دورية أكاديمية

Update in the management of gastroenteropancreatic neuroendocrine tumors.

التفاصيل البيبلوغرافية
العنوان: Update in the management of gastroenteropancreatic neuroendocrine tumors.
المؤلفون: Sedlack AJH; Medical Scientist Training Program, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA., Varghese DG; Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA., Naimian A; Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA., Yazdian Anari P; Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA.; Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA., Bodei L; Memorial Sloan Kettering Cancer Center, New York, New York, USA., Hallet J; Odette Cancer Centre, Sunnybrook Health Sciences Centre, East York, Ontario, Canada., Riechelmann RP; Departamento de Oncologia Clínica, AC Camargo Cancer Center, Sao Paolo, Brazil., Halfdanarson T; Mayo Clinic, Rochester, Minnesota, USA., Capdevilla J; Vall d'Hebron University Hospital, Barcelona, Spain., Del Rivero J; Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA.
المصدر: Cancer [Cancer] 2024 Sep 15; Vol. 130 (18), pp. 3090-3105. Date of Electronic Publication: 2024 Jul 16.
نوع المنشور: Journal Article; Review
اللغة: English
بيانات الدورية: Publisher: Wiley Country of Publication: United States NLM ID: 0374236 Publication Model: Print-Electronic Cited Medium: Internet ISSN: 1097-0142 (Electronic) Linking ISSN: 0008543X NLM ISO Abbreviation: Cancer Subsets: MEDLINE
أسماء مطبوعة: Publication: <2005- >: Hoboken, NJ : Wiley
Original Publication: New York [etc.] Published for the American Cancer Society by J. Wiley [etc.]
مواضيع طبية MeSH: Neuroendocrine Tumors*/therapy , Neuroendocrine Tumors*/pathology , Pancreatic Neoplasms*/therapy , Pancreatic Neoplasms*/pathology , Stomach Neoplasms*/therapy , Stomach Neoplasms*/pathology , Intestinal Neoplasms*/therapy , Intestinal Neoplasms*/pathology, Humans ; Somatostatin/analogs & derivatives ; Somatostatin/therapeutic use ; Molecular Targeted Therapy/methods
مستخلص: Neuroendocrine neoplasms are a diverse group of neoplasms that can occur in various areas throughout the body. Well-differentiated neuroendocrine tumors (NETs) most often arise in the gastrointestinal tract, termed gastroenteropancreatic neuroendocrine tumors (GEP-NETs). Although GEP-NETs are still uncommon, their incidence and prevalence have been steadily increasing over the past decades. The primary treatment for GEP-NETs is surgery, which offers the best chance for a cure. However, because GEP-NETs are often slow-growing and do not cause symptoms until they have spread widely, curative surgery is not always an option. Significant advances have been made in systemic and locoregional treatment options in recent years, including peptide-receptor radionuclide therapy with α and β emitters, somatostatin analogs, chemotherapy, and targeted molecular therapies.
(© 2024 American Cancer Society.)
References: White BE, Rous B, Chandrakumaran K, et al. Incidence and survival of neuroendocrine neoplasia in England 1995‐2018: aa retrospective, population‐based study. Lancet Reg Health Eur. 2022;23:100510. doi:10.1016/j.lanepe.2022.100510.
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.2017.0589.
Basuroy R, Bouvier C, Ramage JK, Sissons M, Srirajaskanthan R. Delays and routes to diagnosis of neuroendocrine tumours. BMC Cancer. 2018;18(1):1122. doi:10.1186/s12885-018-5057-3.
Lee MR, Harris C, Baeg KJ, Aronson A, Wisnivesky JP, Kim MK. Incidence trends of gastroenteropancreatic neuroendocrine tumors in the United States. Clin Gastroenterol Hepatol. 2019;17(11):2212‐2217.e1. doi:10.1016/j.cgh.2018.12.017.
Das S, Dasari A. Novel therapeutics for patients with well‐differentiated gastroenteropancreatic neuroendocrine tumors. Ther Adv Med Oncol. 2021;13:17588359211018047. doi:10.1177/17588359211018047.
Díez M, Teulé A, Salazar R. Gastroenteropancreatic neuroendocrine tumors: diagnosis and treatment. Ann Gastroenterol. 2013;26(1):29‐36.
Thiis‐Evensen E, Boyar Cetinkaya R. Incidence and prevalence of neuroendocrine neoplasms in Norway 1993–2021. J Neuroendocrinol. 2023;35(4):e13264. doi:10.1111/jne.13264.
Das S, Dasari A. Epidemiology, incidence, and prevalence of neuroendocrine neoplasms: are there global differences? Curr Oncol Rep. 2021;23(4):43. doi:10.1007/s11912-021-01029-7.
Ellis L, Shale MJ, Coleman MP. Carcinoid tumors of the gastrointestinal tract: trends in incidence in England since 1971. Am J Gastroenterol. 2010;105(12):2563‐2569. doi:10.1038/ajg.2010.341.
Hallet J, Law CHL, Cukier M, Saskin R, Liu N, Singh S. Exploring the rising incidence of neuroendocrine tumors: a population‐based analysis of epidemiology, metastatic presentation, and outcomes. Cancer. 2015;121(4):589‐597. doi:10.1002/cncr.29099.
Feldman JM. Carcinoid tumors and syndrome. Semin Oncol. 1987;14(3):237‐246.
Cives M, Strosberg JR. Gastroenteropancreatic neuroendocrine tumors. CA Cancer J Clin. 2018;68(6):471‐487. doi:10.3322/caac.21493.
Rosai J. The origin of neuroendocrine tumors and the neural crest saga. Mod Pathol. 2011;24(2):S53‐S57. doi:10.1038/modpathol.2010.166.
Klöppel G. Neuroendocrine neoplasms: dichotomy, origin and classifications. Visc Med. 2017;33(5):324‐330. doi:10.1159/000481390.
Di Domenico A, Pipinikas CP, Maire RS, et al. Epigenetic landscape of pancreatic neuroendocrine tumours reveals distinct cells of origin and means of tumour progression. Commun Biol. 2020;3(1):1‐11. doi:10.1038/s42003-020-01479-y.
Hirabayashi K, Zamboni G, Nishi T, Tanaka A, Kajiwara H, Nakamura N. Histopathology of gastrointestinal neuroendocrine neoplasms. Front Oncol. 2013;3:2. doi:10.3389/fonc.2013.00002.
Asa SL. Pancreatic endocrine tumors. Mod Pathol. 2011;24(suppl 2):S66‐S77. doi:10.1038/modpathol.2010.127.
Rindi G, Mete O, Uccella S, et al. Overview of the 2022 WHO Classification of Neuroendocrine Neoplasms. Endocr Pathol. 2022;33(1):115‐154. doi:10.1007/s12022-022-09708-2.
Bellizzi AM. Immunohistochemistry in the diagnosis and classification of neuroendocrine neoplasms: what can brown do for you? Hum Pathol. 2020;96:8‐33. doi:10.1016/j.humpath.2019.12.002.
Maxwell JE, Sherman SK, Stashek KM, O’Dorisio TM, Bellizzi AM, Howe JR. A practical method to determine the site of unknown primary in metastatic neuroendocrine tumors. Surgery. 2014;156(6):1359‐1366. doi:10.1016/j.surg.2014.08.008.
Mafficini A, Scarpa A. Genomic landscape of pancreatic neuroendocrine tumours: the International Cancer Genome Consortium. J Endocrinol. 2018;236(3):R161‐R167. doi:10.1530/JOE-17-0560.
Scarpa A, Chang DK, Nones K, et al. Whole‐genome landscape of pancreatic neuroendocrine tumours. Nature. 2017;543(7643):65‐71. doi:10.1038/nature21063.
Crona J, Gustavsson T, Norlén O, et al. Somatic mutations and genetic heterogeneity at the CDKN1B locus in small intestinal neuroendocrine tumors. Ann Surg Oncol. 2015;22(suppl 3):S1428‐S1435. doi:10.1245/s10434-014-4351-9.
Francis JM, Kiezun A, Ramos AH, et al. Somatic mutation of CDKN1B in small intestine neuroendocrine tumors. Nat Genet. 2013;45(12):1483‐1486. doi:10.1038/ng.2821.
Banck MS, Kanwar R, Kulkarni AA, et al. The genomic landscape of small intestine neuroendocrine tumors. J Clin Invest. 2013;123(6):2502‐2508. doi:10.1172/JCI67963.
Venizelos A, Elvebakken H, Perren A, et al. The molecular characteristics of high‐grade gastroenteropancreatic neuroendocrine neoplasms. Endocr Relat Cancer. 2021;29(1):1‐14. doi:10.1530/ERC-21-0152.
Jeong SY, Kim ST, Jang JY. The tumor mutation burden in gastro‐entero‐pancreatic‐neuroendocrine tumors. J Clin Orthod. 2023;41(suppl 4):654. doi:10.1200/JCO.2023.41.4_suppl.654.
Padda SK, Aggarwal RR, Ashok A, Mauer E, Shirazi M, Bergsland EK. Prevalence of high tumor mutational burden (TMB‐H) and microsatellite instability‐high (MSI‐H) status in neuroendocrine neoplasms. J Clin Orthod. 2022;40(suppl 16):2625. doi:10.1200/JCO.2022.40.16_suppl.2625.
Niederle B, Selberherr A, Bartsch DK, et al. Multiple endocrine neoplasia type 1 and the pancreas: diagnosis and treatment of functioning and non‐functioning pancreatic and duodenal neuroendocrine neoplasia within the MEN1 syndrome – an international consensus statement. Neuroendocrinology. 2020;111(7):609‐630. doi:10.1159/000511791.
Riechelmann RP, Soares DC, Dias C, Carraro DM, Torrezan GT. Li‐Fraumeni‐associated pancreatic neuroendocrine tumour and XAF1 p.Glu134Ter risk modifier variant. Ecancermedicalscience. 2022;16:1487. doi:10.3332/ecancer.2022.1487.
Aversa JG, De Abreu FB, Yano S, et al. The first pancreatic neuroendocrine tumor in Li‐Fraumeni syndrome: a case report. BMC Cancer. 2020;20(1):256. doi:10.1186/s12885-020-06723-6.
Geurts JL. Inherited syndromes involving pancreatic neuroendocrine tumors. J Gastrointest Oncol. 2020;11(3):559‐566. doi:10.21037/jgo.2020.03.09.
Shah MH, Goldner WS, Benson AB, et al. Neuroendocrine and adrenal tumors, version 2.2021, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw. 2021;19(7):839‐868. doi:10.6004/jnccn.2021.0032.
Zhang JY, Kunz PL. Making sense of a complex disease: a practical approach to managing neuroendocrine tumors. JCO Oncol Pract. 2022;18(4):258‐264. doi:10.1200/OP.21.00240.
Kawamoto S, Johnson PT, Shi C, et al. Pancreatic neuroendocrine tumor with cystlike changes: evaluation with MDCT. AJR Am J Roentgenol. 2013;200(3):W283‐W290. doi:10.2214/AJR.12.8941.
Canellas R, Lo G, Bhowmik S, Ferrone C, Sahani D. Pancreatic neuroendocrine tumor: correlations between MRI features, tumor biology, and clinical outcome after surgery. J Magn Reson Imag. 2018;47(2):425‐432. doi:10.1002/jmri.25756.
Lee NJ, Hruban RH, Fishman EK. Pancreatic neuroendocrine tumor: review of heterogeneous spectrum of CT appearance. Abdom Radiol (NY). 2018;43(11):3025‐3034. doi:10.1007/s00261-018-1574-4.
Navin PJ, Ehman EC, Liu JB, et al. Imaging of Small‐Bowel Neuroendocrine Neoplasms: AJR Expert Panel Narrative Review. Am J Roentgenol. 2023;221(3):289‐301. doi:10.2214/AJR.22.28877.
Martin S, Iravani A, Shetty AS, et al. Neuroendocrine neoplasm imaging: protocols by site of origin. Abdom Radiol. 2022;47(12):4081‐4095. doi:10.1007/s00261-022-03713-z.
Bodei L, Sundin A, Kidd M, Prasad V, Modlin IM. The status of neuroendocrine tumor imaging: from darkness to light? Neuroendocrinology. 2015;101(1):1‐17. doi:10.1159/000367850.
Lee L, Ito T, Jensen RT. Imaging of pancreatic neuroendocrine tumors: recent advances, current status and controversies. Expert Rev Anticancer Ther. 2018;18(9):837‐860. doi:10.1080/14737140.2018.1496822.
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(2):167‐183. doi:10.1159/000184855.
Kirshbom PM, Kherani AR, Onaitis MW, Feldman JM, Tyler DS. Carcinoids of unknown origin: comparative analysis with foregut, midgut, and hindgut carcinoids. Surgery. 1998;124(6):1063‐1070. doi:10.1067/msy.1998.93105.
Khanna L, Prasad SR, Sunnapwar A, et al. Pancreatic neuroendocrine neoplasms: 2020 update on pathologic and imaging findings and classification. Radiographics. 2020;40(5):1240‐1262. doi:10.1148/rg.2020200025.
Chiti G, Grazzini G, Cozzi D, et al. Imaging of pancreatic neuroendocrine neoplasms. Int J Environ Res Public Health. 2021;18(17):8895. doi:10.3390/ijerph18178895.
Grawe F, Rosenberger N, Ingenerf M, et al. Diagnostic performance of PET/CT in the detection of liver metastases in well‐differentiated NETs. Cancer Imag. 2023;23(1):41. doi:10.1186/s40644-023-00556-9.
Ronot M, Clift AK, Baum RP, et al. Morphological and functional imaging for detecting and assessing the resectability of neuroendocrine liver metastases. Neuroendocrinology. 2018;106(1):74‐88. doi:10.1159/000479293.
Dromain C, de Baere T, Baudin E, et al. MR imaging of hepatic metastases caused by neuroendocrine tumors: comparing four techniques. AJR Am J Roentgenol. 2003;180(1):121‐128. doi:10.2214/ajr.180.1.1800121.
Berzaczy D, Giraudo C, Haug AR, et al. Whole‐body 68Ga‐DOTANOC PET/MRI versus 68Ga‐DOTANOC PET/CT in patients with neuroendocrine tumors. Clin Nucl Med. 2017;42(9):669‐674. doi:10.1097/RLU.0000000000001753.
Kim YI, Cheon GJ, Paeng JC, et al. Total lesion glycolysis as the best 18F‐FDG PET/CT parameter in differentiating intermediate‐high risk adrenal incidentaloma. Nucl Med Commun. 2014;35(6):606‐612. doi:10.1097/MNM.0000000000000101.
Magi L, Prosperi D, Lamberti G, et al. Role of [18F]FDG PET/CT in the management of G1 gastro‐entero‐pancreatic neuroendocrine tumors. Endocrine. 2022;76(2):484‐490. doi:10.1007/s12020-022-03000-3.
Varlamov E, Hinojosa‐Amaya JM, Stack M, Fleseriu M. Diagnostic utility of Gallium‐68‐somatostatin receptor PET/CT in ectopic ACTH‐secreting tumors: a systematic literature review and single‐center clinical experience. Pituitary. 2019;22(5):445‐455. doi:10.1007/s11102-019-00972-w.
Conti M, Eriksson L. Physics of pure and non‐pure positron emitters for PET: a review and a discussion. EJNMMI Phys. 2016;3(1):8. doi:10.1186/s40658-016-0144-5.
Khatri W, Spiro E, Henderson A, Rowe SP, Solnes LB. Gastro‐entero‐pancreatic tumors: FDG positron emission tomography/computed tomography. Pet Clin. 2023;18(2):243‐250. doi:10.1016/j.cpet.2022.11.007.
Prosperi D, Carideo L, Russo VM, et al. A systematic review on combined [18F]FDG and 68Ga‐SSA PET/CT in pulmonary carcinoid. J Clin Med. 2023;12(11):3719. doi:10.3390/jcm12113719.
Hindié E. The NETPET score: combining FDG and somatostatin receptor imaging for optimal management of patients with metastatic well‐differentiated neuroendocrine tumors. Theranostics. 2017;7(5):1159‐1163. doi:10.7150/thno.19588.
Chan DL, Pavlakis N, Schembri GP, et al. Dual somatostatin receptor/FDG PET/CT imaging in metastatic neuroendocrine tumours: proposal for a novel grading scheme with prognostic significance. Theranostics. 2017;7(5):1149‐1158. doi:10.7150/thno.18068.
Chan DL, Hayes AR, Karfis I, et al. Dual [68Ga]DOTATATE and [18F]FDG PET/CT in patients with metastatic gastroenteropancreatic neuroendocrine neoplasms: a multicentre validation of the NETPET score. Br J Cancer. 2023;128(4):549‐555. doi:10.1038/s41416-022-02061-5.
Keutgen XM, Schadde E, Pommier RF, Halfdanarson TR, Howe JR, Kebebew E. Metastatic neuroendocrine tumors of the gastrointestinal tract and pancreas: a surgeon’s plea to centering attention on the liver. Semin Oncol. 2018;45(4):232‐235. doi:10.1053/j.seminoncol.2018.07.002.
Howe JR, Cardona K, Fraker DL, et al. The surgical management of small bowel neuroendocrine tumors: consensus guidelines of the North American Neuroendocrine Tumor Society. Pancreas. 2017;46(6):715‐731. doi:10.1097/MPA.0000000000000846.
Howe JR, Merchant NB, Conrad C, et al. The North American Neuroendocrine Tumor Society consensus paper on the surgical management of pancreatic neuroendocrine tumors. Pancreas. 2020;49(1):1‐33. doi:10.1097/MPA.0000000000001454.
Heidsma CM, Engelsman AF, van Dieren S, et al. Watchful waiting for small non‐functional pancreatic neuroendocrine tumours: nationwide prospective cohort study (PANDORA). Br J Surg. 2021;108(8):888‐891. doi:10.1093/bjs/znab088.
Partelli S, Massironi S, Zerbi A, et al. Management of asymptomatic sporadic non‐functioning pancreatic neuroendocrine neoplasms no larger than 2 cm: interim analysis of prospective ASPEN trial. Br J Surg. 2022;109(12):1186‐1190. doi:10.1093/bjs/znac267.
Kos‐Kudła B, Castaño JP, Denecke T, et al. European Neuroendocrine Tumour Society (ENETS) 2023 guidance paper for nonfunctioning pancreatic neuroendocrine tumours. J Neuroendocrinol. 2023;35(12):e13343. doi:10.1111/jne.13343.
Pavel M, Öberg K, Falconi M, et al. Gastroenteropancreatic neuroendocrine neoplasms: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow‐up. Ann Oncol. 2020;31(7):844‐860. doi:10.1016/j.annonc.2020.03.304.
Gudmundsdottir H, Habermann EB, Vierkant RA, et al. Survival and symptomatic relief after cytoreductive hepatectomy for neuroendocrine tumor liver metastases: long‐term follow‐up evaluation of more than 500 patients. Ann Surg Oncol. 2023;30(8):4840‐4851. doi:10.1245/s10434-023-13372-z.
Søreide K, Stättner S, Hallet J. Surgery as a principle and technical consideration for primary tumor resection of small bowel neuroendocrine tumors. Ann Surg Oncol. 2024;31(2):1125‐1137. doi:10.1245/s10434-023-14610-0.
Strosberg JR, Halfdanarson TR, Bellizzi AM, et al. The North American Neuroendocrine Society (NANETS) consensus guidelines for surveillance and medical management of midgut neuroendocrine tumors. Pancreas. 2017;46(6):707‐714. doi:10.1097/MPA.0000000000000850.
Kolasińska‐Ćwikła A, Nowicki ML, Sankowski AJ, et al. Radiological and clinical efficacy of intra‐arterial 90Y‐DOTATATE in patients with unresectable, progressive, liver dominant neuroendocrine neoplasms. J Clin Med. 2021;10(8):1794. doi:10.3390/jcm10081794.
Myrehaug S, Chan DL, Rodriguez‐Freixinos V, et al. A pilot study of everolimus and radiation for neuroendocrine liver metastases. Endocr Relat Cancer. 2021;28(8):541‐548. doi:10.1530/ERC-21-0030.
Chan DL, Thompson R, Lam M, et al. External beam radiotherapy in the treatment of gastroenteropancreatic neuroendocrine tumours: a systematic review. Clin Oncol. 2018;30(7):400‐408. doi:10.1016/j.clon.2018.03.006.
Hudson JM, Chung HTK, Chu W, et al. Stereotactic ablative radiotherapy for the management of liver metastases from neuroendocrine neoplasms: a preliminary study. Neuroendocrinology. 2022;112(2):153‐160. doi:10.1159/000514914.
Pavel M, O’Toole D, Costa F, 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. Neuroendocrinology. 2016;103(2):172‐185. doi:10.1159/000443167.
Mohan H, Nicholson P, Winter DC, et al. Radiofrequency ablation for neuroendocrine liver metastases: a systematic review. J Vasc Intervent Radiol. 2015;26(7):935‐942.e1. doi:10.1016/j.jvir.2014.12.009.
Lee L, Ramos‐Alvarez I, Jensen RT. Predictive factors for resistant disease with medical/radiologic/liver‐directed anti‐tumor treatments in patients with advanced pancreatic neuroendocrine neoplasms: recent advances and controversies. Cancers. 2022;14(5):1250. doi:10.3390/cancers14051250.
Currie BM, Nadolski G, Mondschein J, et al. Chronic hepatotoxicity in patients with metastatic neuroendocrine tumor: transarterial chemoembolization versus transarterial radioembolization. J Vasc Intervent Radiol. 2020;31(10):1627‐1635. doi:10.1016/j.jvir.2020.05.019.
Padia SA. Radioembolization versus chemoembolization for neuroendocrine metastases. J Vasc Interv Radiol. 2021;32(3):482‐483. doi:10.1016/j.jvir.2020.11.017.
Ngo L, Elnahla A, Attia AS, et al. Chemoembolization versus radioembolization for neuroendocrine liver metastases: a meta‐analysis comparing clinical outcomes. Ann Surg Oncol. 2021;28(4):1950‐1958. doi:10.1245/s10434-020-09469-4.
Guo LJ, Tang CW. Somatostatin analogues do not prevent carcinoid crisis. Asian Pac J Cancer Prev. 2014;15(16):6679‐6683. doi:10.7314/apjcp.2014.15.16.6679.
Kwon DH, Paciorek A, Mulvey CK, et al. Periprocedural management of patients undergoing liver resection or embolotherapy for neuroendocrine tumor metastases. Pancreas. 2019;48(4):496‐503. doi:10.1097/MPA.0000000000001271.
Vaughan DJ, Brunner MD. Anesthesia for patients with carcinoid syndrome. Int Anesthesiol Clin. 1997;35(4):129‐142. doi:10.1097/00004311-199703540-00009.
Majeed F, Porter TR, Tarantolo S, Duhachek‐Stapelman A, Xie F, Elhendy A. Carcinoid crisis and reversible right ventricular dysfunction after embolization in untreated carcinoid syndrome. Eur J Echocardiogr. 2007;8(5):386‐389. doi:10.1016/j.euje.2006.04.008.
Davì MV, Bodei L, Francia G, et al. Carcinoid crisis induced by receptor radionuclide therapy with 90Y‐DOTATOC in a case of liver metastases from bronchial neuroendocrine tumor (atypical carcinoid). J Endocrinol Invest. 2006;29(6):563‐567. doi:10.1007/BF03344149.
Hijioka S, Morizane C, Ikeda M, Ishii H, Okusaka T, Furuse J. Current status of medical treatment for gastroenteropancreatic neuroendocrine neoplasms and future perspectives. Jpn J Clin Oncol. 2021;51(8):1185‐1196. doi:10.1093/jjco/hyab076.
Rinke A, Müller 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 Orthod. 2009;27(28):4656‐4663. doi:10.1200/JCO.2009.22.8510.
Caplin ME, Pavel M, Ćwikła JB, et al. Lanreotide in metastatic enteropancreatic neuroendocrine tumors. N Engl J Med. 2014;371(3):224‐233. doi:10.1056/NEJMoa1316158.
Caplin ME, Pavel M, Ćwikła JB, et al. Anti‐tumour effects of lanreotide for pancreatic and intestinal neuroendocrine tumours: the CLARINET open‐label extension study. Endocr Relat Cancer. 2016;23(3):191‐199. doi:10.1530/ERC-15-0490.
Rinke A, Wittenberg M, 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 (PROMID): results of long‐term survival. Neuroendocrinology. 2016;104(1):26‐32. doi:10.1159/000443612.
Sidéris L, Dubé P, Rinke A. Antitumor effects of somatostatin analogs in neuroendocrine tumors. Oncologist. 2012;17(6):747‐755. doi:10.1634/theoncologist.2011-0458.
Rustgi SD, Oh A, Yang JY, et al. Initiation of somatostatin analogues for neuroendocrine tumor patients: a cost‐effectiveness analysis. BMC Cancer. 2021;21(1):597. doi:10.1186/s12885-021-08306-5.
Halfdanarson TR, Strosberg JR, Tang L, et al. The North American Neuroendocrine Tumor Society consensus guidelines for surveillance and medical management of pancreatic neuroendocrine tumors. Pancreas. 2020;49(7):863‐881. doi:10.1097/MPA.0000000000001597.
Krishnan T, Safro M, Furlanetto DM, et al. Clinical impact of unsuccessful subcutaneous administration of octreotide LAR instead of intramuscular administration in patients with metastatic gastroenteropancreatic neuroendocrine tumors. J Neuroendocrinol. 2024;36:e13360. doi:10.1111/jne.13360.
Boyd AE, DeFord LL, Mares JE, et al. Improving the success rate of gluteal intramuscular injections. Pancreas. 2013;42(5):878‐882. doi:10.1097/MPA.0b013e318279d552.
Bolanowski M, Kałużny M, Witek P, Jawiarczyk‐Przybyłowska A. Pasireotide—a novel somatostatin receptor ligand after 20 years of use. Rev Endocr Metab Disord. 2022;23(3):601‐620. doi:10.1007/s11154-022-09710-3.
Andre A, Squittieri N, Patil SB. Evaluation of the octreotide acetate pen injector and its instructions for use in a formative human factors study. Adv Ther. 2021;38(6):3129‐3142. doi:10.1007/s12325-021-01739-1.
Madan A, Markison S, Betz SF, et al. Paltusotine, a novel oral once‐daily nonpeptide SST2 receptor agonist, suppresses GH and IGF‐1 in healthy volunteers. Pituitary. 2022;25(2):328‐339. doi:10.1007/s11102-021-01201-z.
Singh S. Interim safety and exploratory efficacy results of a phase 2, randomized, parallel‐group study of oral paltusotine treatment in patients with carcinoid syndrome. Paper presented at: European Neuroendocrine Tumor Society; March 13, 2024.
Kulke MH, Hörsch 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.2016.69.2780.
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-17-0455.
Khanna L, Halfdanarson TR, Sonbol MB, Eiring R, Prond T, Camilleri M. Bile acid malabsorption in patients with neuroendocrine tumors. Dig Dis Sci. 2022;67(6):2517‐2525. doi:10.1007/s10620-021-07189-7.
Naraev BG, Halland M, Halperin DM, Purvis AJ, O’Dorisio TM, Halfdanarson TR. Management of diarrhea in patients with carcinoid syndrome. Pancreas. 2019;48(8):961‐972. doi:10.1097/MPA.0000000000001384.
Eads JR, Reidy‐Lagunes D, Soares HP, et al. Differential diagnosis of diarrhea in patients with neuroendocrine tumors. Pancreas. 2020;49(9):1123‐1130. doi:10.1097/MPA.0000000000001658.
Mosalem O, Sonbol MB, Halfdanarson TR, Starr JS. Tyrosine kinase inhibitors and immunotherapy updates in neuroendocrine neoplasms. Best Pract Res Clin Endocrinol Metabol. 2023;37(5):101796. doi:10.1016/j.beem.2023.101796.
Grillo F, Florio T, Ferraù F, et al. Emerging multitarget tyrosine kinase inhibitors in the treatment of neuroendocrine neoplasms. Endocr Relat Cancer. 2018;25(9):R453‐R466. doi:10.1530/ERC-17-0531.
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/NEJMoa1003825.
Raymond E, Kulke MH, Qin S, et al. Efficacy and safety of sunitinib in patients with well‐differentiated pancreatic neuroendocrine tumours. Neuroendocrinology. 2018;107(3):237‐245. doi:10.1159/000491999.
Xu J, Shen L, Zhou Z, et al. Surufatinib in advanced extrapancreatic neuroendocrine tumours (SANET‐ep): a randomised, double‐blind, placebo‐controlled, phase 3 study. Lancet Oncol. 2020;21(11):1500‐1512. doi:10.1016/S1470-2045(20)30496-4.
Chan JA, Faris JE, Murphy JE, et al. Phase II trial of cabozantinib in patients with carcinoid and pancreatic neuroendocrine tumors (pNET). J Clin Orthod. 2017;35(suppl 4):228. doi:10.1200/JCO.2017.35.4_suppl.228.
Capdevila J, Fazio N, Lopez C, et al. Lenvatinib in patients with advanced grade 1/2 pancreatic and gastrointestinal neuroendocrine tumors: results of the phase II TALENT trial (GETNE1509). J Clin Oncol. 2021;39(20):2304‐2312. doi:10.1200/JCO.20.03368.
Strosberg JR, Cives M, Hwang J, et al. A phase II study of axitinib in advanced neuroendocrine tumors. Endocr Relat Cancer. 2016;23(5):411‐418. doi:10.1530/ERC-16-0008.
Bergsland EK, Mahoney MR, Asmis TR, et al. Prospective randomized phase II trial of pazopanib versus placebo in patients with progressive carcinoid tumors (CARC) (Alliance A021202). J Clin Oncol. 2019;37(suppl 15):4005. doi:10.1200/JCO.2019.37.15_suppl.4005.
Bongiovanni A, Liverani C, Recine F, et al. Phase‐II trials of pazopanib in metastatic neuroendocrine neoplasia (mNEN): a systematic review and meta‐analysis. Front Oncol. 2020;10:414. doi:10.3389/fonc.2020.00414.
Grande E, Capdevila J, Castellano D, et al. Pazopanib in pretreated advanced neuroendocrine tumors: a phase II, open‐label trial of the Spanish Task Force Group for neuroendocrine tumors (GETNE). Ann Oncol. 2015;26(9):1987‐1993. doi:10.1093/annonc/mdv252.
Phan AT, Halperin DM, Chan JA, et al. Pazopanib and depot octreotide in advanced, well‐differentiated neuroendocrine tumours: a multicentre, single‐group, phase 2 study. Lancet Oncol. 2015;16(6):695‐703. doi:10.1016/S1470-2045(15)70136-1.
Garcia‐Carbonero R, Benavent M, Fonseca PJ, et al. 1097O The AXINET trial (GETNE1107): axitinib plus octreotide LAR improves PFS by blinded central radiological assessment vs placebo plus octreotide LAR in G1‐2 extrapancreatic NETs. Ann Oncol. 2021;32:S907‐S908. doi:10.1016/j.annonc.2021.08.179.
Castellano D, Capdevila J, Sastre J, et al. Sorafenib and bevacizumab combination targeted therapy in advanced neuroendocrine tumour: a phase II study of Spanish Neuroendocrine Tumour Group (GETNE0801). Eur J Cancer. 2013;49(18):3780‐3787. doi:10.1016/j.ejca.2013.06.042.
Xu J, Shen L, Bai C, et al. Surufatinib in advanced pancreatic neuroendocrine tumours (SANET‐p): a randomised, double‐blind, placebo‐controlled, phase 3 study. Lancet Oncol. 2020;21(11):1489‐1499. doi:10.1016/S1470-2045(20)30493-9.
Chan J, Geyer S, Ou FS, et al. LBA53 Alliance A021602: phase III, double‐blinded study of cabozantinib versus placebo for advanced neuroendocrine tumors (NET) after progression on prior therapy (CABINET). Ann Oncol. 2023;34:S1292. doi:10.1016/j.annonc.2023.10.047.
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. 2016;387(10022):968‐977. doi:10.1016/S0140-6736(15)00817-X.
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/NEJMoa1009290.
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. 2011;378(9808):2005‐2012. doi:10.1016/S0140-6736(11)61742-X.
Strosberg J, El‐Haddad G, Wolin E, et al. Phase 3 trial of 177Lu‐dotatate for midgut neuroendocrine tumors. N Engl J Med. 2017;376(2):125‐135. doi:10.1056/NEJMoa1607427.
Lutathera. Advanced Accelerator Applications USA Inc. Lutathera (lutetium Lu 177 dotatate) injection. Package insert. https://www.accessdata.fda.gov/drugsatfda&#95;docs/label/2018/208700s000lbl.pdf.
Das S, Al‐Toubah T, El‐Haddad G, Strosberg J. 177Lu‐DOTATATE for the treatment of gastroenteropancreatic neuroendocrine tumors. Expert Rev Gastroenterol Hepatol. 2019;13(11):1023‐1031. doi:10.1080/17474124.2019.1685381.
Strosberg JR, Caplin ME, Kunz PL, et al. 177Lu‐Dotatate plus long‐acting octreotide versus high‐dose long‐acting octreotide in patients with midgut neuroendocrine tumours (NETTER‐1): final overall survival and long‐term safety results from an open‐label, randomised, controlled, phase 3 trial. Lancet Oncol. 2021;22(12):1752‐1763. doi:10.1016/S1470-2045(21)00572-6.
de Herder WW. [177Lu]Lu‐DOTA‐TATE in newly diagnosed patients with advanced grade 2 and grande 3, well‐differentiated gastroenteropancreatic neuroendocrine tumors: primary analysis of the phase 3 randomized NETTER‐2 study. Presented at: Eureopean Neuroendocrine Tumor Society 2024; March 13, 2024.
Bodei L, Kidd MS, Singh A, et al. PRRT genomic signature in blood for prediction of 177Lu‐octreotate efficacy. Eur J Nucl Med Mol Imaging. 2018;45(7):1155‐1169. doi:10.1007/s00259-018-3967-6.
Bodei L, Raj N, Do RK, et al. Interim analysis of a prospective validation of 2 blood‐based genomic assessments (PPQ and NETest) to determine the clinical efficacy of 177Lu‐Dotatate in neuroendocrine tumors. J Nucl Med. 2023;64(4):567‐573. doi:10.2967/jnumed.122.264363.
Das S, Chauhan A, Du L, et al. External validation of a clinical score for patients with neuroendocrine tumors under consideration for peptide receptor radionuclide therapy. JAMA Netw Open. 2022;5(1):e2144170. doi:10.1001/jamanetworkopen.2021.44170.
Strosberg J, Leeuwenkamp O, Siddiqui MK. Peptide receptor radiotherapy re‐treatment in patients with progressive neuroendocrine tumors: a systematic review and meta‐analysis. Cancer Treat Rev. 2021;93:102141. doi:10.1016/j.ctrv.2020.102141.
Kim YI. Salvage peptide receptor radionuclide therapy in patients with progressive neuroendocrine tumors: a systematic review and meta‐analysis. Nucl Med Commun. 2021;42(4):451‐458. doi:10.1097/mnm.0000000000001350.
Ocak M, Toklu T, Demirci E, Selçuk N, Kabasakal L. Post‐therapy imaging of 225Ac‐DOTATATE treatment in a patient with recurrent neuroendocrine tumor. Eur J Nucl Med Mol Imaging. 2020;47(11):2711‐2712. doi:10.1007/s00259-020-04725-x.
Delpassand E, Tworowska I, Esfandiari R, Torgue J, Hurt JD, Nunez R. Phase I dose‐escalation study of AlphaMedix for targeted‐alpha‐emitter therapy of PRRT‐naive neuroendocrine patients. J Clin Orthod. 2021;39(suppl 15):4117. doi:10.1200/JCO.2021.39.15_suppl.4117.
Yadav MP, Ballal S, Sahoo RK, Bal C. Efficacy and safety of 225Ac‐DOTATATE targeted alpha therapy in metastatic paragangliomas: a pilot study. Eur J Nucl Med Mol Imaging. 2022;49(5):1595‐1606. doi:10.1007/s00259-021-05632-5.
Ballal S, Yadav MP, Tripathi M, Sahoo RK, Bal C. Survival outcomes in metastatic gastroenteropancreatic neuroendocrine tumor patients receiving concomitant 225Ac‐Dotatate–targeted α‐therapy and capecitabine: a real‐world‐scenario management‐based long‐term outcome study. J Nucl Med. 2023;64(2):211‐218. doi:10.2967/jnumed.122.264043.
Delpassand ES, Tworowska I, Esfandiari R, et al. Targeted α‐emitter therapy with 212Pb‐Dotamtate for the treatment of metastatic SSTR‐expressing neuroendocrine tumors: first‐in‐humans dose‐escalation clinical trial. J Nucl Med. 2022;63(9):1326‐1333. doi:10.2967/jnumed.121.263230.
Baudin E, Walter T, Beron A, et al. 8870 First multicentric randomized phase II trial investigating the antitumor efficacy of peptide receptor radionucleide therapy with 177Lutetium‐Octreotate (OCLU) in unresectable progressive neuroendocrine pancreatic tumor: Results of the OCLURANDOM trial. Ann Oncol. 2022;33:S410‐S416.
Zappi A, Persano I, Galvani L, et al. Chemotherapy in well differentiated neuroendocrine tumors (NET) G1, G2, and G3: a narrative review. J Clin Med. 2023;12(2):717. doi:10.3390/jcm12020717.
Espinosa‐Olarte P, La Salvia A, Riesco‐Martinez MC, Anton‐Pascual B, Garcia‐Carbonero R. Chemotherapy in NEN: still has a role? Rev Endocr Metab Disord. 2021;22(3):595‐614. doi:10.1007/s11154-021-09638-0.
Kunz PL, Graham NT, Catalano PJ, et al. Randomized study of temozolomide or temozolomide and capecitabine in patients with advanced pancreatic neuroendocrine tumors (ECOG‐ACRIN E2211). J Clin Orthod. 2023;41(7):1359‐1369. doi:10.1200/JCO.22.01013.
Salazar R, Tafuto S, Krogh M, et al. LBA45 randomized open label phase III study comparing the efficacy and safety of everolimus followed by chemotherapy (CT) with streptozotocin (STZ)‐5FU upon progression or the reverse sequence, in advanced progressive panNETs: the SEQTOR study (GETNE 1206). Ann Oncol. 2022;33:S1412. doi:10.1016/j.annonc.2022.08.044.
Al‐Toubah T, Pelle E, Valone T, Haider M, Strosberg JR. Efficacy and toxicity analysis of capecitabine and temozolomide in neuroendocrine neoplasms. J Natl Compr Cancer Netw. 2022;20(1):29‐36. doi:10.6004/jnccn.2021.7017.
Girot P, Baudin E, Senellart H, et al. Oxaliplatin and 5‐fluorouracil in advanced well‐differentiated digestive neuroendocrine tumors: a multicenter national retrospective study from the French Group of Endocrine Tumors. Neuroendocrinology. 2021;112(6):537‐546. doi:10.1159/000518650.
Riechelmann RP, Taboada RG, de Jesus VHF, Iglesia M, Trikalinos NA. Therapy sequencing in patients with advanced neuroendocrine neoplasms. Am Soc Clin Oncol Educ Book. 2023(43):e389278. doi:10.1200/EDBK_389278.
Chauhan A, Del Rivero J, Ramirez RA, Soares HP, Li D. Treatment sequencing strategies in advanced neuroendocrine tumors: a review. Cancers. 2022;14(21):5248. doi:10.3390/cancers14215248.
Passhak M, McNamara MG, Hubner RA, Ben‐Aharon I, Valle JW. Choosing the best systemic treatment sequence for control of tumour growth in gastro‐enteropancreatic neuroendocrine tumours (GEP‐NETs): what is the recent evidence? Best Pract Res Clin Endocrinol Metab. 2023;37(5):101836. doi:10.1016/j.beem.2023.101836.
معلومات مُعتمدة: United States CA NCI NIH HHS; United States CA NCI NIH HHS
فهرسة مساهمة: Keywords: delayed diagnosis; gastrointestinal neoplasms; molecularly targeted therapy; neuroendocrine carcinoma; neuroendocrine tumors; pancreatic neoplasms; stomach neoplasms
المشرفين على المادة: 51110-01-1 (Somatostatin)
SCR Disease Name: Gastro-enteropancreatic neuroendocrine tumor
تواريخ الأحداث: Date Created: 20240716 Date Completed: 20240824 Latest Revision: 20240824
رمز التحديث: 20240826
DOI: 10.1002/cncr.35463
PMID: 39012928
قاعدة البيانات: MEDLINE
الوصف
تدمد:1097-0142
DOI:10.1002/cncr.35463