Controversy Over Liver Transplantation or Resection for Neuroendocrine Liver Metastasis: Tumor Biology Cuts the Deal.
Journal
Annals of surgery
ISSN: 1528-1140
Titre abrégé: Ann Surg
Pays: United States
ID NLM: 0372354
Informations de publication
Date de publication:
01 May 2023
01 May 2023
Historique:
medline:
29
11
2023
pubmed:
18
8
2022
entrez:
17
8
2022
Statut:
ppublish
Résumé
In patients with neuroendocrine liver metastasis (NELM), liver transplantation (LT) is an alternative to liver resection (LR), although the choice of therapy remains controversial. In this multicenter study, we aim to provide novel insight in this dispute. Following a systematic literature search, 15 large international centers were contacted to provide comprehensive data on their patients after LR or LT for NELM. Survival analyses were performed with the Kaplan-Meier method, while multivariable Cox regression served to identify factors influencing survival after either transplantation or resection. Inverse probability weighting and propensity score matching was used for analyses with balanced and equalized baseline characteristics. Overall, 455 patients were analyzed, including 230 after LR and 225 after LT, with a median follow-up of 97 months [95% confidence interval (CI): 85-110 months]. Multivariable analysis revealed G3 grading as a negative prognostic factor for LR [hazard ratio (HR)=2.22, 95% CI: 1.04-4.77, P =0.040], while G2 grading (HR=2.52, 95% CI: 1.15-5.52, P =0.021) and LT outside Milan criteria (HR=2.40, 95% CI: 1.16-4.92, P =0.018) were negative prognostic factors in transplanted patients. Inverse probability-weighted multivariate analyses revealed a distinct survival benefit after LT. Matched patients presented a median overall survival (OS) of 197 months (95% CI: 143-not reached) and a 73% 5-year OS after LT, and 119 months (95% CI: 74-133 months) and a 52.8% 5-year OS after LR (HR=0.59, 95% CI: 0.3-0.9, P =0.022). However, the survival benefit after LT was lost if patients were transplanted outside Milan criteria. This multicentric study in patients with NELM demonstrates a survival benefit of LT over LR. This benefit depends on adherence to selection criteria, in particular low-grade tumor biology and Milan criteria, and must be balanced against potential risks of LT.
Sections du résumé
BACKGROUND
BACKGROUND
In patients with neuroendocrine liver metastasis (NELM), liver transplantation (LT) is an alternative to liver resection (LR), although the choice of therapy remains controversial. In this multicenter study, we aim to provide novel insight in this dispute.
METHODS
METHODS
Following a systematic literature search, 15 large international centers were contacted to provide comprehensive data on their patients after LR or LT for NELM. Survival analyses were performed with the Kaplan-Meier method, while multivariable Cox regression served to identify factors influencing survival after either transplantation or resection. Inverse probability weighting and propensity score matching was used for analyses with balanced and equalized baseline characteristics.
RESULTS
RESULTS
Overall, 455 patients were analyzed, including 230 after LR and 225 after LT, with a median follow-up of 97 months [95% confidence interval (CI): 85-110 months]. Multivariable analysis revealed G3 grading as a negative prognostic factor for LR [hazard ratio (HR)=2.22, 95% CI: 1.04-4.77, P =0.040], while G2 grading (HR=2.52, 95% CI: 1.15-5.52, P =0.021) and LT outside Milan criteria (HR=2.40, 95% CI: 1.16-4.92, P =0.018) were negative prognostic factors in transplanted patients. Inverse probability-weighted multivariate analyses revealed a distinct survival benefit after LT. Matched patients presented a median overall survival (OS) of 197 months (95% CI: 143-not reached) and a 73% 5-year OS after LT, and 119 months (95% CI: 74-133 months) and a 52.8% 5-year OS after LR (HR=0.59, 95% CI: 0.3-0.9, P =0.022). However, the survival benefit after LT was lost if patients were transplanted outside Milan criteria.
CONCLUSIONS
CONCLUSIONS
This multicentric study in patients with NELM demonstrates a survival benefit of LT over LR. This benefit depends on adherence to selection criteria, in particular low-grade tumor biology and Milan criteria, and must be balanced against potential risks of LT.
Identifiants
pubmed: 35975918
doi: 10.1097/SLA.0000000000005663
pii: 00000658-990000000-00231
doi:
Types de publication
Multicenter Study
Journal Article
Langues
eng
Sous-ensembles de citation
IM
Pagination
e1063-e1071Informations de copyright
Copyright © 2022 The Author(s). Published by Wolters Kluwer Health, Inc.
Déclaration de conflit d'intérêts
The authors report no conflicts of interest.
Références
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;3:1335–1342.
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;46:715–731.
Halfdanarson TR, Rabe KG, Rubin J, et al. Pancreatic neuroendocrine tumors (PNETs): incidence, prognosis and recent trend toward improved survival. Ann Oncol. 2008;19:1727–1733.
Chamberlain RS, Canes D, Brown KT, et al. Hepatic neuroendocrine metastases: does intervention alter outcomes? J Am Coll Surg. 2000;190:432–445.
House MG, Cameron JL, Lillemoe KD, et al. Differences in survival for patients with resectable versus unresectable metastases from pancreatic islet cell cancer. J Gastrointest Surg. 2006;10:138–145.
Modlin IM, Lye KD, Kidd M. A 5-decade analysis of 13,715 carcinoid tumors. Cancer. 2003;97:934–959.
Pavel M, Oberg K, Falconi M, et al. Gastroenteropancreatic neuroendocrine neoplasms: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2020;31:844–860.
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;103:172–185.
Shah MH, Goldner WS, Halfdanarson TR, et al. NCCN Guidelines Insights: Neuroendocrine and Adrenal Tumors, Version 2.2018. J Natl Compr Canc Netw. 2018;16:693–702.
Boudreaux JP, Klimstra DS, Hassan MM, et al. The NANETS consensus guideline for the diagnosis and management of neuroendocrine tumors: well-differentiated neuroendocrine tumors of the jejunum, ileum, appendix, and cecum. Pancreas. 2010;39:753–766.
Niederle B, Pape UF, Costa F, et al. ENETS Consensus Guidelines Update for Neuroendocrine Neoplasms of the Jejunum and Ileum. Neuroendocrinology. 2016;103:125–138.
Frilling A, Modlin IM, Kidd M, et al. Recommendations for management of patients with neuroendocrine liver metastases. Lancet Oncol. 2014;15:e8–e21.
Caplin ME, Pavel M, Cwikla JB, et al. Anti-tumour effects of lanreotide for pancreatic and intestinal neuroendocrine tumours: the CLARINET open-label extension study. Endocr Relat Cancer. 2016;23:191–199.
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;387:968–977.
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;21:1489–1499.
Pavel ME, Baudin E, Öberg KE, et al. Efficacy of everolimus plus octreotide LAR in patients with advanced neuroendocrine tumor and carcinoid syndrome: final overall survival from the randomized, placebo-controlled phase 3 RADIANT-2 study. Ann Oncol. 2017;28:1569–1575.
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;376:125–135.
Strosberg JR, Caplin ME, Kunz PL, et al. Lu-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;22:1752–1763.
Kulke MH, Anthony LB, Bushnell DL, et al. NANETS treatment guidelines: well-differentiated neuroendocrine tumors of the stomach and pancreas. Pancreas. 2010;39:735–752.
Kaltsas G, Caplin M, Davies P, et al. ENETS Consensus Guidelines for the Standards of Care in Neuroendocrine Tumors: pre- and perioperative therapy in patients with neuroendocrine tumors. Neuroendocrinology. 2017;105:245–254.
Mayo SC, de Jong MC, Pulitano C, et al. Surgical management of hepatic neuroendocrine tumor metastasis: results from an international multi-institutional analysis. Ann Surg Oncol. 2010;17:3129–3136.
Lesurtel M, Nagorney DM, Mazzaferro V, et al. When should a liver resection be performed in patients with liver metastases from neuroendocrine tumours? A systematic review with practice recommendations. HPB. 2015;17:17–22.
Moris D, Tsilimigras DI, Ntanasis-Stathopoulos I, et al. Liver transplantation in patients with liver metastases from neuroendocrine tumors: a systematic review. Surgery. 2017;162:525–536.
Elias D, Lefevre JH, Duvillard P, et al. Hepatic metastases from neuroendocrine tumors with a “thin slice” pathological examination: they are many more than you think. Ann Surg. 2010;251:307–310.
Mazzaferro V, Sposito C, Coppa J, et al. The long-term benefit of liver transplantation for hepatic metastases from neuroendocrine tumors. Am J Transplant. 2016;16:2892–2902.
Mazzaferro V, Pulvirenti A, Coppa J. Neuroendocrine tumors metastatic to the liver: how to select patients for liver transplantation? J Hepatol. 2007;47:460–466.
Gurusamy KS, Ramamoorthy R, Sharma D, et al. Liver resection versus other treatments for neuroendocrine tumours in patients with resectable liver metastases. Cochrane Database Syst Rev. 2009;2:CD007060.
Norlen O, Daskalakis K, Oberg K, et al. Indication for liver transplantation in young patients with small intestinal NETs is rare? World J Surg. 2014;38:742–747.
Schreckenbach T, Hübert H, Koch C, et al. Surgical resection of neuroendocrine tumor liver metastases as part of multimodal treatment strategies: a propensity score matching analysis. Eur J Surg Oncol. 2019;45:808–815.
Chakedis J, Beal EW, Lopez-Aguiar AG, et al. Surgery provides long-term survival in patients with metastatic neuroendocrine tumors undergoing resection for non-hormonal symptoms. J Gastrointest Surg. 2019;23:122–134.
Frilling A, Li J, Malamutmann E, et al. Treatment of liver metastases from neuroendocrine tumours in relation to the extent of hepatic disease. Br J Surg. 2009;96:175–184.
Eshmuminov D, Raptis DA, Linecker M, et al. Meta-analysis of associating liver partition with portal vein ligation and portal vein occlusion for two-stage hepatectomy. Br J Surg. 2016;103:1768–1782.
Linecker M, Kambakamba P, Raptis DA, et al. ALPPS in neuroendocrine liver metastases not amenable for conventional resection—lessons learned from an interim analysis of the International ALPPS Registry. HPB (Oxford). 2020;22:537–544.
Lerut J, Iesari S, Vandeplas G, et al. Secondary non-resectable liver tumors: a single-center living-donor and deceased-donor liver transplantation case series. Hepatobiliary Pancreat Dis Int. 2019;18:412–422.
Le Treut YP, Gregoire E, Klempnauer J, et al. Liver transplantation for neuroendocrine tumors in Europe—results and trends in patient selection: a 213-case European liver transplant registry study. Ann Surg. 2013;257:807–815.
Rindi G, Klersy C, Albarello L, et al. Competitive Testing of the WHO 2010 versus the WHO 2017 Grading of Pancreatic Neuroendocrine Neoplasms: data from a large international cohort study. Neuroendocrinology. 2018;107:375–386.
Inzani F, Petrone G, Rindi G. The New World Health Organization Classification for Pancreatic Neuroendocrine Neoplasia. Endocrinol Metab Clin North Am. 2018;47:463–470.
Ricci C, Casadei R, Taffurelli G, et al. Validation of the 2010 WHO classification and a new prognostic proposal: a single centre retrospective study of well-differentiated pancreatic neuroendocrine tumours. Pancreatology. 2016;16:403–410.
Nuñez-Valdovinos B, Carmona-Bayonas A, Jimenez-Fonseca P, et al. Neuroendocrine tumor heterogeneity adds uncertainty to the World Health Organization 2010 Classification: Real-World Data from the Spanish Tumor Registry (R-GETNE). Oncologist. 2018;23:422–432.
Auernhammer CJ, Spitzweg C, Angele MK, et al. Advanced neuroendocrine tumours of the small intestine and pancreas: clinical developments, controversies, and future strategies. Lancet Diabetes Endocrinol. 2018;6:404–415.
Yao JC, Pavel M, Lombard-Bohas C, et al. Everolimus for the treatment of advanced pancreatic neuroendocrine tumors: overall survival and circulating biomarkers from the randomized, phase III RADIANT-3 study. J Clin Oncol. 2016;34:3906–3913.
Raymond E, Dahan L, Raoul JL, et al. Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. N Engl J Med. 2011;364:501–513.
Strosberg JR, Fine RL, Choi J, et al. First-line chemotherapy with capecitabine and temozolomide in patients with metastatic pancreatic endocrine carcinomas. Cancer. 2011;117:268–275.
Ramage J, Naraev BG, Halfdanarson TR. Peptide receptor radionuclide therapy for patients with advanced pancreatic neuroendocrine tumors. Semin Oncol. 2018;45:236–248.
Starr JS, Sonbol MB, Hobday TJ, et al. Peptide receptor radionuclide therapy for the treatment of pancreatic neuroendocrine tumors: recent insights. Onco Targets Ther. 2020;13:3545–3555.
Parghane RV, Bhandare M, Chaudhari V, et al. Surgical feasibility, determinants, and overall efficacy of neoadjuvant. J Nucl Med. 2021;62:1558–1563.
Schnitzbauer AA, Filmann N, Adam R, et al. mTOR inhibition is most beneficial after liver transplantation for hepatocellular carcinoma in patients with active tumors. Ann Surg. 2020;272:855–862.
Lerut J, Mathys J, Verbaandert C, et al. Tacrolimus monotherapy in liver transplantation: one-year results of a prospective, randomized, double-blind, placebo-controlled study. Ann Surg. 2008;248:956–967.
Line PD, Dueland S. Liver transplantation for secondary liver tumours: the difficult balance between survival and recurrence. J Hepatol. 2020;73:1557–1562.
Broering DC, Schulte am Esch J, Fischer L, et al. Split liver transplantation. HPB (Oxford). 2004;6:76–82.
Nobel YR, Goldberg DS. Variable use of model for end-stage liver disease exception points in patients with neuroendocrine tumors metastatic to the liver and its impact on patient outcomes. Transplantation. 2015;99:2341–2346.
The New York Times. A transplant that is raising many questions; 2009. Available at: https://www.nytimes.com/2009/06/23/business/23liver.html?_r=1&adxnnl=1&pagewanted=all&adxnnlx=1411048884-Sv9SQvWJ1y4EzJnaQvuX3w . Accessed April 4, 2022.