Advances in the Diagnosis and Therapeutic Management of Gastroenteropancreatic Neuroendocrine Neoplasms (GEP-NENs)
Abstract
:Simple Summary
Abstract
1. Introduction
1.1. Epidemiology
1.2. Classification
1.3. Prognosis
2. Laboratory Diagnostics
2.1. NETest
2.2. NETest vs. CgA
2.3. NETest vs. Imaging
2.4. NETest vs. PRRT Predictor Quotient (PPQ)
2.5. HOX Gene Expression Analysis
2.6. Epigenome Analysis
2.7. Histopathological Findings
3. Imaging Tests
3.1. Morphological Imaging
3.2. Nuclear Medicine
4. Systemic Treatment
4.1. Somatostatin Analogs
4.2. INF Alpha
4.3. Radionuclide Treatment
4.4. Transarterial Treatment
4.5. Targeted Therapy
4.6. Immunotherapy
4.7. CAR-T-Cell Therapy
4.8. Bispecific Antibodies
4.9. Vaccines
4.10. Cytotoxic Chemotherapy of Highly Differentiated G1 and G2 GEP NETs
4.11. Cytotoxic Chemotherapy of Low-Differentiated G3 GEP NETs and GEP NECs
5. Surgery
5.1. Stomach (Gastric NETs, G-NETs)
- Type I most commonly manifests as small multiple, low-grade tumors [32]; they account for 70–80% of G-NET patients, and are more frequent in females. It is histologically composed of enterochromaffin-like cells [122]. Small tumors should be surveilled or resected yearly, depending on the size and depth of invasion [32,123]. According to ENETS guidelines from 2016, resection is recommended for lesions equal or above 1 cm, and those for which infiltration of the mucosa propria is anticipated. If feasible, resection could be performed using EMR (endoscopic mucosal resection) or preferred ESD (endoscopic submucosal dissection). In case of positive margins or T2 stage, local excision or partial gastrectomy should be considered [123].
- Type III is usually diagnosed in late lesions, which require partial or complete gastrectomy with lymph nodes dissection [123], while the treatment of tumors below 2 cm is controversial and may be considered in a narrow range of patients [62]. In small tumors, endoscopic resection is recommended [123].
5.2. Duodenum (D-NETs)
5.3. Pancreas (PanNETs)
5.4. Small Intestine (SI-NETs, SB-NETs)
5.5. Appendix (Appendiceal NETs, A-NETs)
5.6. Colon (C-NETs)
5.7. Rectum (R-NETs)
5.8. Liver
6. Radio Guided Surgery
7. Advances in Minimally Invasive Surgery
8. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
Abbreviations
NENs | neuroendocrine neoplasms |
GEP-NETs | gastroenteropancreatic neuroendocrine tumors |
IARC-WHO | International Agency for Research on Cancer—World Health Organization |
NECs | neuroendocrine cancers |
MiNEN | mixed neuroendocrine-nonneuroendocrine neoplasms |
MEN | multiple endocrine neoplasia |
SINET | small intestine |
PanNET | pancreatic neuroendocrine tumor |
RECIST | response evaluation criteria in solid tumors |
CT | computed tomography |
US | ultrasonography |
MRI | magnetic resonance imaging |
PRRT | peptide receptor radionuclide therapy |
PPQ | Predictor quotient |
CI | confidence interval |
ISH | in situ hybridization |
CTC | circulating tumor cells |
EpNET | extrapancreatic neuroendocrine tumor |
ER | endoscopic resection |
HR | hazard ratio |
LAR | long-acting release |
mOS | median overall survival |
mPFS | median progression-free survival |
ORS | objective response rate |
PET | positron emitting radionuclide |
PFS | progression-free survival |
RR | response rate |
SPECT | gamma emitting radionuclide |
SSA | somatostatin analogs |
SSTR | somatostatin receptor |
References
- Pavel, M.; Öberg, K.; Falconi, M.; Krenning, E.P.; Sundin, A.; Perren, A.; Berruti, A. Gastroenteropancreatic neuroendocrine neoplasms: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 2020, 31, 844–860. [Google Scholar] [CrossRef] [PubMed]
- Kos-Kudła, B.; Blicharz-Dorniak, J.; Strzelczyk, J.; Bałdys-Waligórska, A.; Bednarczuk, T.; Bolanowski, M.; Boratyn-Nowicka, A.; Borowska, M.; Cichocki, A.; Ćwikła, J.B.; et al. Diagnostic and therapeutic guidelines for gastro-entero-pancreatic neuroendocrine neoplasms (recommended by the Polish Network of Neuroendocrine Tumours). Endokrynol. Pol. 2017, 68, 79–110. [Google Scholar] [CrossRef] [PubMed]
- Das, S.; Dasari, A. Epidemiology, incidence, and prevalence of neuroendocrine neoplasms: Are there global differences? Curr. Oncol. Rep. 2021, 23, 43. [Google Scholar] [CrossRef] [PubMed]
- Cetinkaya, R.B.; Aagnes, B.; Thiis-Evensen, E.; Tretli, S.; Bergestuen, D.S.; Hansen, S. Trends in incidence of neuroendocrine neoplasms in Norway: A report of 16,075 cases from 1993 through 2010. Neuroendocrinology 2017, 104, 1–10. [Google Scholar] [CrossRef] [Green Version]
- Wyld, D.; Wan, M.H.; Moore, J.; Dunn, N.; Youl, P. Epidemiological trends of neuroendocrine tumours over three decades in Queensland, Australia. Cancer Epidemiol. 2019, 63, 101598. [Google Scholar] [CrossRef]
- Xu, Z.; Wang, L.; Dai, S.; Chen, M.; Li, F.; Sun, J.; Luo, F. Epidemiologic trends of and factors associated with overall survival for patients with gastroenteropancreatic neuroendocrine tumors in the United States. JAMA Netw. Open 2021, 4, e2124750. [Google Scholar] [CrossRef]
- Dasari, A.; Shen, C.; Halperin, D.; Zhao, B.; Zhou, S.; Xu, Y.; Shih, T.; Yao, J.C. Trends in the incidence, prevalence, and survival outcomes in patients with neuroendocrine tumors in the United States. JAMA Oncol. 2017, 3, 1335–1342. [Google Scholar] [CrossRef]
- Massironi, S.; Sciola, V.; Peracchi, M.; Ciafardini, C.; Spampatti, M.P.; Conte, D. Neuroendocrine tumors of the gastro-entero-pancreatic system. World J. Gastroenterol. 2008, 14, 5377–5384. [Google Scholar] [CrossRef]
- Frilling, A.; Akerström, G.; Falconi, M.; Pavel, M.; Ramos, J.; Kidd, M.; Modlin, I.M. Neuroendocrine tumor disease: An evolving landscape. Endocr. Relat. Cancer 2012, 19, R163–R185. [Google Scholar] [CrossRef] [Green Version]
- Takayanagi, D.; Cho, H.; Machida, E.; Kawamura, A.; Takashima, A.; Wada, S.; Tsunoda, T.; Kohno, T.; Shiraishi, K. Update on Epidemiology, Diagnosis, and Biomarkers in Gastroenteropancreatic Neuroendocrine Neoplasms. Cancers 2022, 14, 1119. [Google Scholar] [CrossRef]
- Nagtegaal, I.D.; Odze, R.D.; Klimstra, D.; Paradis, V.; Rugge, M.; Schirmacher, P.; Washington, K.M.; Carneiro, F.; Cree, I.A. The 2019 WHO classification of tumours of the digestive system. Histopathology 2020, 76, 182–188. [Google Scholar] [CrossRef] [Green Version]
- Bałdys-Waligórska, A.; Nowak, A. Neuroendocrine neoplasms of the digestive system—Current classification and terminology. Nowotw. J. Oncol. 2021, 71, 26–37. [Google Scholar] [CrossRef]
- Crona, J.; Skogseid, B. Genetics of neuroendocrine tumors. Eur. J. Endocrinol. 2016, 174, R275–R290. [Google Scholar] [CrossRef] [Green Version]
- Noor, M.; Huber, A.R.; Cates, J.M.M.; Gonzalez, R.S. Risk factors for progression of appendiceal neuroendocrine tumours: Low-stage tumours <5 mm appear to be overwhelmingly indolent and may merit a separate designation. Histopathology 2021, 79, 416–426. [Google Scholar] [CrossRef]
- Cloyd, J.M.; Poultsides, G.A. Non-functional neuroendocrine tumors of the pancreas: Advances in diagnosis and management. World J. Gastroenterol. 2015, 21, 9512–9525. [Google Scholar] [CrossRef]
- Liu, E.; Paulson, S.; Gulati, A.; Freudman, J.; Grosh, W.; Kafer, S.; Wickremesinghe, P.C.; Salem, R.R.; Bodei, L. Assessment of NETest clinical utility in a U.S. registry-based study. Oncologist 2019, 24, 783–790. [Google Scholar] [CrossRef] [Green Version]
- Modlin, I.M.; Kidd, M.; Bodei, L.; Malczewska, A.; Kitz, A.; Oberg, K. Circulating biomarkers of gastroenteropancreatic and lung neuroendocrine neoplasms: “The times they are a changin”. Curr. Opin. Endocr. Metab. Res. 2021, 18, 243–253. [Google Scholar] [CrossRef]
- Koch, C.; Koca, E.; Filmann, N.; Husmann, G.; Bojunga, J. Time from first tumor manifestation to diagnosis in patients with GEP-NET: Results from a large German tertiary referral center. Medicine 2021, 100, e27276. [Google Scholar] [CrossRef]
- van Treijen, M.J.C.; van der Zee, D.; Heeres, B.C.; Staal, F.C.R.; Vriens, M.R.; Saveur, L.J.; Verbeek, W.H.M.; Korse, C.M.; Maas, M.; Valk, G.D.; et al. Blood molecular genomic analysis predicts the disease course of gastroenteropancreatic neuroendocrine tumor patients: A validation study of the predictive value of the NETest®. Neuroendocrinology 2021, 111, 586–598. [Google Scholar] [CrossRef]
- van Treijen, M.J.C.; Korse, C.M.; van Leeuwaarde, R.S.; Saveur, L.J.; Vriens, M.R.; Verbeek, W.H.M.; Tesselaar, M.E.T.; Valk, G.D. Blood transcript profiling for the detection of neuroendocrine tumors: Results of a large independent validation study. Front. Endocrinol. 2018, 9, 740. [Google Scholar] [CrossRef]
- Di Giacinto, P.; Rota, F.; Rizza, L.; Campana, D.; Isidori, A.; Lania, A.; Lenzi, A.; Zuppi, P.; Baldelli, R. Chromogranin A: From laboratory to clinical aspects of patients with neuroendocrine tumors. Int. J. Endocrinol. 2018, 2018, 8126087. [Google Scholar] [CrossRef]
- Malczewska, A.; Witkowska, M.; Makulik, K.; Bocian, A.; Walter, A.; Pilch-Kowalczyk, J.; Zajęcki, W.; Bodei, L.; Oberg, K.E.; Kos-Kudła, B. NETest liquid biopsy is diagnostic of small intestine and pancreatic neuroendocrine tumors and correlates with imaging. Endocr. Connect. 2019, 8, 442–453. [Google Scholar] [CrossRef] [Green Version]
- Bodei, L.; Kidd, M.S.; Singh, A.; van der Zwan, W.A.; Severi, S.; Drozdov, I.A.; Malczewska, A.; Baum, R.P.; Kwekkeboom, D.J.; Paganelli, G.; et al. PRRT neuroendocrine tumor response monitored using circulating transcript analysis: The NETest. Eur. J. Nucl. Med. Mol. Imaging 2020, 47, 895–906. [Google Scholar] [CrossRef]
- Rinke, A.; Auernhammer, C.J.; Bodei, L.; Kidd, M.; Krug, S.; Lawlor, R.; Marinoni, I.; Perren, A.; Scarpa, A.; Sorbye, H.; et al. Treatment of advanced gastroenteropancreatic neuroendocrine neoplasia, are we on the way to personalised medicine? Gut 2021, 70, 1768–1781. [Google Scholar] [CrossRef]
- Öberg, K.; Califano, A.; Strosberg, J.R.; Ma, S.; Pape, U.; Bodei, L.; Kaltsas, G.; Toumpanakis, C.; Goldenring, J.R.; Frilling, A.; et al. A meta-analysis of the accuracy of a neuroendocrine tumor mRNA genomic biomarker (NETest) in blood. Ann. Oncol. 2020, 31, 202–212. [Google Scholar] [CrossRef] [Green Version]
- Di Mauro, A.; Scognamiglio, G.; Aquino, G.; Cerrone, M.; Liguori, G.; Clemente, O.; Di Bonito, M.; Cantile, M.; Botti, G.; Tafuto, S.; et al. Aberrant expression of long non coding RNA HOTAIR and de-regulation of the paralogous 13 HOX genes are strongly associated with aggressive behavior of gastro-entero-pancreatic neuroendocrine tumors. Int. J. Mol. Sci. 2021, 22, 7049. [Google Scholar] [CrossRef]
- Holmager, P.; Langer, S.W.; Federspiel, B.; Willemoe, G.L.; Garbyal, R.S.; Melchior, L.; Klose, M.; Kjaer, A.; Hansen, C.P.; Andreassen, M.; et al. Increase of Ki-67 index and influence on mortality in patients with neuroendocrine neoplasms. J. Neuroendocrinol. 2021, 33, e13018. [Google Scholar] [CrossRef]
- Jamali, M.; Chetty, R. Predicting prognosis in gastroentero-pancreatic neuroendocrine tumors: An overview and the value of Ki-67 immunostaining. Endocr. Pathol. 2008, 19, 282–288. [Google Scholar] [CrossRef] [PubMed]
- Janson, E.T.; Knigge, U.; Dam, G.; Federspiel, B.; Grønbaek, H.; Stålberg, P.; Langer, S.W.; Kjaer, A.; Arola, J.; Schalin-Jäntti, C.; et al. Nordic guidelines 2021 for diagnosis and treatment of gastroenteropancreatic neuroendocrine neoplasms. Acta Oncol. 2021, 60, 931–941. [Google Scholar] [CrossRef] [PubMed]
- Półtorak-Szymczak, G.; Budlewski, T.; Furmanek, M.I.; Wierzba, W.; Sklinda, K.; Walecki, J.; Mruk, B. Radiological imaging of gastro-entero-pancreatic neuroendocrine tumors. The review of current literature emphasizing the diagnostic value of chosen imaging methods. Front. Oncol. 2021, 11, 670233. [Google Scholar] [CrossRef] [PubMed]
- Zilli, A.; Arcidiacono, P.G.; Conte, D.; Massironi, S. Clinical impact of endoscopic ultrasonography on the management of neuroendocrine tumors: Lights and shadows. Dig. Liver Dis. 2018, 50, 6–14. [Google Scholar] [CrossRef] [Green Version]
- Andreasi, V.; Partelli, S.; Muffatti, F.; Manzoni, M.F.; Capurso, G.; Falconi, M. Update on gastroenteropancreatic neuroendocrine tumors. Dig. Liver Dis. 2021, 53, 171–182. [Google Scholar] [CrossRef]
- Mikulová, M.B.; Mikuš, P. Advances in development of radiometal labeled amino acid-based compounds for cancer imaging and diagnostics. Pharmaceuticals 2021, 14, 167. [Google Scholar] [CrossRef]
- Hu, Y.; Ye, Z.; Wang, F.; Qin, Y.; Xu, X.; Yu, X.; Ji, S. Role of somatostatin receptor in pancreatic neuroendocrine tumor development, diagnosis, and therapy. Front. Endocrinol. 2021, 12, 679000. [Google Scholar] [CrossRef]
- Papadakis, G.Z.; Karantanas, A.H.; Marias, K.; Millo, C. Current status and future prospects of PET-imaging applications in patients with gastro-entero-pancreatic neuroendocrine tumors (GEP-NETs). Eur. J. Radiol. 2021, 143, 109932. [Google Scholar] [CrossRef]
- Kanzaki, T.; Takahashi, Y.; Higuchi, T.; Zhang, X.; Mogi, N.; Suto, T.; Tsushima, Y. Evaluation of a correction method for (111)In-pentetreotide SPECT imaging of gastroenteropancreatic neuroendocrine tumors. J. Nucl. Med. Technol. 2020, 48, 326–330. [Google Scholar] [CrossRef]
- Chan, D.L.; Pavlakis, N.; Schembri, G.P.; Bernard, E.J.; Hsiao, E.; Hayes, A.; Barnes, T.; Diakos, C.; Khasraw, M.; Samra, J.; 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, 1149–1158. [Google Scholar] [CrossRef]
- Karfis, I.; Marin, G.; Levillain, H.; Drisis, S.; Muteganya, R.; Critchi, G.; Taraji-Schiltz, L.; Guix, C.A.; Shaza, L.; Elbachiri, M.; et al. Prognostic value of a three-scale grading system based on combining molecular imaging with (68)Ga-DOTATATE and (18)F-FDG PET/CT in patients with metastatic gastroenteropancreatic neuroendocrine neoplasias. Oncotarget 2020, 11, 589–599. [Google Scholar] [CrossRef] [Green Version]
- Yao, J.; Strosberg, J.; Fazio, N.; Pavel, M.; Ruszniewski, P.; Bergsland, E.; Li, D.; Tafuto, S.; Raj, N.; Campana, D. Activity & safety of spartalizumab (PDR001) in patients (pts) with advanced neuroendocrine tumors (NET) of pancreatic (Pan), gastrointestinal (GI), or thoracic (T) origin, & gastroenteropancreatic neuroendocrine carcinoma (GEP NEC) who have progressed on prior treatment (Tx). Ann. Oncol. 2018, 29, viii467–viii468. [Google Scholar] [CrossRef]
- Zhang, J.; Liu, Q.; Singh, A.; Schuchardt, C.; Kulkarni, H.R.; Baum, R.P. Prognostic value of (18)F-FDG PET/CT in a large cohort of patients with advanced metastatic neuroendocrine neoplasms treated with peptide receptor radionuclide therapy. J. Nucl. Med. 2020, 61, 1560–1569. [Google Scholar] [CrossRef]
- Delpassand, E.S.; Ranganathan, D.; Wagh, N.; Shafie, A.; Gaber, A.; Abbasi, A.; Kjaer, A.; Tworowska, I.; Núñez, R. (64)Cu-DOTATATE PET/CT for imaging patients with known or suspected somatostatin receptor-positive neuroendocrine tumors: Results of the first U.S. prospective, reader-masked clinical trial. J. Nucl. Med. 2020, 61, 890–896. [Google Scholar] [CrossRef]
- Johnbeck, C.B.; Knigge, U.; Loft, A.; Berthelsen, A.K.; Mortensen, J.; Oturai, P.; Langer, S.W.; Elema, D.R.; Kjaer, A. Head-to-head comparison of (64)Cu-DOTATATE and (68)Ga-DOTATOC PET/CT: A prospective study of 59 patients with neuroendocrine tumors. J. Nucl. Med. 2017, 58, 451–457. [Google Scholar] [CrossRef] [Green Version]
- Nicolas, G.P.; Schreiter, N.; Kaul, F.; Uiters, J.; Bouterfa, H.; Kaufmann, J.; Erlanger, T.E.; Cathomas, R.; Christ, E.; Fani, M.; et al. Sensitivity comparison of (68)Ga-OPS202 and (68)Ga-DOTATOC PET/CT in patients with gastroenteropancreatic neuroendocrine tumors: A prospective phase II imaging study. J. Nucl. Med. 2018, 59, 915–921. [Google Scholar] [CrossRef] [Green Version]
- Xie, Q.; Liu, T.; Ding, J.; Zhou, N.; Meng, X.; Zhu, H.; Li, N.; Yu, J.; Yang, Z. Synthesis, preclinical evaluation, and a pilot clinical imaging study of [(18)F]AlF-NOTA-JR11 for neuroendocrine neoplasms compared with [(68)Ga]Ga-DOTA-TATE. Eur. J. Nucl. Med. Mol. Imaging 2021, 48, 3129–3140. [Google Scholar] [CrossRef]
- Antwi, K.; Fani, M.; Heye, T.; Nicolas, G.; Rottenburger, C.; Kaul, F.; Merkle, E.; Zech, C.J.; Boll, D.; Vogt, D.R.; et al. Comparison of glucagon-like peptide-1 receptor (GLP-1R) PET/CT, SPECT/CT and 3T MRI for the localisation of occult insulinomas: Evaluation of diagnostic accuracy in a prospective crossover imaging study. Eur. J. Nucl. Med. Mol. Imaging 2018, 45, 2318–2327. [Google Scholar] [CrossRef]
- Kim, Y.I.; Yoo, C.; Oh, S.J.; Lee, S.J.; Kang, J.; Hwang, H.S.; Hong, S.M.; Ryoo, B.Y.; Ryu, J.S. Tumour-to-liver ratio determined by [(68)Ga]Ga-DOTA-TOC PET/CT as a prognostic factor of lanreotide efficacy for patients with well-differentiated gastroenteropancreatic-neuroendocrine tumours. EJNMMI Res. 2020, 10, 63. [Google Scholar] [CrossRef]
- Lee, H.; Eads, J.R.; Pryma, D.A. (68)Ga-DOTATATE positron emission tomography-computed tomography quantification predicts response to somatostatin analog therapy in gastroenteropancreatic neuroendocrine tumors. Oncologist 2021, 26, 21–29. [Google Scholar] [CrossRef]
- Rinke, A.; Müller, H.H.; Schade-Brittinger, C.; Klose, K.J.; Barth, P.; Wied, M.; Mayer, C.; Aminossadati, B.; Pape, U.F.; Bläker, M.; et al. Placebo-controlled, double-blind, prospective, randomized study on the effect of octreotide LAR in the control of tumor growth in patients with metastatic neuroendocrine midgut tumors: A report from the PROMID Study Group. J. Clin. Oncol. 2009, 27, 4656–4663. [Google Scholar] [CrossRef] [PubMed]
- Caplin, M.E.; Pavel, M.; Ćwikła, J.B.; Phan, A.T.; Raderer, M.; Sedláčková, E.; Cadiot, G.; Wolin, E.M.; Capdevila, J.; Wall, L.; et al. Lanreotide in metastatic enteropancreatic neuroendocrine tumors. N. Engl. J. Med. 2014, 371, 224–233. [Google Scholar] [CrossRef] [PubMed]
- Pavel, M.E.; Ćwikła, J.; Lombard-Bohas, C.; Borbath, I.; Shah, T.; Pape, U.; Thanh, X.T.; Houchard, A.; Ruszniewski, P. Efficacy and safety of lanreotide autogel (LAN) 120 mg every 14 days in progressive pancreatic or midgut neuroendocrine tumours (NETs): CLARINET FORTE study results. Ann. Oncol. 2020, 31, S711–S724. [Google Scholar] [CrossRef]
- Oberg, K.; Kvols, L.; Caplin, M.; Delle Fave, G.; de Herder, W.; Rindi, G.; Ruszniewski, P.; Woltering, E.A.; Wiedenmann, B. Consensus report on the use of somatostatin analogs for the management of neuroendocrine tumors of the gastroenteropancreatic system. Ann. Oncol. 2004, 15, 966–973. [Google Scholar] [CrossRef]
- Rinke, A.; Krug, S. Neuroendocrine tumours—Medical therapy: Biological. Best Pract. Res. Clin. Endocrinol. Metab. 2016, 30, 79–91. [Google Scholar] [CrossRef]
- Yao, J.C.; Guthrie, K.A.; Moran, C.; Strosberg, J.R.; Kulke, M.H.; Chan, J.A.; LoConte, N.; McWilliams, R.R.; Wolin, E.M.; Mattar, B.; 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, 1695–1703. [Google Scholar] [CrossRef]
- Pavel, M.; Gross, D.J.; Benavent, M.; Perros, P.; Srirajaskanthan, R.; Warner, R.R.P.; Kulke, M.H.; Anthony, L.B.; Kunz, P.L.; Hörsch, D.; et al. Telotristat ethyl in carcinoid syndrome: Safety and efficacy in the TELECAST phase 3 trial. Endocr. Relat. Cancer 2018, 25, 309–322. [Google Scholar] [CrossRef] [Green Version]
- Kwekkeboom, D.J.; de Herder, W.W.; Kam, B.L.; van Eijck, C.H.; van Essen, M.; Kooij, P.P.; Feelders, R.A.; van Aken, M.O.; Krenning, E.P. Treatment with the radiolabeled somatostatin analog [177 Lu-DOTA 0,Tyr3]octreotate: Toxicity, efficacy, and survival. J. Clin. Oncol. 2008, 26, 2124–2130. [Google Scholar] [CrossRef] [Green Version]
- Kwekkeboom, D.J.; Teunissen, J.J.; Bakker, W.H.; Kooij, P.P.; de Herder, W.W.; Feelders, R.A.; van Eijck, C.H.; Esser, J.P.; Kam, B.L.; Krenning, E.P. Radiolabeled somatostatin analog [177Lu-DOTA0,Tyr3]octreotate in patients with endocrine gastroenteropancreatic tumors. J. Clin. Oncol. 2005, 23, 2754–2762. [Google Scholar] [CrossRef] [Green Version]
- Brabander, T.; Teunissen, J.J.; Van Eijck, C.H.; Franssen, G.J.; Feelders, R.A.; de Herder, W.W.; Kwekkeboom, D.J. Peptide receptor radionuclide therapy of neuroendocrine tumours. Best Pract. Res. Clin. Endocrinol. Metab. 2016, 30, 103–114. [Google Scholar] [CrossRef]
- Strosberg, J.; El-Haddad, G.; Wolin, E.; Hendifar, A.; Yao, J.; Chasen, B.; Mittra, E.; Kunz, P.L.; Kulke, M.H.; Jacene, H.; et al. Phase 3 trial of (177)Lu-Dotatate for midgut neuroendocrine tumors. N. Engl. J. Med. 2017, 376, 125–135. [Google Scholar] [CrossRef]
- Brabander, T.; van der Zwan, W.A.; Teunissen, J.J.M.; Kam, B.L.R.; Feelders, R.A.; de Herder, W.W.; van Eijck, C.H.J.; Franssen, G.J.H.; Krenning, E.P.; Kwekkeboom, D.J. Long-term efficacy, survival, and safety of [(177)Lu-DOTA(0),Tyr(3)]octreotate in patients with gastroenteropancreatic and bronchial neuroendocrine tumors. Clin. Cancer Res. 2017, 23, 4617–4624. [Google Scholar] [CrossRef] [Green Version]
- Bodei, L.; Kwekkeboom, D.J.; Kidd, M.; Modlin, I.M.; Krenning, E.P. Radiolabeled somatostatin analogue therapy of gastroenteropancreatic cancer. Semin. Nucl. Med. 2016, 46, 225–238. [Google Scholar] [CrossRef]
- Yao, J.C.; Strosberg, J.; Fazio, N.; Pavel, M.E.; Bergsland, E.; Ruszniewski, P.; Halperin, D.M.; Li, D.; Tafuto, S.; Raj, N.; et al. Spartalizumab in metastatic, well/poorly-differentiated neuroendocrine neoplasms. Endocr. Relat. Cancer 2021, 28, 161–172. [Google Scholar] [CrossRef]
- de Mestier, L.; Lepage, C.; Baudin, E.; Coriat, R.; Courbon, F.; Couvelard, A.; Do Cao, C.; Frampas, E.; Gaujoux, S.; Gincul, R.; et al. Digestive neuroendocrine neoplasms (NEN): French intergroup clinical practice guidelines for diagnosis, treatment and follow-up (SNFGE, GTE, RENATEN, TENPATH, FFCD, GERCOR, UNICANCER, SFCD, SFED, SFRO, SFR). Dig. Liver Dis. 2020, 52, 473–492. [Google Scholar] [CrossRef]
- Wild, D.; Fani, M.; Fischer, R.; Del Pozzo, L.; Kaul, F.; Krebs, S.; Fischer, R.; Rivier, J.E.; Reubi, J.C.; Maecke, H.R.; et al. Comparison of somatostatin receptor agonist and antagonist for peptide receptor radionuclide therapy: A pilot study. J. Nucl. Med. 2014, 55, 1248–1252. [Google Scholar] [CrossRef] [Green Version]
- Lindner, T.; Loktev, A.; Altmann, A.; Giesel, F.; Kratochwil, C.; Debus, J.; Jäger, D.; Mier, W.; Haberkorn, U. Development of quinoline-based theranostic ligands for the targeting of fibroblast activation protein. J. Nucl. Med. 2018, 59, 1415–1422. [Google Scholar] [CrossRef] [Green Version]
- Hicks, R.J.; Kwekkeboom, D.J.; Krenning, E.; Bodei, L.; Grozinsky-Glasberg, S.; Arnold, R.; Borbath, I.; Cwikla, J.; Toumpanakis, C.; Kaltsas, G.; et al. ENETS consensus guidelines for the standards of care in neuroendocrine neoplasia: Peptide receptor radionuclide therapy with radiolabeled somatostatin analogues. Neuroendocrinology 2017, 105, 295–309. [Google Scholar] [CrossRef]
- Tian, R.; Jacobson, O.; Niu, G.; Kiesewetter, D.O.; Wang, Z.; Zhu, G.; Ma, Y.; Liu, G.; Chen, X. Evans blue attachment enhances somatostatin receptor subtype-2 imaging and radiotherapy. Theranostics 2018, 8, 735–745. [Google Scholar] [CrossRef]
- Howe, J.R.; Merchant, N.B.; Conrad, C.; Keutgen, X.M.; Hallet, J.; Drebin, J.A.; Minter, R.M.; Lairmore, T.C.; Tseng, J.F.; Zeh, H.J.; et al. The North American neuroendocrine tumor society consensus paper on the surgical management of pancreatic neuroendocrine tumors. Pancreas 2020, 49, 1–33. [Google Scholar] [CrossRef]
- Pavlakis, N.; Ransom, D.T.; Wyld, D.; Sjoquist, K.M.; Asher, R.; Gebski, V.; Wilson, K.; Kiberu, A.D.; Burge, M.E.; Macdonald, W.; et al. Australasian gastrointestinal trials group (AGITG) CONTROL NET study: Phase II study evaluating the activity of 177Lu-Octreotate peptide receptor radionuclide therapy (LuTate PRRT) and capecitabine, temozolomide CAPTEM)—First results for pancreas and updated midgut neuroendocrine tumors (pNETS, mNETS). J. Clin. Oncol. 2020, 38, 4608. [Google Scholar] [CrossRef]
- Aljubran, A.H.; Alrowaili, M.; Raef, H.; Bazarbashi, S.; Alzahrani, A.M.; Almuhaideb, A.; Almanea, H.; Badran, A.A.; Al-Dalee, A.; Tuli, M. Combination of everolimus and lu-177 PRRT in treatment of G1-2 neuroendocrine tumors (NET): Phase 1-2 study. J. Clin. Oncol. 2019, 37, 386. [Google Scholar] [CrossRef]
- Putzer, D.; Schullian, P.; Jaschke, W.; Bale, R. NEN: Advancement in diagnosis and minimally invasive therapy. Rofo 2020, 192, 422–430. [Google Scholar] [CrossRef] [PubMed]
- Touloupas, C.; Faron, M.; Hadoux, J.; Deschamps, F.; Roux, C.; Ronot, M.; Yevich, S.; Joskin, J.; Gelli, M.; Barbé, R.; et al. Long term efficacy and assessment of tumor response of transarterial chemoembolization in neuroendocrine liver metastases: A 15-year monocentric experience. Cancers 2021, 13, 5366. [Google Scholar] [CrossRef] [PubMed]
- Chan, J.; Kulke, M. Targeting the mTOR signaling pathway in neuroendocrine tumors. Curr. Treat. Options Oncol. 2014, 15, 365–379. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pavel, M.; Valle, J.W.; Eriksson, B.; Rinke, A.; Caplin, M.; Chen, J.; Costa, F.; Falkerby, J.; Fazio, N.; Gorbounova, V.; et al. ENETS consensus guidelines for the standards of care in neuroendocrine neoplasms: Systemic therapy—Biotherapy and novel targeted agents. Neuroendocrinology 2017, 105, 266–280. [Google Scholar] [CrossRef] [PubMed]
- Yao, J.C.; Shah, M.H.; Ito, T.; Bohas, C.L.; Wolin, E.M.; Van Cutsem, E.; Hobday, T.J.; Okusaka, T.; Capdevila, J.; de Vries, E.G.; et al. Everolimus for advanced pancreatic neuroendocrine tumors. N. Engl. J. Med. 2011, 364, 514–523. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yao, J.C.; Fazio, N.; Singh, S.; Buzzoni, R.; Carnaghi, C.; Wolin, E.; Tomasek, J.; Raderer, M.; Lahner, H.; Voi, M.; 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. [Google Scholar] [CrossRef]
- Raymond, E.; Dahan, L.; Raoul, J.L.; Bang, Y.J.; Borbath, I.; Lombard-Bohas, C.; Valle, J.; Metrakos, P.; Smith, D.; Vinik, A.; et al. Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. N. Engl. J. Med. 2011, 364, 501–513. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xu, J.; Shen, L.; Bai, C.; Wang, W.; Li, J.; Yu, X.; Li, Z.; Li, E.; Yuan, X.; Chi, Y.; 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. [Google Scholar] [CrossRef]
- Xu, J.; Shen, L.; Zhou, Z.; Li, J.; Bai, C.; Chi, Y.; Li, Z.; Xu, N.; Li, E.; Liu, T.; et al. Surufatinib in advanced extrapancreatic neuroendocrine tumours (SANET-ep): A randomised, double-blind, placebo-controlled, phase 3 study. Lancet Oncol. 2020, 21, 1500–1512. [Google Scholar] [CrossRef]
- Dasari, A.; Li, D.; Sung, M.W.; Tucci, C.; Kauh, J.S.; Kania, M.K.; Paulson, A.S. Efficacy and safety of surufatinib in United States (US) patients (pts) with neuroendocrine tumors (NETs). J. Clin. Oncol. 2020, 38, 4610. [Google Scholar] [CrossRef]
- Beyens, M.; Vandamme, T.; Peeters, M.; Van Camp, G.; Op de Beeck, K. Resistance to targeted treatment of gastroenteropancreatic neuroendocrine tumors. Endocr. Relat. Cancer 2019, 26, R109–R130. [Google Scholar] [CrossRef] [Green Version]
- Das, S.; Dasari, A. Novel therapeutics for patients with well-differentiated gastroenteropancreatic neuroendocrine tumors. Ther. Adv. Med. Oncol. 2021, 13, e17588359211018047. [Google Scholar] [CrossRef]
- Yi, M.; Jiao, D.; Qin, S.; Chu, Q.; Wu, K.; Li, A. Synergistic effect of immune checkpoint blockade and anti-angiogenesis in cancer treatment. Mol. Cancer 2019, 18, 60. [Google Scholar] [CrossRef]
- McNamara, M.G.; Scoazec, J.Y.; Walter, T. Extrapulmonary poorly differentiated NECs, including molecular and immune aspects. Endocr. Relat. Cancer 2020, 27, R219–R238. [Google Scholar] [CrossRef]
- Maggio, I.; Manuzzi, L.; Lamberti, G.; Ricci, A.D.; Tober, N.; Campana, D. Landscape and future perspectives of immunotherapy in neuroendocrine neoplasia. Cancers 2020, 12, 832. [Google Scholar] [CrossRef] [Green Version]
- Weber, M.M.; Fottner, C. Immune checkpoint inhibitors in the treatment of patients with neuroendocrine neoplasia. Oncol. Res. Treat. 2018, 41, 306–312. [Google Scholar] [CrossRef]
- Zhang, W.H.; Wang, W.Q.; Gao, H.L.; Yu, X.J.; Liu, L. The tumor immune microenvironment in gastroenteropancreatic neuroendocrine neoplasms. Biochim. Biophys. Acta Rev. Cancer 2019, 1872, 188311. [Google Scholar] [CrossRef]
- Yang, M.W.; Fu, X.L.; Jiang, Y.S.; Chen, X.J.; Tao, L.Y.; Yang, J.Y.; Huo, Y.M.; Liu, W.; Zhang, J.F.; Liu, P.F.; et al. Clinical significance of programmed death 1/programmed death ligand 1 pathway in gastric neuroendocrine carcinomas. World J. Gastroenterol. 2019, 25, 1684–1696. [Google Scholar] [CrossRef]
- Bösch, F.; Brüwer, K.; Altendorf-Hofmann, A.; Auernhammer, C.J.; Spitzweg, C.; Westphalen, C.B.; Boeck, S.; Schubert-Fritschle, G.; Werner, J.; Heinemann, V.; et al. Immune checkpoint markers in gastroenteropancreatic neuroendocrine neoplasia. Endocr. Relat. Cancer 2019, 26, 293–301. [Google Scholar] [CrossRef]
- Ferrata, M.; Schad, A.; Zimmer, S.; Musholt, T.J.; Bahr, K.; Kuenzel, J.; Becker, S.; Springer, E.; Roth, W.; Weber, M.M.; et al. PD-L1 expression and immune cell infiltration in gastroenteropancreatic (GEP) and non-GEP neuroendocrine neoplasms with high proliferative activity. Front. Oncol. 2019, 9, 343. [Google Scholar] [CrossRef] [Green Version]
- Strosberg, J.; Mizuno, N.; Doi, T.; Grande, E.; Delord, J.P.; Shapira-Frommer, R.; Bergsland, E.; Shah, M.; Fakih, M.; Takahashi, S.; et al. Efficacy and safety of pembrolizumab in previously treated advanced neuroendocrine tumors: Results from the phase II KEYNOTE-158 study. Clin. Cancer Res. 2020, 26, 2124–2130. [Google Scholar] [CrossRef]
- Patel, S.P.; Othus, M.; Chae, Y.K.; Giles, F.J.; Hansel, D.E.; Singh, P.P.; Fontaine, A.; Shah, M.H.; Kasi, A.; Baghdadi, T.A.; et al. A phase II basket trial of dual anti-CTLA-4 and anti-PD-1 blockade in rare tumors (DART SWOG 1609) in patients with nonpancreatic neuroendocrine tumors. Clin. Cancer Res. 2020, 26, 2290–2296. [Google Scholar] [CrossRef] [Green Version]
- Mandriani, B.; Pelle, E.; Pezzicoli, G.; Strosberg, J.; Abate-Daga, D.; Guarini, A.; Cives, M.; Porta, C. Adoptive T-cell immunotherapy in digestive tract malignancies: Current challenges and future perspectives. Cancer Treat. Rev. 2021, 100, 102288. [Google Scholar] [CrossRef]
- Park, J.H.; Rivière, I.; Gonen, M.; Wang, X.; Sénéchal, B.; Curran, K.J.; Sauter, C.; Wang, Y.; Santomasso, B.; Mead, E.; et al. Long-term follow-up of CD19 CAR therapy in acute lymphoblastic leukemia. N. Engl. J. Med. 2018, 378, 449–459. [Google Scholar] [CrossRef]
- Mandriani, B.; Cives, M.; Pelle, E.; Quaresmini, D.; Ramello, M.; Strosberg, J.; Abatte-Daga, D.; Silvestris, F. Development of anti-SSTR CAR-T cells for future treatment of NETs. J. Neuroendocr. Tumors Pancreat. Dis. Sci. 2020, 49, 479. [Google Scholar]
- Whalen, K.A.; White, B.H.; Quinn, J.M.; Kriksciukaite, K.; Alargova, R.; Au Yeung, T.P.; Bazinet, P.; Brockman, A.; DuPont, M.M.; Oller, H.; et al. Targeting the somatostatin receptor 2 with the miniaturized drug conjugate, PEN-221: A potent and novel therapeutic for the treatment of small cell lung cancer. Mol. Cancer Ther. 2019, 18, 1926–1936. [Google Scholar] [CrossRef] [Green Version]
- Wang, G.; Kang, X.; Chen, K.S.; Jehng, T.; Jones, L.; Chen, J.; Huang, X.F.; Chen, S.Y. An engineered oncolytic virus expressing PD-L1 inhibitors activates tumor neoantigen-specific T cell responses. Nat. Commun. 2020, 11, 1395. [Google Scholar] [CrossRef]
- Yu, D.; Leja-Jarblad, J.; Loskog, A.; Hellman, P.; Giandomenico, V.; Oberg, K.; Essand, M. Preclinical evaluation of AdVince, an oncolytic adenovirus adapted for treatment of liver metastases from neuroendocrine cancer. Neuroendocrinology 2017, 105, 54–66. [Google Scholar] [CrossRef]
- Kloker, L.D.; Berchtold, S.; Smirnow, I.; Beil, J.; Krieg, A.; Sipos, B.; Lauer, U.M. Oncolytic vaccinia virus GLV-1h68 exhibits profound antitumoral activities in cell lines originating from neuroendocrine neoplasms. BMC Cancer 2020, 20, 628. [Google Scholar] [CrossRef]
- Matsushima, H.; Kaibori, M.; Hatta, M.; Ishizaki, M.; Nakatake, R.; Okumura, T.; Yoshii, K.; Todo, T. Efficacy of a third-generation oncolytic herpes simplex virus in neuroendocrine tumor xenograft models. Oncotarget 2019, 10, 7132–7141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, J.Y.; Kunz, P.L. Making sense of a complex disease: A practical approach to managing neuroendocrine tumors. JCO Oncol. Pract. 2021, 18, 4. [Google Scholar] [CrossRef]
- Kouvaraki, M.A.; Ajani, J.A.; Hoff, P.; Wolff, R.; Evans, D.B.; Lozano, R.; Yao, J.C. Fluorouracil, doxorubicin, and streptozocin in the treatment of patients with locally advanced and metastatic pancreatic endocrine carcinomas. J. Clin. Oncol. 2004, 22, 4710–4719. [Google Scholar] [CrossRef] [PubMed]
- Arnold, R.; Rinke, A.; Schmidt, C.; Hofbauer, L. Chemotherapy. Best Pract. Res. Clin. Gastroenterol. 2005, 19, 649–656. [Google Scholar] [CrossRef] [PubMed]
- Thomas, K.; Voros, B.A.; Meadows-Taylor, M.; Smeltzer, M.P.; Griffin, R.; Boudreaux, J.P.; Thiagarajan, R.; Woltering, E.A.; Ramirez, R.A. Outcomes of capecitabine and temozolomide (CAPTEM) in advanced neuroendocrine neoplasms (NENs). Cancers 2020, 12, 206. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kunz, P.L.; Catalano, P.J.; Nimeiri, H.; Fisher, G.A.; Longacre, T.A.; Suarez, C.J.; Yao, J.C.; Kulke, M.H.; Hendifar, A.E.; Shanks, J.C.; et al. A randomized study of temozolomide or temozolomide and capecitabine in patients with advanced pancreatic neuroendocrine tumors: A trial of the ECOG-ACRIN Cancer Research Group (E2211). J. Clin. Oncol. 2018, 36, 4004. [Google Scholar] [CrossRef]
- Strosberg, J.R.; Fine, R.L.; Choi, J.; Nasir, A.; Coppola, D.; Chen, D.T.; Helm, J.; Kvols, L. First-line chemotherapy with capecitabine and temozolomide in patients with metastatic pancreatic endocrine carcinomas. Cancer 2011, 117, 268–275. [Google Scholar] [CrossRef]
- Shah, M.H.; Goldner, W.S.; Benson, A.B.; Bergsland, E.; Blaszkowsky, L.S.; Brock, P.; Chan, J.; Das, S.; Dickson, P.V.; Fanta, P.; et al. Neuroendocrine and adrenal tumors, version 2.2021, NCCN clinical practice guidelines in oncology. J. Natl. Compr. Cancer Netw. 2021, 19, 839–868. [Google Scholar] [CrossRef]
- Pavel, M.; O’Toole, D.; Costa, F.; Capdevila, J.; Gross, D.; Kianmanesh, R.; Krenning, E.; Knigge, U.; Salazar, R.; Pape, U.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. [Google Scholar] [CrossRef]
- Yoo, C.; Oh, C.R.; Kim, S.T.; Bae, W.K.; Choi, H.J.; Oh, D.Y.; Lee, M.A.; Ryoo, B.Y. Systemic treatment of advanced gastroenteropancreatic neuroendocrine tumors in Korea: Literature review and expert opinion. Cancer Res. Treat. 2021, 53, 291–300. [Google Scholar] [CrossRef]
- Cives, M.; Strosberg, J.R. Gastroenteropancreatic neuroendocrine tumors. CA Cancer J. Clin. 2018, 68, 471–487. [Google Scholar] [CrossRef]
- Chan, D.; Bergsland, E.K.; Chan, J.A.; Gadgil, R.; Halfdanarson, T.R.; Hornbacker, K.; Kelly, V.; Kunz, P.L.; McGarrah, P.W.; Raj, N.P.; et al. Temozolomide in grade III neuroendocrine neoplasms (G3 NENs): A multicenter retrospective review. J. Clin. Oncol. 2019, 37, 321. [Google Scholar] [CrossRef]
- Sorbye, H.; Welin, S.; Langer, S.W.; Vestermark, L.W.; Holt, N.; Osterlund, P.; Dueland, S.; Hofsli, E.; Guren, M.G.; Ohrling, K.; et al. Predictive and prognostic factors for treatment and survival in 305 patients with advanced gastrointestinal neuroendocrine carcinoma (WHO G3): The NORDIC NEC study. Ann. Oncol. 2013, 24, 152–160. [Google Scholar] [CrossRef]
- Sorbye, H.; Baudin, E.; Perren, A. The problem of high-grade gastroenteropancreatic neuroendocrine neoplasms: Well-differentiated neuroendocrine tumors, neuroendocrine carcinomas, and beyond. Endocrinol. Metab. Clin. N. Am. 2018, 47, 683–698. [Google Scholar] [CrossRef]
- Raj, N.; Valentino, E.; Capanu, M.; Tang, L.H.; Basturk, O.; Untch, B.R.; Allen, P.J.; Klimstra, D.S.; Reidy-Lagunes, D. Treatment response and outcomes of grade 3 pancreatic neuroendocrine neoplasms based on morphology: Well differentiated versus poorly differentiated. Pancreas 2017, 46, 296–301. [Google Scholar] [CrossRef]
- Garcia-Carbonero, R.; Sorbye, H.; Baudin, E.; Raymond, E.; Wiedenmann, B.; Niederle, B.; Sedlackova, E.; Toumpanakis, C.; Anlauf, M.; Cwikla, J.B.; et al. ENETS consensus guidelines for high-grade gastroenteropancreatic neuroendocrine tumors and neuroendocrine carcinomas. Neuroendocrinology 2016, 103, 186–294. [Google Scholar] [CrossRef] [Green Version]
- Walter, T.; Tougeron, D.; Baudin, E.; Le Malicot, K.; Lecomte, T.; Malka, D.; Hentic, O.; Manfredi, S.; Bonnet, I.; Guimbaud, R.; et al. Poorly differentiated gastro-entero-pancreatic neuroendocrine carcinomas: Are they really heterogeneous? Insights from the FFCD-GTE national cohort. Eur. J. Cancer 2017, 79, 158–165. [Google Scholar] [CrossRef]
- Brixi-Benmansour, H.; Jouve, J.L.; Mitry, E.; Bonnetain, F.; Landi, B.; Hentic, O.; Bedenne, L.; Cadiot, G. Phase II study of first-line FOLFIRI for progressive metastatic well-differentiated pancreatic endocrine carcinoma. Dig. Liver Dis. 2011, 43, 912–916. [Google Scholar] [CrossRef]
- Couronne, T.; Girot, P.; Hadoux, J.; Lecomte, T.; Durand, A.; Fine, C.; Vandevoorde, K.; Lombard-Bohas, C.; Walter, T. Post first-line dacarbazine or temozolomide in neuroendocrine carcinoma. Endocr. Connect. 2020, 9, 498–505. [Google Scholar] [CrossRef]
- Søreide, J.A.; Kvaløy, J.T.; Lea, D.; Sandvik, O.M.; Al-Saiddi, M.; Haslerud, T.M.; Garresori, H.; Karlsen, L.N.; Gudlaugsson, E.; Søreide, K. The overriding role of surgery and tumor grade for long-term survival in patients with gastroenteropancreatic neuroendocrine neoplasms: A population-based cohort study. Cancer Rep. 2022, 5, e1462. [Google Scholar] [CrossRef]
- Pommergaard, H.C.; Nielsen, K.; Sorbye, H.; Federspiel, B.; Tabaksblat, E.M.; Vestermark, L.W.; Janson, E.T.; Hansen, C.P.; Ladekarl, M.; Garresori, H.; et al. Surgery of the primary tumour in 201 patients with high-grade gastroenteropancreatic neuroendocrine and mixed neuroendocrine-non-neuroendocrine neoplasms. J. Neuroendocrinol. 2021, 33, e12967. [Google Scholar] [CrossRef]
- Merola, E.; Falconi, M.; Rinke, A.; Staettner, S.; Krendl, F.; Partelli, S.; Andreasi, V.; Gress, T.M.; Pascher, A.; Arsenic, R.; et al. Radical intended surgery for highly selected stage IV neuroendocrine neoplasms G3. Am. J. Surg. 2020, 220, 284–289. [Google Scholar] [CrossRef]
- Desai, G.S.; Pande, P.; Chhabra, V.; Shah, R.C.; Jagannath, P. Multimodality management, recurrence patterns, and long-term outcome of gastroenteropancreatic neuroendocrine neoplasms: Progress over 17 years. Indian J. Gastroenterol. 2019, 38, 399–410. [Google Scholar] [CrossRef]
- Sato, Y.; Hashimoto, S.; Mizuno, K.; Takeuchi, M.; Terai, S. Management of gastric and duodenal neuroendocrine tumors. World J. Gastroenterol. 2016, 22, 6817–6828. [Google Scholar] [CrossRef]
- Fave, G.D.; O’Toole, D.; Sundin, A.; Taal, B.; Ferolla, P.; Ramage, J.K.; Ferone, D.; Ito, T.; Weber, W.; Zheng-Pei, Z.; et al. ENETS consensus guidelines update for gastroduodenal neuroendocrine neoplasms. Neuroendocrinology 2016, 103, 119–124. [Google Scholar] [CrossRef] [Green Version]
- Tang, L.H. Pancreatic neuroendocrine neoplasms landscape and horizon. Arch. Pathol. Lab. Med. 2020, 144, 816–828. [Google Scholar] [CrossRef] [Green Version]
- Partelli, S.; Cirocchi, R.; Crippa, S.; Cardinali, L.; Fendrich, V.; Bartsch, D.K.; Falconi, M. Systematic review of active surveillance versus surgical management of asymptomatic small non-functioning pancreatic neuroendocrine neoplasms. Br. J. Surg. 2017, 104, 34–41. [Google Scholar] [CrossRef]
- Partelli, S.; Javed, A.A.; Andreasi, V.; He, J.; Muffatti, F.; Weiss, M.J.; Sessa, F.; La Rosa, S.; Doglioni, C.; Zamboni, G.; et al. The number of positive nodes accurately predicts recurrence after pancreaticoduodenectomy for nonfunctioning neuroendocrine neoplasms. Eur. J. Surg. Oncol. 2018, 44, 778–783. [Google Scholar] [CrossRef]
- Memeh, K.O.; Vaghaiwalla, T.; Keutgen, X.M. Surgical treatment of non-functioning pancreatic neuroendocrine tumors: Current controversies and challenges. J. Pancreatol. 2020, 3, 51–58. [Google Scholar] [CrossRef]
- Partelli, S.; Bartsch, D.K.; Capdevila, J.; Chen, J.; Knigge, U.; Niederle, B.; Nieveen van Dijkum, E.J.M.; Pape, U.F.; Pascher, A.; Ramage, J.; et al. ENETS consensus guidelines for standard of care in neuroendocrine tumours: Surgery for small intestinal and pancreatic neuroendocrine tumours. Neuroendocrinology 2017, 105, 255–265. [Google Scholar] [CrossRef] [PubMed]
- Rault-Petit, B.; Do Cao, C.; Guyétant, S.; Guimbaud, R.; Rohmer, V.; Julié, C.; Baudin, E.; Goichot, B.; Coriat, R.; Tabarin, A.; et al. Current management and predictive factors of lymph node metastasis of appendix neuroendocrine tumors: A national study from the French group of endocrine tumors (GTE). Ann. Surg. 2019, 270, 165–171. [Google Scholar] [CrossRef] [PubMed]
- Ito, T.; Masui, T.; Komoto, I.; Doi, R.; Osamura, R.Y.; Sakurai, A.; Ikeda, M.; Takano, K.; Igarashi, H.; Shimatsu, A.; et al. JNETS clinical practice guidelines for gastroenteropancreatic neuroendocrine neoplasms: Diagnosis, treatment, and follow-up: A synopsis. J. Gastroenterol. 2021, 56, 1033–1044. [Google Scholar] [CrossRef] [PubMed]
- Pape, U.F.; Niederle, B.; Costa, F.; Gross, D.; Kelestimur, F.; Kianmanesh, R.; Knigge, U.; Öberg, K.; Pavel, M.; Perren, A.; et al. ENETS consensus guidelines for neuroendocrine neoplasms of the appendix (excluding goblet cell carcinomas). Neuroendocrinology 2016, 103, 144–152. [Google Scholar] [CrossRef] [Green Version]
- Brighi, N.; La Rosa, S.; Rossi, G.; Grillo, F.; Pusceddu, S.; Rinzivillo, M.; Spada, F.; Tafuto, S.; Massironi, S.; Faggiano, A.; et al. Morphological factors related to nodal metastases in neuroendocrine tumors of the appendix: A multicentric retrospective study. Ann. Surg. 2020, 271, 527–533. [Google Scholar] [CrossRef]
- Albers, M.B.; Almquist, M.; Bergenfelz, A.; Nordenström, E. Complications of surgery for gastro-entero-pancreatic neuroendocrine neoplasias. Langenbecks Arch. Surg. 2020, 405, 137–143. [Google Scholar] [CrossRef] [Green Version]
- Caplin, M.; Sundin, A.; Nillson, O.; Baum, R.P.; Klose, K.J.; Kelestimur, F.; Plöckinger, U.; Papotti, M.; Salazar, R.; Pascher, A. ENETS consensus guidelines for the management of patients with digestive neuroendocrine neoplasms: Colorectal neuroendocrine neoplasms. Neuroendocrinology 2012, 95, 88–97. [Google Scholar] [CrossRef]
- Fields, A.C.; McCarty, J.C.; Lu, P.; Vierra, B.M.; Pak, L.M.; Irani, J.; Goldberg, J.E.; Bleday, R.; Chan, J.; Melnitchouk, N. Colon neuroendocrine tumors: A new lymph node staging classification. Ann. Surg. Oncol. 2019, 26, 2028–2036. [Google Scholar] [CrossRef]
- Chagpar, R.; Chiang, Y.J.; Xing, Y.; Cormier, J.N.; Feig, B.W.; Rashid, A.; Chang, G.J.; You, Y.N. Neuroendocrine tumors of the colon and rectum: Prognostic relevance and comparative performance of current staging systems. Ann. Surg. Oncol. 2013, 20, 1170–1178. [Google Scholar] [CrossRef]
- Ramage, J.K.; De Herder, W.W.; Delle Fave, G.; Ferolla, P.; Ferone, D.; Ito, T.; Ruszniewski, P.; Sundin, A.; Weber, W.; Zheng-Pei, Z.; et al. ENETS consensus guidelines update for colorectal neuroendocrine neoplasms. Neuroendocrinology 2016, 103, 139–143. [Google Scholar] [CrossRef]
- de Mestier, L.; Lorenzo, D.; Fine, C.; Cros, J.; Hentic, O.; Walter, T.; Panis, Y.; Couvelard, A.; Cadiot, G.; Ruszniewski, P. Endoscopic, transanal, laparoscopic, and transabdominal management of rectal neuroendocrine tumors. Best Pract. Res. Clin. Endocrinol. Metab. 2019, 33, 101293. [Google Scholar] [CrossRef]
- Cockburn, K.C.; Toumi, Z.; Mackie, A.; Julyan, P. Radioguided surgery for gastroenteropancreatic neuroendocrine tumours: A systematic literature review. J. Gastrointest. Surg. 2021, 25, 3244–3257. [Google Scholar] [CrossRef]
- Kaçmaz, E.; Engelsman, A.F.; Bemelman, W.A.; Tanis, P.J.; Nieveen van Dijkum, E.J.M. International survey on opinions and use of minimally invasive surgery in small bowel neuroendocrine neoplasms. Eur. J. Surg. Oncol. 2021. [Google Scholar] [CrossRef]
- Kaçmaz, E.; Klümpen, H.J.; Bemelman, W.A.; Nieveen van Dijkum, E.J.M.; Engelsman, A.F.; Tanis, P.J. Evaluating nationwide application of minimally invasive surgery for treatment of small bowel neuroendocrine neoplasms. World J. Surg. 2021, 45, 2463–2470. [Google Scholar] [CrossRef]
Terminology | Differentiation | Grade | Mitotic Rate (mitoses/2 mm2) | Ki-67 Index |
---|---|---|---|---|
NET, G1 | Well differentiated | Low | <2 | <3% |
NET, G2 | Intermediate | 2–20 | 3–20% | |
NET, G3 | High | >20 | >20% | |
NEC, small-cell type | Poorly differentiated | High | >20 | >20% |
NEC, large-cell | >20 | >20% | ||
MiNEN | Well or poorly | Variable | Variable | Variable |
Illness/Phenotype | Pattern of Inheritance | Causative Gene(s) | Products of Genes Expression |
---|---|---|---|
Lynch syndrome | AD | hMSH2, hMLH1 (hPMS1, hPMS2, hMSH6) | MSH2, MLH1, MSH6, PMS2, PMS1 |
Familial adenomatous polyposis 1 (FAP) | AD (AR) | APC (MUTYH) | APC (hMYH) |
Li-Fraumeni syndrome | AD | TP53 | TP53 |
NF1 | AD | NF1 | Neurofibromin |
TSC-1 | AD | TSC1 | Hamartin |
TSC-2 | AD | TSC2 | Tuberin |
VHL (Von Hippel–Lindau disease) | AD | VHL | pVHL |
MEN-1 | AD | MEN1 | MEN1 |
MEN-2B | AD | RET | RET |
MEN-4 | AD | CDKN1B | p27 |
Polycythemia paraganglioma syndrome | - | EPAS1 | HIF2A |
Mahvash disease (MVAH) | AR | GCGR | Glucagon receptor |
Heading | Sensitivity | Advantages | Disadvantages |
---|---|---|---|
US | PanNEN 13–27% 85–90% liver metastases (CEUS—99%) | Cheap, widely accessible Can be enhanced by using contrast | Not recommended for the other parts of the GI tract |
EUS | PanNET 54–97% Insulinomas 71–94% | Sensitivity of insulinoma detection higher than that achieved using CT (20–63%) Possibility of fine needle biopsy of the lesion | The quality of the test depends greatly on the skill of the person performing the test Possibility of mistaking high-grade NENs for adenocarcinoma |
CT contrast enhanced | PanNET 63–82% Metastases to the liver 82%, lymph nodes 60–70%, bones 58% | Good visualization of vascular infiltration | Low sensitivity for detecting lesions < 1 cm and small lesions in duodenum, stomach and small intestine and bone metastases |
MRI contrast enhanced | PanNEN 79% | Less radiation to the patient than that in CT | Low sensitivity for detecting small lesions in the duodenum, stomach and small intestine |
DWI | 83% liver metastases | Detection of lesions after treatment Easier differentiation of hepatic NEN metastases from hemangiomas Measures the effectiveness of PRRT | |
111In-pentetreotide SPECT/CT | PanNET 60–80% 90–100% liver metastases | Better sensitivity than standard methods | More radiation Less effective than 68Ga-DOTA |
68Ga-DOTA-SSA PET/CT | PanNETs 79.6% 95–100% liver metastases (DOTATAC) | Less radiation to the patient The most effective method listed | Low half-life time—68 min |
Locoregional/Resectable | Metastatic/Unresectable | |||||||||
---|---|---|---|---|---|---|---|---|---|---|
G1 G2 G3 | G1/G2 | G3 | ||||||||
Surgery | Asymptomatic/stable | Symptomatic/progressive | NET | NEC | ||||||
Observe SSA | Hepatic dominant/metastases | Widespread | Cisplatin/Eotpside Everolimus Sunitinib ** Platinum/Etopside CAP/TEM PRRT SSTR- rapid SSTR+ slow Studies | Ki-67 50–60% | Ki-67 >60% | |||||
Platinum/Etoposide CAP/TEM FOLFIRI FOLFOX Studies | Plapinum/Etopside FOLFIRI FOLFOX Studies | |||||||||
Chemoembolization STZ | panNET | Midgut NET | ||||||||
SSTR+ | SSTR+ Ki-67 >10% | SSTR- | SSTR+ | SSTR- | ||||||
Everolimus Studies Sunitinib Lanreotide SSA PRRT | STZ/FU CAP/TEM * | Everolimus Studies Sunitinib STZ/FU CAP/TEM | Everolimus IFN alpha Studies Octreotide Lanreotide SSA PRRT | Everolimus IFN alpha Studies |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Kaliszewski, K.; Ludwig, M.; Greniuk, M.; Mikuła, A.; Zagórski, K.; Rudnicki, J. Advances in the Diagnosis and Therapeutic Management of Gastroenteropancreatic Neuroendocrine Neoplasms (GEP-NENs). Cancers 2022, 14, 2028. https://doi.org/10.3390/cancers14082028
Kaliszewski K, Ludwig M, Greniuk M, Mikuła A, Zagórski K, Rudnicki J. Advances in the Diagnosis and Therapeutic Management of Gastroenteropancreatic Neuroendocrine Neoplasms (GEP-NENs). Cancers. 2022; 14(8):2028. https://doi.org/10.3390/cancers14082028
Chicago/Turabian StyleKaliszewski, Krzysztof, Maksymilian Ludwig, Maria Greniuk, Agnieszka Mikuła, Karol Zagórski, and Jerzy Rudnicki. 2022. "Advances in the Diagnosis and Therapeutic Management of Gastroenteropancreatic Neuroendocrine Neoplasms (GEP-NENs)" Cancers 14, no. 8: 2028. https://doi.org/10.3390/cancers14082028