Skip to main content

Advertisement

Log in

In patients with well-differentiated neuroendocrine tumours, there is no apparent benefit of somatostatin analogues after disease control by peptide receptor radionuclide therapy

  • Original Article
  • Published:
European Journal of Nuclear Medicine and Molecular Imaging Aims and scope Submit manuscript

Abstract

Purpose

Peptide receptor radionuclide therapy (PRRT) and somatostatin analogues (SSAs) are commonly combined as primary treatment for neuroendocrine neoplasms (NEN), and SSAs given as maintenance. We sought to evaluate whether sequential therapy with PRRT followed by SSAs has progression or survival benefits in patients with NEN after disease control by PRRT.

Methods

This prospective, randomised, single-centre study had as principal eligibility criteria: unresectable, locally advanced, or metastatic, histologically confirmed well-differentiated NEN; no symptoms/biochemical diagnosis of carcinoid syndrome; no SSAs or ≤ 3 months of SSAs before PRRT; and stable disease or partial or complete response after PRRT. Altogether, 115 patients were randomised 2:1 to an SSA group (n = 74) given octreotide acetate LAR every 4 weeks, or a control group (n = 41) receiving only best supportive care. Octreotide treatment was to stop upon intolerable toxicity or patient refusal, or, at physician/patient discretion, upon NEN progression. The primary endpoint was progression-free survival (PFS), the secondary endpoint, and overall survival (OS).

Results

Median (25th–75th percentile) follow-up from the first PRRT activity to death or latest observation was 6.6 (3.18–10.22) years. During that time, 71/115 patients (62%) progressed, 52/74 (70%) in the SSA group, and 19/41 (46%) in the control group (p = 0.01). Eighty-eight/115 patients (76%) died, 58/74 (78%) in the SSA group, and 30/41 (73%) in the control group (p = 0.52). Median (95% CI) PFS was 4.7 (2.8–7.7) years in the SSA group, and 6.4 (4.1–not reached) years in controls. Overall, median OS was 6.6 years. Neither PFS nor OS differed between groups (p = 0.129, p = 0.985, respectively).

Conclusions

In patients with disease control after PRRT, subsequent SSA treatment appeared not to be associated with better PFS or OS. Whether to continue SSA administration upon progression after PRRT requires evaluation in a prospective, randomised, controlled multicentre study with a relatively homogeneous sample.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2

Similar content being viewed by others

References

  1. Reubi JC. Peptide receptor expression in GEP-NET [eng]. Virchows Arch. 2007;451 Suppl 1:S47–S50. https://doi.org/10.1007/s00428-007-0443-2.

    Article  PubMed  Google Scholar 

  2. Ahlman H, Wängberg B, Nilsson O, Grimelius L, Granérus G, Modlin IM, et al. Aspects on diagnosis and treatment of the foregut carcinoid syndrome [eng]. Scand J Gastroenterol. 1992;27:59–71. https://doi.org/10.3109/00365529209000106.

    Article  PubMed  Google Scholar 

  3. Oberg K, Ferone D, Kaltsas G, Knigge U-P, Taal B, Plöckinger U. ENETS consensus guidelines for the standards of care in neuroendocrine tumors: biotherapy [eng]. Neuroendocrinology. 2009;90:09–213. https://doi.org/10.1159/000183751.

    Article  PubMed  Google Scholar 

  4. Schally AV. Oncological applications of somatostatin analogues [eng]. Cancer Res. 1988;48(24 Pt 1):6977–85.

    CAS  PubMed  Google Scholar 

  5. Patel YC, Greenwood MT, Panetta R, Demchyshyn L, Niznik H, Srikant CB. The somatostatin receptor family [eng]. Life Sci. 1995;57:9–65. https://doi.org/10.1016/0024-3205(95)02082-t.

    Article  PubMed  Google Scholar 

  6. Delaunoit T, Rubin J, Neczyporenko F, Erlichman C, Hobday TJ. Somatostatin analogues in the treatment of gastroenteropancreatic neuroendocrine tumors [eng]. Mayo Clin Proc. 2005;80:502–6. https://doi.org/10.4065/80.4.502.

    Article  PubMed  Google Scholar 

  7. Eriksson B, Skogseid B, Lundqvist G, Wide L, Wilander E, Oberg K. Medical treatment and long-term survival in a prospective study of 84 patients with endocrine pancreatic tumors [eng]. Cancer. 1990;65:1883–90. https://doi.org/10.1002/1097-0142(19900501)65:9%3c1883::aid-cncr2820650902%3e3.0.co;2-3.

    Article  PubMed  Google Scholar 

  8. Aparicio T, Ducreux M, Baudin E, Sabourin JC, de Baere T, Mitry E, et al. Antitumour activity of somatostatin analogues in progressive metastatic neuroendocrine tumours [eng]. Eur J Cancer. 2001;37:1014–9. https://doi.org/10.1016/s0959-8049(01)00073-9.

    Article  PubMed  Google Scholar 

  9. Hejna M, Schmidinger M, Raderer M. The clinical role of somatostatin analogues as antineoplastic agents: much ado about nothing? Ann Oncol. 2002;13:653–68. https://doi.org/10.1093/annonc/mdf142.

    Article  CAS  PubMed  Google Scholar 

  10. Rinke A, Müller H-H, Schade-Brittinger C, Klose K-J, 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 [eng]. J Clin Oncol. 2009;27:4656–3. https://doi.org/10.1200/JCO.2009.22.8510.

    Article  CAS  PubMed  Google Scholar 

  11. Caplin ME, Pavel M, Ćwikła JB, Phan AT, Raderer M, Sedláčková E, et al. Lanreotide in metastatic enteropancreatic neuroendocrine tumors [eng]. N Engl J Med. 2014;371:224–33. https://doi.org/10.1056/NEJMoa1316158.

    Article  PubMed  Google Scholar 

  12. de Jong M, Valkema R, Jamar F, Kvols LK, Kwekkeboom DJ, Breeman WAP, et al. Somatostatin receptor-targeted radionuclide therapy of tumors: preclinical and clinical findings [eng]. Semin Nucl Med. 2002;32:133–40. https://doi.org/10.1053/snuc.2002.31027.

    Article  PubMed  Google Scholar 

  13. Krenning EP, Kooij PP, Bakker WH, Breeman WA, Postema PT, Kwekkeboom DJ, et al. Radiotherapy with a radiolabeled somatostatin analogue, [111In-DTPA-D-Phe1]-octreotide. A case history [eng]. Ann N Y Acad Sci. 1994;733:496–506. https://doi.org/10.1111/j.1749-6632.1994.tb17300.x.

  14. Paganelli G, Bodei L, Handkiewicz Junak D, Rocca P, Papi S, Lopera Sierra M, et al. 90Y-DOTA-D-Phe1-Try3-octreotide in therapy of neuroendocrine malignancies [eng]. Biopolymers. 2002;66:393–8. https://doi.org/10.1002/bip.10349.

    Article  PubMed  Google Scholar 

  15. Strosberg J, El-Haddad G, Wolin E, Hendifar A, Yao J, Chasen B, et al. Phase 3 trial of 177Lu-Dotatate for midgut neuroendocrine tumors [eng]. N Engl J Med. 2017;376:125–35. https://doi.org/10.1056/NEJMoa1607427.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Strosberg JR, Caplin ME, Kunz PL, Ruszniewski PB, Bodei L, Hendifar AE, et al. Final overall survival in the phase 3 NETTER-1 study of lutetium-177-DOTATATE in patients with midgut neuroendocrine tumors. J Clin Oncol. 2021;39:4112. https://doi.org/10.1200/JCO.2021.39.15_suppl.4112.

    Article  Google Scholar 

  17. Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer. 2009;45:228–47. https://doi.org/10.1016/j.ejca.2008.10.026.

  18. Bison SM, Haeck JC, Bol K, Koelewijn SJ, Groen HC, Melis M, et al. Optimization of combined temozolomide and peptide receptor radionuclide therapy (PRRT) in mice after multimodality molecular imaging studies [eng]. EJNMMI Res. 2015;5:62. https://doi.org/10.1186/s13550-015-0142-y.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Claringbold PG, Brayshaw PA, Price RA, Turner JH. Phase II study of radiopeptide 177Lu-octreotate and capecitabine therapy of progressive disseminated neuroendocrine tumours [eng]. Eur J Nucl Med Mol Imaging. 2011;38:302–11. https://doi.org/10.1007/s00259-010-1631-x.

    Article  PubMed  Google Scholar 

  20. Claringbold PG, Price RA, Turner JH. Phase I-II study of radiopeptide 177Lu-octreotate in combination with capecitabine and temozolomide in advanced low-grade neuroendocrine tumors [eng]. Cancer Biother Radiopharm. 2012;27561–9. https://doi.org/10.1089/cbr.2012.1276.

    Article  PubMed  Google Scholar 

  21. Yordanova A, Ahrens H, Feldmann G, Brossart P, Gaertner FC, Fottner C, et al. Peptide receptor radionuclide therapy combined with chemotherapy in patients with neuroendocrine tumors [eng]. Clin Nucl Med. 2019;44:e329–e335. https://doi.org/10.1097/RLU.0000000000002532.

    Article  PubMed  Google Scholar 

  22. Purohit NK, Shah RG, Adant S, Hoepfner M, Shah GM, Beauregard J-M. Potentiation of 177Lu-octreotate peptide receptor radionuclide therapy of human neuroendocrine tumor cells by PARP inhibitor [eng]. Oncotarget. 2018;9:24694–706. https://doi.org/10.18632/oncotarget.25266.

  23. Cullinane C, Waldeck K, Kirby L, Rogers BE, Eu P, Tothill RW, et al. Enhancing the anti-tumour activity of (177)Lu-DOTA-octreotate radionuclide therapy in somatostatin receptor-2 expressing tumour models by targeting PARP [eng]. Sci Rep. 2020;10:10169. https://doi.org/10.1038/s41598-020-67199-9.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Pignata SA, Ferrantelli A, Baldari S. Peptide receptor radionuclide therapy plus somatostatin analogues for a neuroendocrine tumour combined and maintenance treatment. Clinical and Translational Imaging. 2019;7(5):373–6.

    Article  Google Scholar 

  25. Bodei L, Schöder H, Baum RP, Herrmann K, Strosberg J, Caplin M, et al. Molecular profiling of neuroendocrine tumours to predict response and toxicity to peptide receptor radionuclide therapy [eng]. Lancet Oncol. 2020;21:e431–e443. https://doi.org/10.1016/S1470-2045(20)30323-5.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Aalbersberg EA, de Wit-van der Veen, B J, Versleijen MWJ, Saveur LJ, Valk GD, Tesselaar MET, et al. Influence of lanreotide on uptake of 68Ga-DOTATATE in patients with neuroendocrine tumours: a prospective intra-patient evaluation [eng]. Eur J Nucl Med Mol Imaging. 2019;46:696–703. https://doi.org/10.1007/s00259-018-4117-x.

  27. Bodei L, Mueller-Brand J, Baum RP, Pavel ME, Hörsch D, O’Dorisio MS, et al. The joint IAEA, EANM, and SNMMI practical guidance on peptide receptor radionuclide therapy (PRRNT) in neuroendocrine tumours [eng]. Eur J Nucl Med Mol Imaging. 2013;40:800–16. https://doi.org/10.1007/s00259-012-2330-6.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Plöckinger U, Wiedenmann B. Neuroendocrine tumors. Biotherapy [eng]. Best Pract Res Clin Endocrinol Metab. 2007;21:45–62. https://doi.org/10.1016/j.beem.2007.01.002.

  29. Toumpanakis C, Garland J, Marelli L, Srirajaskanthan R, Soh J, Davies P, et al. Long-term results of patients with malignant carcinoid syndrome receiving octreotide LAR [eng]. Aliment Pharmacol Ther. 2009;30:733–40. https://doi.org/10.1111/j.1365-2036.2009.04083.x.

    Article  PubMed  Google Scholar 

  30. Stueven AK, Kayser A, Wetz C, Amthauer H, Wree A, Tacke F, et al. Somatostatin analogues in the treatment of neuroendocrine tumors: past, present and future [eng]. Int J Mol Sci. 2019;20. https://doi.org/10.3390/ijms20123049.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Pavel ME, Hainsworth JD, Baudin E, Peeters M, Hörsch D, Winkler RE, 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 [eng]. Lancet. 2011;378:2005–12. https://doi.org/10.1016/S0140-6736(11)61742-X.

    Article  PubMed  Google Scholar 

  32. Gatto F, Hofland LJ. The role of somatostatin and dopamine D2 receptors in endocrine tumors [eng]. Endocr Relat Cancer. 2011;18:R223–51. https://doi.org/10.1530/ERC-10-0334.

    Article  PubMed  Google Scholar 

  33. Oh S, Prasad V, Lee DS, Baum RP, Chen X. Effect of peptide receptor radionuclide therapy on somatostatin receptor status and glucose metabolism in neuroendocrine tumors: intraindividual comparison of Ga-68 DOTANOC PET/CT and F-18 FDG PET/CT. Int J Mol Imaging. 2011;2011524130. https://doi.org/10.1155/2011/524130.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Feijtel D, Doeswijk GN, Verkaik NS, Haeck JC, Chicco D, Angotti C, et al. Inter and intra-tumor somatostatin receptor 2 heterogeneity influences peptide receptor radionuclide therapy response [eng]. Theranostics. 2021;11:491–505. https://doi.org/10.7150/thno.51215.

    Article  PubMed  PubMed Central  Google Scholar 

  35. Sowa-Staszczak A, Stefanska A, Chrapczynski P, Trofimiuk-Müldner M, Szura M, Hubalewska-Dydejczyk A. Does combination of “cold” and “hot” somatostatin analogs prolong survival of patients with neuroendocrine neoplasms? [eng]. Endocr J. 2017;64:171–7. https://doi.org/10.1507/endocrj.EJ16-0219.

    Article  PubMed  Google Scholar 

  36. Yordanova A, Wicharz MM, Mayer K, Brossart P, Gonzalez-Carmona MA, Strassburg CP, et al. The role of adding somatostatin analogues to peptide receptor radionuclide therapy as a combination and maintenance therapy [eng]. Clin Cancer Res. 2018;24:4672–79. https://doi.org/10.1158/1078-0432.CCR-18-0947.

    Article  PubMed  Google Scholar 

  37. Kvols LK, Reubi JC. Metastatic carcinoid tumors and the malignant carcinoid syndrome. Acta Oncol. 1993;32:197–201. https://doi.org/10.3109/02841869309083912.

    Article  PubMed  Google Scholar 

  38. Ruszniewski P, Ducreux M, Chayvialle JA, Blumberg J, Cloarec D, Michel H, et al. Treatment of the carcinoid syndrome with the longacting somatostatin analogue lanreotide: a prospective study in 39 patients. Gut. 1996;39:279. https://doi.org/10.1136/gut.39.2.279.

    Article  PubMed  PubMed Central  Google Scholar 

  39. Caplin ME, Pavel M, Ćwikła JB, Phan AT, Raderer M, Sedláčková E, et al. Anti-tumour effects of lanreotide for pancreatic and intestinal neuroendocrine tumours: the CLARINET open-label extension study [eng]. Endocr Relat Cancer. 2016;23:191–9. https://doi.org/10.1530/ERC-15-0490.

    Article  PubMed  PubMed Central  Google Scholar 

  40. Sullivan I, Le Teuff G, Guigay J, Caramella C, Berdelou A, Leboulleux S, et al. Antitumour activity of somatostatin analogues in sporadic, progressive, metastatic pulmonary carcinoids [eng]. Eur J Cancer. 2017;75:59–67. https://doi.org/10.1016/j.ejca.2016.11.034.

    Article  PubMed  Google Scholar 

  41. Lenotti E, Alberti A, Spada F, Amoroso V, Maisonneuve P, Grisanti S, et al. Outcome of patients with metastatic lung neuroendocrine tumors submitted to first line monotherapy with somatostatin analogs [eng]. Front Endocrinol (Lausanne). 2021;12:669484. https://doi.org/10.3389/fendo.2021.669484.

    Article  Google Scholar 

  42. Kulke MH, Ruszniewski P, van Cutsem E, Lombard-Bohas C, Valle JW, de Herder WW, et al. A randomized, open-label, phase 2 study of everolimus in combination with pasireotide LAR or everolimus alone in advanced, well-differentiated, progressive pancreatic neuroendocrine tumors: COOPERATE-2 trial [eng]. Ann Oncol. 2017;28:1309–15. https://doi.org/10.1093/annonc/mdx078.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Ferolla P, Brizzi MP, Meyer T, Mansoor W, Mazieres J, Do Cao C, et al. Efficacy and safety of long-acting pasireotide or everolimus alone or in combination in patients with advanced carcinoids of the lung and thymus (LUNA): an open-label, multicentre, randomised, phase 2 trial [eng]. Lancet Oncol. 2017;18:1652–64. https://doi.org/10.1016/S1470-2045(17)30681-2.

    Article  PubMed  Google Scholar 

  44. Dasari A, Shen C, Halperin D, Zhao B, Zhou S, Xu Y, et al. Trends in the incidence, prevalence, and survival outcomes in patients with neuroendocrine tumors in the United States [eng]. JAMA Oncol. 2017;3:1335–42. https://doi.org/10.1001/jamaoncol.2017.0589.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Levy S, van Veenendaal LM, Korse CM, Breekveldt ECH, Verbeek WHM, Vriens MR, et al. Survival in patients with neuroendocrine tumours of the small intestine: nomogram validation and predictors of survival [eng]. J Clin Med. 2020;9. https://doi.org/10.3390/jcm9082502.

    Article  PubMed  PubMed Central  Google Scholar 

  46. Chen L, Gnanasegaran G, Mandair D, Toumpanakis C, Caplin M, Navalkissoor S. Prognostic stratification for patients with neuroendocrine tumours receiving 177Lu-Dotatate [eng]. Endocr Relat Cancer. 2022;29:111–20. https://doi.org/10.1530/ERC-21-0248.

    Article  PubMed  Google Scholar 

  47. Marotta V, Zatelli MC, Sciammarella C, Ambrosio MR, Bondanelli M, Colao A, et al. Chromogranin A as circulating marker for diagnosis and management of neuroendocrine neoplasms: more flaws than fame [eng]. Endocr Relat Cancer. 2018;25:R11–R29. https://doi.org/10.1530/ERC-17-0269.

    Article  PubMed  Google Scholar 

  48. Bodei L, Kidd MS, Singh A, van der Zwan, Wouter A, Severi S, Drozdov IA, et al. PRRT neuroendocrine tumor response monitored using circulating transcript analysis: the NETest [eng]. Eur J Nucl Med Mol Imaging. 2020;47:895–906. https://doi.org/10.1007/s00259-019-04601-3.

  49. Pavel M, O’Toole D, Costa F, Capdevila J, Gross D, Kianmanesh R, et al. ENETS consensus guidelines update for the management of distant metastatic disease of intestinal, pancreatic, bronchial neuroendocrine neoplasms (NEN) and NEN of unknown primary site [eng]. Neuroendocrinology. 2016;103:172–85. https://doi.org/10.1159/000443167.

    Article  PubMed  Google Scholar 

  50. Pavel M, Öberg K, Falconi M, Krenning EP, Sundin A, Perren A, et al. Gastroenteropancreatic neuroendocrine neoplasms: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up [eng]. Ann Oncol. 2020;31:844–60. https://doi.org/10.1016/j.annonc.2020.03.304.

    Article  PubMed  Google Scholar 

  51. Klöppel G, Couvelard A, Perren A, Komminoth P, McNicol A-M, Nilsson O, et al. ENETS consensus guidelines for the standards of care in neuroendocrine tumors: towards a standardized approach to the diagnosis of gastroenteropancreatic neuroendocrine tumors and their prognostic stratification [eng]. Neuroendocrinology. 2009;90:162–6. https://doi.org/10.1159/000182196.

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

Robert J. Marlowe, Spencer-Fontayne Corporation, Jersey City, NJ, USA, edited this manuscript.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Daria Handkiewicz-Junak.

Ethics declarations

Conflict of interest

BJ received travel grants and speaker honoraria from Ipsen, Novartis; DH-J received travel grants and speaker honoraria from Ipsen, Novartis, and Teva; KHL and BJ-L received travel grants from Ipsen; other authors have nothing to declare.

Ethics approval

All procedures performed in studies involving human participants were in accordance with the ethical standards of the institutional and/or national research committee and with the 1964 Helsinki declaration and its later amendments or comparable ethical standards. The study protocol was approved by the institutional Ethics Committee.

Additional information

Publisher's note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

This article is part of the Topical Collection on Theragnostic.

Supplementary Information

Below is the link to the electronic supplementary material.

Supplementary file1 (PPTX 48 KB)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Syguła, A., Ledwon, A., Hasse-Lazar, K. et al. In patients with well-differentiated neuroendocrine tumours, there is no apparent benefit of somatostatin analogues after disease control by peptide receptor radionuclide therapy. Eur J Nucl Med Mol Imaging 49, 3841–3851 (2022). https://doi.org/10.1007/s00259-022-05792-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00259-022-05792-y

Keywords

Navigation