/cncr.25425.
    1. Kulke MH, Stuart K, Enzinger PC, Ryan DP, Clark JW, Muzikansky A, et al. Phase II study of temozolomide and thalidomide in patients with metastatic neuroendocrine tumors. J Clin Oncol. 2006;24:401–406. doi: 10.1200/JCO.2005.03.6046.
    1. Eriksson B, Annibale B, Bajetta E, Mitry E, Pavel M, Platania M, et al. ENETS Consensus Guidelines for the Standards of Care in Neuroendocrine Tumors: chemotherapy in patients with neuroendocrine tumors. Neuroendocrinology. 2009;90:214–219. doi: 10.1159/000225950.
    1. Pavel M, Hainsworth JD, Baudin E, et al. A randomized, double-blind, placebo-controlled, multicenter phase III trial of everolimus plus octreotide LAR vs placebo plus octreotide LAR in patients with advanced neuroendocrine tumors (NET) (RADIANT-2) Ann Oncol. 2010;21(Suppl 8):viii4.
    1. Yao JC, Phan A, Hoff PM, Chen HX, Charnsangavej C, Yeung SC, et al. Targeting vascular endothelial growth factor in advanced carcinoid tumor: a random assignment phase II study of depot octreotide with bevacizumab and pegylated interferon alpha-2b. J Clin Oncol. 2008;26:1316–1323. doi: 10.1200/JCO.2007.13.6374.
    1. Raymond E, Dahan L, Raoul JL, Bang YJ, Borbath I, Lombard-Bohas C, et al. Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. N Engl J Med. 2011;364:501–513. doi: 10.1056/NEJMoa1003825.
    1. Yao JC, Lombard-Bohas C, Baudin E, Kvols LK, Rougier P, Ruszniewski P, et al. Daily oral everolimus activity in patients with metastatic pancreatic neuroendocrine tumors after failure of cytotoxic chemotherapy: a phase II trial. J Clin Oncol. 2010;28:69–76. doi: 10.1200/JCO.2009.24.2669.
    1. Yao JC, Shah MH, Ito T, Bohas CL, Wolin EM, Van Cutsem E, et al. Everolimus for advanced pancreatic neuroendocrine tumors. N Engl J Med. 2011;364:514–523. doi: 10.1056/NEJMoa1009290.
    1. Patrick DL, Ferketich SL, Frame PS, Harris JJ, Hendricks CB, Levin B, et al. National Institutes of Health State-of-the-Science Panel. National Institutes of Health State-of-the-Science Conference Statement: Symptom Management in Cancer: Pain, Depression, and Fatigue, July 15–17, 2002. J Natl Cancer Inst. 2003;95:1110–1117. doi: 10.1093/jnci/djg014.
    1. Valkema R, De Jong M, Bakker WH, Breeman WA, Kooij PP, Lugtenburg PJ, et al. Phase I study of peptide receptor radionuclide therapy with [In-DTPA]-octreotide: the Rotterdam experience. Semin Nucl Med. 2002;32:110–122. doi: 10.1053/snuc/2002.31025.
    1. De Jong M, Bakker WH, Breeman WA, Bernard BF, Hofland LJ, Visser TJ, et al. Pre-clinical comparison of [DTPA0] octreotide, [DTPA0,Tyr3] octreotide and [DOTA0,Tyr3] octreotide as carriers for somatostatin receptor-targeted scintigraphy and radionuclide therapy. Int J Cancer. 1998;75:406–411. doi: 10.1002/(SICI)1097-0215(19980130)75:3<406::AID-IJC14>;2-6.
    1. Kwekkeboom DJ, Mueller-Brand J, Paganelli G, Anthony LB, Pauwels S, Kvols LK, et al. Overview of results of peptide receptor radionuclide therapy with 3 radiolabeled somatostatin analogs. J Nucl Med. 2005;46(Suppl 1):62S–66S.
    1. Kwekkeboom DJ, Kam BL, van Essen M, Teunissen JJ, van Eijck CH, Valkema R, et al. Somatostatin receptor-based imaging and therapy of gastroenteropancreatic neuroendocrine tumors. Endocr Relat Cancer. 2010;17:R53–R73. doi: 10.1677/ERC-09-0078.
    1. Bodei L, Ferone D, Grana CM, Cremonesi M, Signore A, Dierckx RA, et al. Peptide receptor therapies in neuroendocrine tumors. J Endocrinol Invest. 2009;32:360–369.
    1. Kwekkeboom DJ, de Herder WW, Kam BL, van Eijck CH, van Essen M, Kooij PP, et al. Treatment with the radiolabeled somatostatin analog [177Lu-DOTA0,Tyr3]octreotate: toxicity, efficacy, and survival. J Clin Oncol. 2008;26:2124–2130. doi: 10.1200/JCO.2007.15.2553.
    1. Bushnell DL, Jr, O’Dorisio TM, O’Dorisio MS, Menda Y, Hicks RJ, Van Cutsem E, et al. 90Y-Edotreotide for metastatic carcinoid refractory to octreotide. J Clin Oncol. 2010;28:1652–1659. doi: 10.1200/JCO.2009.22.8585.
    1. Imhof A, Brunner P, Marincek N, Briel M, Schindler C, Rasch H, et al. Response, survival, and long-term toxicity after therapy with the radiolabeled somatostatin analogue [90Y-DOTA]-TOC in metastasized neuroendocrine cancers. J Clin Oncol. 2011;29:2416–2423. doi: 10.1200/JCO.2010.33.7873.
    1. Gains JE, Bomanji JB, Fersht NL, Sullivan T, D’Souza D, Sullivan KP, et al. 177Lu-DOTATATE molecular radiotherapy for childhood neuroblastoma. J Nucl Med. 2011;52:1041–1047. doi: 10.2967/jnumed.110.085100.
    1. Bartolomei M, Bodei L, De Cicco C, Grana CM, Cremonesi M, Botteri E, et al. Peptide receptor radionuclide therapy with (90)Y-DOTATOC in recurrent meningioma. Eur J Nucl Med Mol Imaging. 2009;36:1407–1416. doi: 10.1007/s00259-009-1115-z.
    1. Iten F, Müller B, Schindler C, Rochlitz C, Oertli D, Mäcke HR, et al. Response to [90Yttrium-DOTA]-TOC treatment is associated with long-term survival benefit in metastasized medullary thyroid cancer: a phase II clinical trial. Clin Cancer Res. 2007;13:6696–6702. doi: 10.1158/1078-0432.CCR-07-0935.
    1. Bodei L, Handkiewicz-Junak D, Grana C, Mazzetta C, Rocca P, Bartolomei M, et al. Receptor radionuclide therapy with 90Y-DOTATOC in patients with medullary thyroid carcinomas. Cancer Biother Radiopharm. 2004;19:65–71. doi: 10.1089/108497804773391694.
    1. Iten F, Muller B, Schindler C, Rasch H, Rochlitz C, Oertli D, et al. [(90)Yttrium-DOTA]-TOC response is associated with survival benefit in iodine-refractory thyroid cancer: long-term results of a phase 2 clinical trial. Cancer. 2009;115:2052–2062. doi: 10.1002/cncr.24272.
    1. Teunissen JJ, Kwekkeboom DJ, Kooij PP, Bakker WH, Krenning EP. Peptide receptor radionuclide therapy for non-radioiodine-avid differentiated thyroid carcinoma. J Nucl Med. 2005;46(Suppl 1):107S–114S.
    1. Bodei L, Cremonesi M, Ferrari M, Pacifici M, Grana CM, Bartolomei M, et al. Long-term evaluation of renal toxicity after peptide receptor radionuclide therapy with 90Y-DOTATOC and 177Lu-DOTATATE: the role of associated risk factors. Eur J Nucl Med Mol Imaging. 2008;35:1847–1856. doi: 10.1007/s00259-008-0778-1.
    1. de Jong M, Krenning EP. New advances in peptide receptor radionuclide therapy. J Nucl Med. 2002;43:617–620.
    1. Jamar F, Barone R, Mathieu I, Walrand S, Labar D, Carlier P, et al. (86YDOTA0)-DPhe1-Tyr3-octreotide (SMT487) – a phase 1 clinical study: pharmacokinetics, biodistribution and renal protective effect of different regimens of amino acid co-infusion. Eur J Nucl Med Mol Imaging. 2003;30:510–518. doi: 10.1007/s00259-003-1117-1.
    1. Bodei L, Cremonesi M, Grana C, Rocca P, Bartolomei M, Chinol M, et al. Receptor radionuclide therapy with 90Y-[DOTA]0-Tyr3-octreotide (90Y-DOTATOC) in neuroendocrine tumors. Eur J Nucl Med Mol Imaging. 2004;31:1038–1046. doi: 10.1007/s00259-004-1571-4.
    1. Giovacchini G, Nicolas G, Freidank H, Mindt TL, Forrer F. Effect of amino acid infusion on potassium serum levels in neuroendocrine tumour patients treated with targeted radiopeptide therapy. Eur J Nucl Med Mol Imaging. 2011;38:1675–1682. doi: 10.1007/s00259-011-1826-9.
    1. Watkins J. Reactions to gelatin plasma expanders. Lancet. 1994;344:328–329. doi: 10.1016/S0140-6736(94)91366-8.
    1. Rolleman EJ, Melis M, Valkema R, Boerman OC, Krenning EP, de Jong M. Kidney protection during peptide receptor radionuclide therapy with somatostatin analogues. Eur J Nucl Med Mol Imaging. 2010;37:1018–1031. doi: 10.1007/s00259-009-1282-y.
    1. Barron ME, Wilkes MM, Navickis RJ. A systematic review of the comparative safety of colloids. Arch Surg. 2004;139:552–563. doi: 10.1001/archsurg.139.5.552.
    1. Wehrmann C, Senftleben S, Zachert C, Müller D, Baum RP. Results of individual patient dosimetry in peptide receptor radionuclide therapy with 177Lu DOTA-TATE and 177Lu DOTA-NOC. Cancer Biother Radiopharm. 2007;22:406–416. doi: 10.1089/cbr.2006.325.
    1. Breeman WAP, de Blois E, Bakker WH, Krenning EP. Radiolabeling DOTA-peptides with 90Y and 177Lu to a high specific activity. In: Chinol M, Paganelli G, editors. Radionuclide peptide cancer therapy. New York: Taylor & Francis; 2006. pp. 119–126.
    1. Menda Y, O’Dorisio MS, Kao S, Khanna G, Michael S, Connolly M, et al. Phase I trial of 90Y-DOTATOC therapy in children and young adults with refractory solid tumors that express somatostatin receptors. J Nucl Med. 2010;51:1524–1531. doi: 10.2967/jnumed.110.075226.
    1. Schmidt M, Baum RP, Simon T, Howman-Giles R. Therapeutic nuclear medicine in pediatric malignancy. Q J Nucl Med Mol Imaging. 2010;54:411–428.
    1. Stabin MG, Sparks RB, Crowe E. OLINDA/EXM: the second-generation personal computer software for internal dose assessment in nuclear medicine. J Nucl Med. 2005;46:1023–1027.
    1. RADAR (RAdiation Dose Assessment Resource) [Internet]. Available from: .
    1. Siegel JA, Thomas SR, Stubbs JB, Stabin MG, Hays MT, Koral KF, et al. Techniques for quantitative radiopharmaceutical biodistribution data acquisition and analysis for use in human radiation dose estimates. MIRD Pamphlet No.16. J Nucl Med. 1999;40:S37–S61.
    1. Foster D, Barret P. Developing and testing integrated multicompartment models to describe a single-input multiple-output study using the SAAM II software system. In: Stelson A, Stabin M, Sparks R, editors. Proceedings of the Sixth International Radiopharmaceutical. Dosimetry Symposium, Oak Ridge Institute for Science and Education; 1996 May 7–10; Oak Ridge, TN: Associated Universities; 1998
    1. Sgouros G. Dosimetry of internal emitters. J Nucl Med. 2005;46(Suppl 1):18S–27S.
    1. Forrer F, Krenning EP, Kooij PP, Bernard BF, Konijnenberg M, Bakker WH, et al. Bone marrow dosimetry in peptide receptor radionuclide therapy with [177Lu-DOTA(0),Tyr(3)]octreotate. Eur J Nucl Med Mol Imaging. 2009;36:1138–1146. doi: 10.1007/s00259-009-1072-6.
    1. Cremonesi M, Ferrari M, Bodei L, Tosi G, Paganelli G. Dosimetry in peptide radionuclide receptor therapy: a review. J Nucl Med. 2006;47:1467–1475.
    1. Cremonesi M, Botta F, Di Dia A, Ferrari M, Bodei L, De Cicco C, et al. Dosimetry for treatment with radiolabelled somatostatin analogues. A review. Q J Nucl Med Mol Imaging. 2010;54:37–51.
    1. Minarik D, Ljungberg M, Segars P, Gleisner KS. Evaluation of quantitative planar 90Y bremsstrahlung whole-body imaging. Phys Med Biol. 2009;54:5873–5883. doi: 10.1088/0031-9155/54/19/014.
    1. Walrand S, Flux GD, Konijnenberg MW, Valkema R, Krenning EP, Lhommel R, et al. Dosimetry of yttrium-labelled radiopharmaceuticals for internal therapy: 86Y or 90Y imaging? Eur J Nucl Med Mol Imaging. 2011;38:S57–S68. doi: 10.1007/s00259-011-1771-7.
    1. Forrer F, Uusijärvi H, Waldherr C, Cremonesi M, Bernhardt P, Mueller-Brand J, et al. A comparison of 111In-DOTATOC and 111In-DOTATATE: biodistribution and dosimetry in the same patients with metastatic neuroendocrine tumours. Eur J Nucl Med Mol Imaging. 2004;31:1257–1262. doi: 10.1007/s00259-004-1553-6.
    1. Rodrigues M, Traub-Weidinger T, Li S, Ibi B, Virgolini I. Comparison of 111In-DOTA-DPhe1-Tyr3-octreotide and 111In-DOTA-lanreotide scintigraphy and dosimetry in patients with neuroendocrine tumours. Eur J Nucl Med Mol Imaging. 2006;33:532–540. doi: 10.1007/s00259-005-0020-3.
    1. Forster GJ, Engelbach MJ, Brockmann JJ, Reber HJ, Buchholz HG, Macke HR, et al. Preliminary data on biodistribution and dosimetry for therapy planning of somatostatin receptor positive tumours: comparison of 86Y-DOTATOC and 111In-DTPA-octreotide. Eur J Nucl Med. 2001;28:1743–1750. doi: 10.1007/s002590100628.
    1. Helisch A, Förster GJ, Reber H, Buchholz HG, Arnold R, Goke B, et al. Pre-therapeutic dosimetry and biodistribution of 86Y-DOTA-Phe 1-Tyr3-octreotide versus 111In-pentetreotide in patients with advanced neuroendocrine tumours. Eur J Nucl Med Mol Imaging. 2004;31:1386–1392. doi: 10.1007/s00259-004-1561-6.
    1. Kwekkeboom DJ, Kooij PP, Bakker WH, Macke HR, Krenning EP. Comparison of 111In-DOTA-Tyr3-octreotide and 111In-DTPA-octreotide in the same patients: biodistribution, kinetics, organ and tumor uptake. J Nucl Med. 1999;40:762–767.
    1. Hindorf C, Chittenden S, Causer L, Lewington VJ, Mäcke HR, Flux GD. Dosimetry for (90)Y-DOTATOC therapies in patients with neuroendocrine tumors. Cancer Biother Radiopharm. 2007;22:130–135. doi: 10.1089/cbr.2007.306.
    1. Kwekkeboom DJ, Bakker WH, Kooij PP, Konijnenberg MW, Srinivasan A, Erion JL, et al. [177Lu-DOTAOTyr3]octreotate: comparison with [111In-DTPAo]octreotide in patients. Eur J Nucl Med. 2001;28:1319–1325. doi: 10.1007/s002590100574.
    1. Cremonesi M, Ferrari M, Bodei L, Bartolomei M, Chinol M, Mei R, et al. Dosimetry in patients undergoing 177Lu-DOTATATE therapy with indications for 90Y- DOTATATE. Eur J Nucl Med Mol Imaging. 2006;33:S102. doi: 10.1007/s00259-006-0151-1.
    1. Sandström M, Garske U, Granberg D, Sundin A, Lundqvist H. Individualized dosimetry in patients undergoing therapy with 177Lu-DOTA-D-Phe1-Tyr3-octreotate. Eur J Nucl Med Mol Imaging. 2010;37:212–225. doi: 10.1007/s00259-009-1216-8.
    1. Rolleman EJ, Valkema R, de Jong M, Kooij PP, Krenning EP. Safe and effective inhibition of renal uptake of radiolabelled octreotide by a combination of lysine and arginine. Eur J Nucl Med Mol Imaging. 2003;30:9–15. doi: 10.1007/s00259-002-0982-3.
    1. de Keizer B, van Aken MO, Feelders RA, de Herder WW, Kam BL, van Essen M, et al. Hormonal crises following receptor radionuclide therapy with the radiolabeled somatostatin analogue [177Lu-DOTA0,Tyr3]octreotate. Eur J Nucl Med Mol Imaging. 2008;35:749–755. doi: 10.1007/s00259-007-0691-z.
    1. Valkema R, Pauwels SA, Kvols LK, Kwekkeboom DJ, Jamar F, de Jong M, et al. Long-term follow-up of renal function after peptide receptor radiation therapy with 90Y-DOTA0,Tyr3-octreotide and 177Lu-DOTA0,Tyr3-octreotate. J Nucl Med. 2005;46:83S–91S.
    1. Teunissen JJ, Krenning EP, de Jong FH, de Rijke YB, Feelders RA, van Aken MO, et al. Effects of therapy with [177Lu-DOTA0,Tyr3]octreotate on endocrine function. Eur J Nucl Med Mol Imaging. 2009;36:1758–1766. doi: 10.1007/s00259-009-1151-8.
    1. van Essen M, Krenning EP, Bakker WH, de Herder WW, van Aken MO, Kwekkeboom DJ. Peptide receptor radionuclide therapy with 177Lu-octreotate in patients with foregut carcinoid tumors of bronchial, gastric and thymic origin. Eur J Nucl Med Mol Imaging. 2007;34:1219–1227. doi: 10.1007/s00259-006-0355-4.
    1. Beutler D, Avoledo P, Reubi JC, Mäcke HR, Müller-Brand J, Merlo A, et al. Three-year recurrence-free survival in a patient with recurrent medulloblastoma after resection, high-dose chemotherapy, and intrathecal Yttrium-90-labeled DOTA0-D-Phe1-Tyr3-octreotide radiopeptide brachytherapy. Cancer. 2005;103:869–873. doi: 10.1002/cncr.20822.
    1. Villard L, Romer A, Marincek N, Brunner P, Koller MT, Schindler C, et al. Cohort study of somatostatin-based radiopeptide therapy with [(90)Y-DOTA]-TOC versus [(90)Y-DOTA]-TOC plus [(177)Lu-DOTA]-TOC in neuroendocrine cancers. J Clin Oncol. 2012;30:1100–1106. doi: 10.1200/JCO.2011.37.2151.
    1. Kunikowska J, Królicki L, Hubalewska-Dydejczyk A, Mikołajczak R, Sowa-Staszczak A, Pawlak D. Clinical results of radionuclide therapy of neuroendocrine tumours with 90Y-DOTATATE and tandem 90Y/177Lu-DOTATATE: which is a better therapy option? Eur J Nucl Med Mol Imaging. 2011;38:1788–1797. doi: 10.1007/s00259-011-1833-x.
    1. Arnold R, Chen YJ, Costa F, Falconi M, Gross D, Grossman AB, et al. ENETS Consensus Guidelines for the Standards of Care in Neuroendocrine Tumors: follow-up and documentation. Neuroendocrinology. 2009;90:227–233. doi: 10.1159/000225952.
    1. World Health Organization. WHO handbook for reporting results of cancer treatment. Geneva: World Health Organization; 1979. Available from:
    1. Green S, Weiss G. Southwest Oncology Group standard response criteria, endpoint, definitions and toxicity criteria. Invest New Drugs. 1992;10:239–253. doi: 10.1007/BF00944177.
    1. Van Persijn Van Meerten EL, Gelderblom H, Bloem J. RECIST revised: implications for the radiologist. A review article on the modified RECIST guideline. Eur Radiol. 2010;20:1456–1467. doi: 10.1007/s00330-009-1685-y.

Source: PubMed

3
구독하다