79
Views
3
CrossRef citations to date
0
Altmetric
Review

Spotlight on telotristat ethyl for the treatment of carcinoid syndrome diarrhea: patient selection and reported outcomes

, , , &
Pages 7537-7556 | Published online: 08 Aug 2019

References

  • Klimstra David SYZ. Pathology, classification, and grading of neuroendocrine neoplasms arising in the digestive system In: Goldberg Richard MSDM, editor. Waltham, MA: UpToDate; 2018.
  • Dasari A, Shen C, Halperin D, et al. Trends in the incidence, prevalence, and survival outcomes in patients with neuroendocrine tumors in the United States. JAMA Oncol. 2017;3(10):1335–1342. doi:10.1001/jamaoncol.2017.058928448665
  • Ohike NAN. Mixed neuroendocrine-non-neuroendocrine neoplasms In: Lloyd RVOR, editor. WHO classification of tumours of endocrine organs. 4th ed. Lyon: IARC Press; 2017:238.
  • Vinik AI, Woltering EA, Warner RRP, et al. NANETS consensus guidelines for the diagnosis of neuroendocrine tumor. Pancreas. 2010;39(6):713–734. doi:10.1097/MPA.0b013e3181ebaffd20664471
  • Halperin DM, Shen C, Dasari A, et al. The frequency of carcinoid syndrome at neuroendocrine tumor diagnosis: a large population-based study using SEER-medicare data. Lancet Oncol. 2017;18(4):525–534. doi:10.1016/S1470-2045(17)30072-428238592
  • Lamarca A, Barriuso J, McNamara MG, Hubner RA, Valle JW. Telotristat ethyl: a new option for the management of carcinoid syndrome. Expert Opin Pharmacother. 2016;17(18):2487–2498. doi:10.1517/14656566.2016.112940227817224
  • Boutzios G, Kaltsas G. Clinical syndromes related to gastrointestinal neuroendocrine neoplasms. Front Horm Res. 2015;44:40–57.26303703
  • Druce M, Rockall A, Grossman AB. Fibrosis and carcinoid syndrome: from causation to future therapy. Nat Rev Endocrinol. 2009;5(5):276–283. doi:10.1038/nrendo.2009.5119444261
  • Mota JM, Sousa LG, Riechelmann RP. Complications from carcinoid syndrome: review of the current evidence. Ecancermedicalscience. 2016;10:662. doi:10.3332/ecancer.2016.66227594907
  • Zacks J, Lavine R, Ratner L, Warner R. Telotristat etiprate appears to halt carcinoid heart disease. Neuroendocrinology. 103(suppl 1):90.
  • Dobson R, Burgess MI, Banks M, et al. The Association of a Panel of Biomarkers with the Presence and Severity of Carcinoid Heart Disease: a cross-sectional study Holvoet P, editor PLoS One. 2013; 8(9):e73679. doi:10.1371/journal.pone.007367924069222
  • Moller JE, Connolly HM, Rubin J, Seward JB, Modesto K, Pellikka PA. Factors associated with progression of carcinoid heart disease. N Engl J Med. 2003;348(11):1005–1015. doi:10.1056/NEJMicm02003712637610
  • Connolly HM, Crary JL, McGoon MD, et al. Valvular heart disease associated with fenfluramine-phentermine. N Engl J Med. 1997;337(9):581–588. doi:10.1056/NEJM1997082833709019271479
  • Fox DJ, Khattar RS. Carcinoid heart disease: presentation, diagnosis, and management. Heart. 2004;90(10):1224–1228. doi:10.1136/hrt.2004.04032915367531
  • Dobson R, Burgess MI, Valle JW, et al. Serial surveillance of carcinoid heart disease: factors associated with echocardiographic progression and mortality. Br J Cancer. 2014;111(9):1703–1709. doi:10.1038/bjc.2014.34825211656
  • Plockinger U, Gustafsson B, Ivan D, Szpak W, Davar J. ENETS consensus guidelines for the standards of care in neuroendocrine tumors: echocardiography. Neuroendocrinology. 2009;90(2):190–193. doi:10.1159/00022594719713710
  • Beaumont JL, Cella D, Phan AT, Choi S, Liu Z, Yao JC. Comparison of health-related quality of life in patients with neuroendocrine tumors with quality of life in the general US population. Pancreas. 2012;41(3):461–466. doi:10.1097/MPA.0b013e318232804522422138
  • Haugland T, Vatn MH, Veenstra M, Wahl AK, Natvig GK. Health related quality of life in patients with neuroendocrine tumors compared with the general Norwegian population. Qual Life Res. 2009;18(6):719–726. doi:10.1007/s11136-009-9478-y19479341
  • Frojd C, Larsson G, Lampic C, von Essen L. Health related quality of life and psychosocial function among patients with carcinoid tumours. A longitudinal, prospective, and comparative study. Health Qual Life Outcomes. 2007;11(5):18. doi:10.1186/1477-7525-5-18
  • Lamarca A, McCallum L, Nuttall C, et al. Somatostatin analogue-induced pancreatic exocrine insufficiency in patients with neuroendocrine tumors: results of a prospective observational study. Expert Rev Gastroenterol Hepatol. 2018;12(7):723–731. doi:10.1080/17474124.2018.148923229923433
  • Pearman TP, Beaumont JL, Cella D, Neary MP, Yao J. Health-related quality of life in patients with neuroendocrine tumors: an investigation of treatment type, disease status, and symptom burden. Support Care Cancer. 2016;24(9):3695–3703. doi:10.1007/s00520-016-3189-z27029477
  • Singh S, Granberg D, Wolin E, et al. Patient-reported burden of a neuroendocrine tumor (NET) diagnosis: results from the first global survey of patients with NETs. J Glob Oncol. 2017;3(1):43–53. doi:10.1200/JGO.2015.00298028717741
  • Wolin EM, Leyden J, Goldstein G, Kolarova T, Hollander R, Warner RRP. Patient-reported experience of diagnosis, management, and burden of neuroendocrine tumors: results from a large patient survey in the United States. Pancreas. 2017;46(5):639–647. doi:10.1097/MPA.000000000000081828328615
  • Wymenga AN, de Vries EG, Leijsma MK, Kema IP, Kleibeuker JH. Effects of ondansetron on gastrointestinal symptoms in carcinoid syndrome. Eur J Cancer. 1998;34(8):1293–1294. doi:10.1016/S0959-8049(98)00009-49849494
  • Kiesewetter B, Duan H, Lamm W, et al. Oral ondansetron offers effective antidiarrheal activity for carcinoid syndrome refractory to somatostatin analogs. Oncologist. 2019;24(2):255–258. doi:10.1634/theoncologist.2018-019130171068
  • Niederle B, Pape U-F, Costa F, et al. ENETS consensus guidelines update for neuroendocrine neoplasms of the Jejunum and Ileum. Neuroendocrinology. 2016;103(2):125–138. doi:10.1159/00044317026758972
  • Vinik AI, Wolin EM, Liyanage N, Gomez-Panzani E, GA F; ELECT Study Group. Evaluation of lanreotide Depot/autogel Efficacy And Safety As A Carcinoid Syndrome Treatment (ELECT): a randomized, double-blind, placebo-controlled trial. Endocr Pract. 2016;22(9):1068–1080. doi:10.4158/1934-2403-22.12.e127214300
  • Fisher GA, Wolin EM, Liyanage N, et al. Lanreotide therapy in carcinoid syndrome: prospective analysis of patient-reported symptoms in patients responsive to prior octreotide therapy and patients naïve to somatostatin analogue therapy in the elect phase 3 study. Endocr Pract. 2018;24(3):243–255. doi:10.4158/EP172000.OR29547049
  • Modlin IM, Pavel M, Kidd M, Gustafsson BI. Review article: somatostatin analogues in the treatment of gastroenteropancreatic neuroendocrine (carcinoid) tumours. Aliment Pharmacol Ther. 2010;31(2):169–188.19845567
  • Ricci S, Antonuzzo A, Galli L, et al. Long-acting depot lanreotide in the treatment of patients with advanced neuroendocrine tumors. Am J Clin Oncol. 2000;23(4):412–415. doi:10.1097/00000421-200008000-0002010955874
  • Kvols LK, Moertel CG, O’Connell MJ, Schutt AJ, Rubin J, Hahn RG. Treatment of the malignant carcinoid syndrome. Evaluation of a long-acting somatostatin analogue. N Engl J Med. 1986;315(11):663–666. doi:10.1056/NEJM1986091131511022427948
  • Wymenga AN, Eriksson B, Salmela PI, et al. Efficacy and safety of prolonged-release lanreotide in patients with gastrointestinal neuroendocrine tumors and hormone-related symptoms. J Clin Oncol. 1999;17(4):1111. doi:10.1200/JCO.1999.17.4.111110561168
  • Hofland LJ, Lamberts SW. The pathophysiological consequences of somatostatin receptor internalization and resistance. Endocr Rev. 2003;24(1):28–47. doi:10.1210/er.2000-000112588807
  • Carmona-Bayonas A, Jimenez-Fonseca P, Custodio A, et al. Optimizing somatostatin analog use in well or moderately differentiated gastroenteropancreatic neuroendocrine tumors. Curr Oncol Rep. 2017;19(11):72. doi:10.1007/s11912-017-0633-228920153
  • Molina-Cerrillo J, Alonso-Gordoa T, Martinez-Saez O, Grande E. Inhibition of peripheral synthesis of serotonin as a new target in neuroendocrine tumors. Oncologist. 2016;21(6):701–707. doi:10.1634/theoncologist.2015-033627107003
  • Cordoba-Chacon J, Gahete MD, Duran-Prado M, et al. Identification and characterization of new functional truncated variants of somatostatin receptor subtype 5 in rodents. Cell Mol Life Sci. 2010;67(7):1147–1163. doi:10.1007/s00018-010-0401-z20063038
  • Rinke A, Muller HH, Schade-Brittinger C, 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(28):4656–4663. doi:10.1200/JCO.2009.22.851019704057
  • Caplin ME, Pavel M, Cwikla JB, et al. Lanreotide in metastatic enteropancreatic neuroendocrine tumors. N Engl J Med. 2014;371(3):224–233. doi:10.1056/NEJMoa141049025014687
  • Strosberg JR, Benson AB, Huynh L, et al. Clinical benefits of above-standard dose of octreotide LAR in patients with neuroendocrine tumors for control of carcinoid syndrome symptoms: a multicenter retrospective chart review study. Oncologist. 2014;19(9):930–936. doi:10.1634/theoncologist.2014-012025096997
  • Kvols LK, Oberg KE, O’Dorisio TM, et al. Pasireotide (SOM230) shows efficacy and tolerability in the treatment of patients with advanced neuroendocrine tumors refractory or resistant to octreotide LAR: results from a phase II study. Endocr Relat Cancer. 2012;19(5):657–666. doi:10.1530/ERC-11-036222807497
  • Wolin EM, Jarzab B, Eriksson B, et al. Phase III study of pasireotide long-acting release in patients with metastatic neuroendocrine tumors and carcinoid symptoms refractory to available somatostatin analogues. Drug Des Devel Ther. 2015;9:5075–5086. doi:10.2147/DDDT.S84177
  • Ferolla P, Brizzi MP, Meyer T, 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. Lancet Oncol. 2017;18(12):1652–1664. doi:10.1016/S1470-2045(17)30072-429074099
  • Kulke MH, Ruszniewski P, Van Cutsem E, 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. Ann Oncol. 2017;28(6):1309–1315. doi:10.1093/annonc/mdx07528327907
  • Oberg K, Funa K, Alm G. Effects of leukocyte interferon on clinical symptoms and hormone levels in patients with mid-gut carcinoid tumors and carcinoid syndrome. N Engl J Med. 1983;309(3):129–133. doi:10.1056/NEJM1983072130903016191217
  • Smith DB, Scarffe JH, Wagstaff J, Johnston RJ. Phase II trial of rDNA alfa 2b interferon in patients with malignant carcinoid tumor. Cancer Treat Rep. 1987;71(12):1265–1266.3690534
  • Moertel CG, Rubin J, Kvols LK. Therapy of metastatic carcinoid tumor and the malignant carcinoid syndrome with recombinant leukocyte A interferon. J Clin Oncol. 1989;7(7):865–868. doi:10.1200/JCO.1989.7.7.8652738623
  • Valimaki M, Jarvinen H, Salmela P, Sane T, Sjoblom SM, Pelkonen R. Is the treatment of metastatic carcinoid tumor with interferon not as successful as suggested? Cancer. 1991;67(3):547–549. doi:10.1002/1097-0142(19910101)67:1<126::aid-cncr2820670123>3.0.co;2-71702352
  • Veenhof CH, de Wit R, Taal BG, et al. A dose-escalation study of recombinant interferon-alpha in patients with a metastatic carcinoid tumour. Eur J Cancer. 1992;28(1):75–78. doi:10.1016/0959-8049(92)90389-J1373635
  • Janson ET, Oberg K. Long-term management of the carcinoid syndrome. Treatment with octreotide alone and in combination with alpha-interferon. Acta Oncol. 1993;32(2):225–229. doi:10.3109/028418693090839167686765
  • Di Bartolomeo M, Bajetta E, Zilembo N, et al. Treatment of carcinoid syndrome with recombinant interferon alpha-2a. Acta Oncol. 1993;32(2):235–238. doi:10.3109/028418693090839187686766
  • Yao JC, Guthrie KA, Moran C, et al. Phase III prospective randomized comparison trial of depot octreotide plus interferon alfa-2b versus depot octreotide plus bevacizumab in patients with advanced carcinoid tumors: SWOG S0518. J Clin Oncol. 2017;35(15):1695–1703. doi:10.1200/JCO.2016.70.407228384065
  • Pavel ME, Hainsworth JD, Baudin E, et al. Everolimus plus octreotide long-acting repeatable for the treatment of advanced neuroendocrine tumours associated with carcinoid syndrome (RADIANT-2): a randomised, placebo-controlled, phase 3 study. Lancet (London, England). 2011;378(9808):2005–2012. doi:10.1016/S0140-6736(11)60984-7
  • Yao JC, Shah MH, Ito T, et al. Everolimus for advanced pancreatic neuroendocrine tumors. N Engl J Med. 2011;364(6):514–523. doi:10.1056/NEJMoa100799421306238
  • Yao JC, Fazio N, Singh S, et al. Everolimus for the treatment of advanced, non-functional neuroendocrine tumours of the lung or gastrointestinal tract (RADIANT-4): a randomised, placebo-controlled, phase 3 study. Lancet (London, England). 2016;387(10022):968–977. doi:10.1016/S0140-6736(15)00817-X
  • Pavel M, Unger N, Borbath I, et al. Safety and QOL in patients with advanced NET in a phase 3b expanded access study of everolimus. Target Oncol. 2016;11(5):667–675. doi:10.1007/s11523-016-0440-y27193465
  • Bainbridge HE, Larbi E, Middleton G. Symptomatic control of neuroendocrine tumours with everolimus. Horm Cancer. 2015;6(5–6):254–259. doi:10.1007/s12672-015-0233-226245686
  • Raymond E, Dahan L, Raoul JL, et al. Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. N Engl J Med. 2011;364(6):501–513. doi:10.1056/NEJMoa100799421306237
  • 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(8):519–523. doi:10.1056/NEJM1992022032608041310159
  • 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(23):4762–4771. doi:10.1200/JCO.2004.08.04015570077
  • Strosberg JR, Fine RL, Choi J, et al. First-line chemotherapy with capecitabine and temozolomide in patients with metastatic pancreatic endocrine carcinomas. Cancer. 2011;117(2):268–275. doi:10.1002/cncr.2542520824724
  • Meyer T, Qian W, Caplin ME, et al. Capecitabine and streptozocin ± cisplatin in advanced gastroenteropancreatic neuroendocrine tumours. Eur J Cancer. 2014;50(5):902–911. doi:10.1016/j.ejca.2013.12.01124445147
  • Fine RL, Gulati AP, Krantz BA, et al. Capecitabine and temozolomide (CAPTEM) for metastatic, well-differentiated neuroendocrine cancers: the Pancreas Center at Columbia University experience. Cancer Chemother Pharmacol. 2013;71(3):663–670. doi:10.1007/s00280-012-2055-z23370660
  • Lamarca A, Elliott E, Barriuso J, et al. Chemotherapy for advanced non-pancreatic well-differentiated neuroendocrine tumours of the gastrointestinal tract, a systematic review and meta-analysis: a lost cause? Cancer Treat Rev. 2016;44:26–41. doi:10.1016/j.ctrv.2016.01.00526855376
  • Strosberg J, El-Haddad G, Wolin E, et al. Phase 3 trial of (177)lu-dotatate for midgut neuroendocrine tumors. N Engl J Med. 2017;376(2):125–135. doi:10.1056/NEJMoa160742728076709
  • Strosberg J, Wolin E, Chasen B, et al. Health-related quality of life in patients with progressive midgut neuroendocrine tumors treated with (177)Lu-dotatate in the phase III NETTER-1 trial. J Clin Oncol. 2018;36(25):2578–2584. doi:10.1200/JCO.2018.78.586529878866
  • Matthes S, Bader M. Peripheral serotonin synthesis as a new drug target. Trends Pharmacol Sci. 2018;39(6):560–572. doi:10.1016/j.tips.2018.03.00429628275
  • Fidalgo S, Ivanov DK, Wood SH. Serotonin: from top to bottom. Biogerontology. 2013;14(1):21–45. doi:10.1007/s10522-012-9406-323100172
  • Walther DJ, Peter JU, Bashammakh S, et al. Synthesis of serotonin by a second tryptophan hydroxylase isoform. Science (New York, NY). 2003;299(5603):76. doi:10.1126/science.1078197
  • Adaway J, Dobson R, Walsh J, et al. Serum 5HIAA: a better biomarker than urine for detecting and monitoring neuroendocrine tumours? BioScientifica. 2014 [cited January 16, 2019] Available from: https://www.endocrine-abstracts.org/ea/0034/ea0034p45. Accessed June 24, 2019.
  • European Medicines Agency. Xermelo [Internet]. 2017 [cited 928, 2018]. Available from: https://www.ema.europa.eu/documents/assessment-report/xermelo-epar-public-assessment-report_en.pdf. Accessed 624, 2019.
  • Margolis KG, Stevanovic K, Li Z, et al. Pharmacological reduction of mucosal but not neuronal serotonin opposes inflammation in mouse intestine. Gut. 2014;63(6):928–937. doi:10.1136/gutjnl-2013-30490123749550
  • Engelman K, Lovenberg W, Sjoerdsma A. Inhibition of serotonin synthesis by para-chlorophenylalanine in patients with the carcinoid syndrome. N Engl J Med. 1967;277(21):1103–1108. doi:10.1056/NEJM1967112327721016054996
  • A Phase 1, Randomized, Double-blind, Placebo-controlled, Ascending Single Dose Study to Determine the Safety and Tolerability of Orally Administered LX1606 in Healthy Human Subjects. The Woodlands, TX: Lexicon Pharmaceuticals, Inc.; 2008.
  • Lapuerta P, Zambrowicz B, Fleming D, Wheeler D, Arthur S. Telotristat etiprate, a novel inhibitor of serotonin synthesis for the treatment of carcinoid syndrome. Clin Invest (Lond). 2015;5:447–456. doi:10.4155/cli.15.10
  • Pavel M, Horsch D, Caplin M, et al. Telotristat etiprate for carcinoid syndrome: a single-arm, multicenter trial. J Clin Endocrinol Metab. 2015;100(4):1511–1519. doi:10.1210/jc.2014-394125636046
  • Kulke MH, O’Dorisio T, Phan A, et al. Telotristat etiprate, a novel serotonin synthesis inhibitor, in patients with carcinoid syndrome and diarrhea not adequately controlled by octreotide. Endocr Relat Cancer. 2014;21(5):705–714. doi:10.1530/ERC-14-017325012985
  • Kulke MH, Horsch D, Caplin ME, et al. Telotristat ethyl, a tryptophan hydroxylase inhibitor for the treatment of carcinoid syndrome. J Clin Oncol. 2017;35(1):14–23. doi:10.1200/JCO.2016.69.278027918724
  • Pavel M, Gross DJ, Benavent M, et al. Telotristat ethyl in carcinoid syndrome: safety and efficacy in the TELECAST phase 3 trial. Endocr Relat Cancer. 2018;25(3):309–322. doi:10.1530/ERC-17-045529330194
  • Hörsch D, Rocio Garcia-Carbonero JW, Valle PP, et al. Efficacy and safety of telotristat ethyl in combination with lanreotide in patients with a neuroendocrine tumour and carcinoid syndrome diarrhoea: meta-analysis of phase 3 double-blind placebo-controlled TELESTAR and TELECAST studies. Ann Oncol. 2018;29(suppl_8):viii467–78. doi:10.1093/annonc/mdx807
  • Markham A. Telotristat ethyl: first global approval. Drugs. 2017;77(7):793–798. doi:10.1007/s40265-017-0731-328382568
  • Oates JA, Melmon K, Sjoerdsma A, Gillespie L, Mason DT. Release of a kinin peptide in the carcinoid syndrome. Lancet. 1964;1(7332):514–517. doi:10.1016/S0140-6736(64)92907-114100182
  • Norheim I, Theodorsson-Norheim E, Brodin E, Oberg K. Tachykinins in carcinoid tumors: their use as a tumor marker and possible role in the carcinoid flush. J Clin Endocrinol Metab. 1986;63(3):605–612. doi:10.1210/jcem-63-3-6052426299
  • Cunningham JL, Janson ET, Agarwal S, Grimelius L, Stridsberg M. Tachykinins in endocrine tumors and the carcinoid syndrome. Eur J Endocrinol. 2008;159(3):275–282. doi:10.1530/EJE-08-027118524798
  • Gelhorn HL, Kulke MH, O’Dorisio T, et al. Patient-reported symptom experiences in patients with carcinoid syndrome after participation in a study of telotristat etiprate: a qualitative interview approach. Clin Ther. 2016;38(4):759–768. doi:10.1016/j.clinthera.2016.03.00227041406
  • Anthony L, Ervin C, Lapuerta P, et al. Understanding the patient experience with carcinoid syndrome: exit interviews from a randomized, placebo-controlled study of telotristat ethyl. Clin Ther. 2017;39(11):2158–2168. doi:10.1016/j.clinthera.2017.09.01329074312
  • Weickert MO, Kaltsas G, Horsch D, et al. Changes in weight associated with telotristat ethyl in the treatment of carcinoid syndrome. Clin Ther. 2018;40(6):952–962.e2. doi:10.1016/j.clinthera.2018.04.00629724499
  • Scott Neil W, Fayers Peter M, Aaronson Neil K, et al. EORTC QLQ-C30 Reference values [Internet]. 2008 [cited 1015, 2018]. Available from: http://www.eortc.org/app/uploads/sites/2/2018/02/reference_values_manual2008.pdf. Accessed 624, 2019.
  • Oberg K, Janson ET, Eriksson B. Tumour markers in neuroendocrine tumours. Ital J Gastroenterol Hepatol. 1999;31(Suppl 2):S160–S162.10604122
  • Eriksson B, Oberg K, Stridsberg M. Tumor markers in neuroendocrine tumors. Digestion. 2000;62(Suppl 1):33–38. doi:10.1159/00005185310940685
  • Maroun J, Kocha W, Kvols L, et al. Guidelines for the diagnosis and management of carcinoid tumours. Part 1: the gastrointestinal tract. A statement from a Canadian National Carcinoid Expert Group. Curr Oncol. 2006;13(2):67–76.17576444
  • Feinberg Y, Law C, Singh S, Wright FC. Patient experiences of having a neuroendocrine tumour: a qualitative study. Eur J Oncol Nurs. 2013;17(5):541–545. doi:10.1016/j.ejon.2013.02.00323522828
  • Goldstein G, Evans K, Shah A, Lapuerta P. Carcinoid syndrome impact survey: evaluation of the burden of illness and the gap between patient and oncologist perceptions. J Clin Oncol. 2017;35:8_suppl.
  • Yoo H, Shin DW, Jeong A, et al. Perceived social support and its impact on depression and health-related quality of life: a comparison between cancer patients and general population. Jpn J Clin Oncol. 2017;47(8):728–734. doi:10.1093/jjco/hyx06428531292
  • Hinz A, Mehnert A, Degi C, Reissmann DR, Schotte D, Schulte T. The relationship between global and specific components of quality of life, assessed with the EORTC QLQ-C30 in a sample of 2019 cancer patients. Eur J Cancer Care (Engl). 2017;26:2. doi:10.1111/ecc.12416
  • Jimenez-Fonseca P, Carmona-Bayonas A, Martin-Perez E, et al. Health-related quality of life in well-differentiated metastatic gastroenteropancreatic neuroendocrine tumors. Cancer Metastasis Rev. 2015;34(3):381–400. doi:10.1007/s10555-015-9573-126245646
  • Ramage JK, Davies AH. Measurement of quality of life in carcinoid/neuroendocrine tumours. Endocr Relat Cancer. 2003;10(4):483–486. doi:10.1677/erc.0.010048314713261
  • Chau I, Casciano R, Willet J, Wang X, Yao JC. Quality of life, resource utilisation and health economics assessment in advanced neuroendocrine tumours: a systematic review. Eur J Cancer Care (Engl). 2013;22(6):714–725. doi:10.1111/ecc.2013.22.issue-623895457
  • Martini C, Gamper E-M, Wintner L, et al. Systematic review reveals lack of quality in reporting health-related quality of life in patients with gastroenteropancreatic neuroendocrine tumours. Health Qual Life Outcomes. 2016;14(1):127. doi:10.1186/s12955-016-0426-627614762
  • Vinik E, Silva MP, Vinik AI. Measuring the relationship of quality of life and health status, including tumor burden, symptoms, and biochemical measures in patients with neuroendocrine tumors. Endocrinol Metab Clin North Am. 2011;40(1):97–109, viii. doi:10.1016/j.ecl.2010.12.00821349413
  • Fayers P, Bottomley A. Quality of life research within the EORTC-the EORTC QLQ-C30. European Organisation for Research and Treatment of Cancer. Eur J Cancer. 2002;38(Suppl 4):S125–S133.11858978
  • Davies AHG, Larsson G, Ardill J, et al. Development of a disease-specific Quality of Life questionnaire module for patients with gastrointestinal neuroendocrine tumours. Eur J Cancer. 2006;42(4):477–484. doi:10.1016/j.ejca.2005.10.02516412628
  • Yadegarfar G, Friend L, Jones L, et al. Validation of the EORTC QLQ-GINET21 questionnaire for assessing quality of life of patients with gastrointestinal neuroendocrine tumours. Br J Cancer. 2013;108(2):301–310. doi:10.1038/bjc.2012.49423322194
  • Vinik E, Carlton CA, Silva MP, Vinik AI. Development of the Norfolk quality of life tool for assessing patients with neuroendocrine tumors. Pancreas. 2009;38(3):e87–e95. doi:10.1097/MPA.0b013e31819b644119276865
  • Lamarca A, Ronot M, Moall S, et al. Tumour growth rate (TGR) in neuroendocrine tumours (NETs): changes following systemic treatment; the GREPONET-2 study [Internet]; 2019. doi:10.1634/theoncologist.2018-0672
  • Siddiqui EJ, Thompson CS, Mikhailidis DP, Mumtaz FH. The role of serotonin in tumour growth (review). Oncol Rep. 2005;14(6):1593–1597.16273262
  • Jiang S-H, Li J, Dong F-Y, et al. Increased serotonin signaling contributes to the Warburg effect in pancreatic tumor cells under metabolic stress and promotes growth of pancreatic tumors in mice. Gastroenterology. 2017;153(1):277–291.e19. doi:10.1053/j.gastro.2017.03.00828315323
  • Alpini G, Invernizzi P, Gaudio E, et al. Serotonin metabolism is dysregulated in cholangiocarcinoma, which has implications for tumor growth. Cancer Res. 2008;68(22):9184–9193. doi:10.1158/0008-5472.CAN-08-213319010890
  • Svejda B, Kidd M, Timberlake A, et al. Serotonin and the 5-HT7 receptor: the link between hepatocytes, IGF-1 and small intestinal neuroendocrine tumors. Cancer Sci. 2013;104(7):844–855. doi:10.1111/cas.1217423578138
  • Svejda B, Kidd M, Giovinazzo F, et al. The 5-HT(2B) receptor plays a key regulatory role in both neuroendocrine tumor cell proliferation and the modulation of the fibroblast component of the neoplastic microenvironment. Cancer. 2010;116(12):2902–2912. doi:10.1002/cncr.2504920564397
  • Drozdov I, Kidd M, Gustafsson BI, et al. Autoregulatory effects of serotonin on proliferation and signaling pathways in lung and small intestine neuroendocrine tumor cell lines. Cancer. 2009;115(21):4934–4945. doi:10.1002/cncr.2453319634160
  • Vicaut E, Laemmel E, Stucker O. Impact of serotonin on tumour growth. Ann Med. 2000;32(3):187–194. doi:10.3109/0785389000899882610821326
  • Vinik A, Bottomley A, Korytowsky B, et al. Patient-reported outcomes and quality of life with sunitinib versus placebo for pancreatic neuroendocrine tumors: results from an international phase III trial. Target Oncol. 2016;11(6):815–824. doi:10.1007/s11523-016-0462-527924459
  • Fjallskog M-LH, 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(1):53–58. doi:10.1159/00011757518285678