Revision of Pancreatic Neuroendocrine Neoplasms (pNENs) from Mon, 2015-11-02 16:48

Ospedali Riuniti-Università Politecnica delle Marche, Ancona, Italy & Ospedale San Raffaele-Università Vita e Salute, Milan, Italy
m.falconi@univpm.it

Entry Version: 

Version 1.0, January 23, 2015

Citation: 

Falconi, Massimo. Maurizi, Angela. Partelli,Stefano. Crippa,Stefano. (2015). Pancreatic Neuroendocrine Neoplasms (pNENs).
Pancreapedia: Exocrine Pancreas Knowledge Base, DOI: 10.3998/panc.2015.4

Abstract

Pancreatic neuroendocrine neoplasms (pNENs) are relatively rare lesions that have been increasing in incidence over the past few decades largely because of the diagnosis of pancreatic incidentalomas on cross-sectional imaging. Most of these tumors are nonfunctioning presenting with symptoms secondary to mass effect, metastatic disease or as incidental findings. Diagnostic work-up aims to assess the stage and the grade of the disease as these parameters represent the main driver for treatment choice. The treatment of pNENs varies from conservative management to extensive surgical resection. Different medical treatments have been recently validated in large randomized clinical trials.    

1. Introduction

According to a population–based study, malignant pancreatic neuroendocrine neoplasms (pNENs) account for approximately 1% of pancreatic cancers by prevalence (55). An estimated 40% to 91% of pNENs are non-functioning. The remainder exhibit clinically evident hormonal symptoms (32). In the last decade, three different classifications have been proposed by the World Health Organization (WHO) (45). In the last classification, pNENs are divided according to tumor grading that is still the most powerful predictor of survival in these tumors. Recent years have seen a dramatic increase in interest and effort in conducting large randomized trials in order to explore different treatments. In this chapter, we focus on the current status of clinical and pathological aspects of pNENs.

2. Epidemiology

pNENs are relatively rare as their incidence is below 1 case per 100,000 inhabitants representing approximately 8-10% of all pancreatic neoplasms (19). The reported incidence of pNENs is about 0.32 per 100,000 inhabitants per year, which is lower than the incidence of lung, ileal and rectal NET (i.e. 1.35, 0.67, 0.86 per 100,000 respectively) (55). In the largest and most recent series of pNENs, including gastroenteropancreatic, thoracic and unknown primary NEN, the age adjusted incidence has risen from 1.1 to 5.2 per 100,000 inhabitants per year over the years 1973-2004 (20). Several factors may have contributed to this rise, including the improvement in classification of NENs, the widespread use of endoscopy for cancer screening and of other sensitive imaging procedures such as endoscopy ultrasound (EUS) and computed tomography CT (50). As a direct consequence, the incidence of pNETs <2 cm in the United States has increased by 710% over the last 22 years (20).

3. Classification and staging

The WHO 2010 classification (7, 45) distinguishes between well- or moderately-differentiated neuroendocrine tumors and poorly differentiated neuroendocrine carcinomas (NEC) of small or large cell type. This classification is mainly constructed on a grading scheme based on mitotic count or Ki67 index. pNENs are then classified into three categories: NET-G1 (with a mitotic count < 2 per 10 high-power fields (HPF) and/or < 2% Ki67 index), NET-G2 (with a mitotic count 2–20 per 10 HPF and/or 3–20% Ki67 index) and NEC-G3 (with a mitotic count > 20 per 10 HPF and/or > 20% Ki67 index) (Table 1).

The grading requires mitotic count in at least 50 HPFs (1 HPF = 2mm) and Ki67 index using the Dako MIB-1 antibody as a percentage of 500-2000 cells counted in areas of strongest nuclear labeling (“hot spots”) (Figure 1). If grade differs for mitotic count compared with Ki67 index, it is suggested that the higher grade must be assumed (45). 

The tumor-node-metastasis (TNM) system, proposed by the European Neuroendocrine Tumor Society (ENETS), is based on the evaluation of the following parameters: size, extra pancreatic invasion, lymph-node and liver metastasis. In this classification, pT1tumors are < 2 cm, pT2 are between 2 cm and 4 cm and pT3 are > 4cm (44). In 2009 an additional TNM staging classification for pNET was suggested in the seventh edition of the AJCC/UICC TNM classification (47). The AJCC/UICC and ENETS classifications differ in the definitions of the T stages although a cut-off of 2 cm to distinguish pT1 from pT2 is used in both. On the contrary, the AJCC/UICC system requires recognition of peripancreatic soft tissue invasion in order to distinguish pT2 from pT3 whereas the ENETS classification based this distinction on tumor size using a cut-off of 4 cm. (Table 2)


Figure 1. Non-functioning pNET: well-differentiated solid tumor cell nests with immunostaining for Ki67 (Ki-67: 6%)

A large retrospective multi-institutional study that included 1072 patients affected by pNENs demonstrated that the ENETS TNM staging system is superior to the AJCC/UICC 2010 TNM staging system (43).

4. Pathology

pNENs are usually solitary and solid masses with rounded borders. The most common pNEN is rich in small vessels and has little fibrotic stroma. Evident features of malignancy after macroscopic examination include involvement of perivisceral fat, invasion of duodenal wall or adjacent organs (2). Microscopically, the majority of pNENs are well-differentiated tumors, which grow as solid nests or arranged on trabecular patterns (Figure 2)

 

Figure 2. Example of the most common form of a pNEN, a well differentiated tumor.Architectural patterns included nesting and micro trabecular, with numerous small vessels. Nuclear features show the coarsely clumped, "salt and pepper" chromatin pattern

Glandular, acinar and cribriform features are also observed. A rich vascularization is typical. Immunohistochemistry is used to differentiate a pNEN from other neoplasms using antibodies to at least one general endocrine marker such as synaptophysin or chromogranin A (CgA). pNEN may express normally produced pancreatic hormones (i.e. insulin, glucagon, somatostatin, pancreatic polypeptide), hormones of ectopic origin (i.e. gastrin, vasoactive intestinal polypeptide, adrenocorticotrophic hormone) and bioamines (i.e. serotonin).

5. Diagnosis

Clinical findings

Clinical suspect of PNEN should be considered in one of the following conditions:

  1. presence of a clinical endocrine syndrome, which pushes the patient to seek medical care. In this case, diagnosis is considered “early” and allows the identification of the endocrine neoplasm when it is still small;
  2. presence of clinical symptoms related to the growth of the neoplasm or to the presence of pain related to retroperitoneal infiltration. Those cases usually involve a non-functioning pNEN (NF-pNEN) and do not lead to early symptoms, also they are diagnosed by symptoms caused by the increase in tumoral mass;
  3. incidental diagnosis of a pancreatic mass, which is the most common way these lesions are diagnosed.

If patients have symptoms, the most common presenting symptoms are abdominal pain (35-78%), weight loss (20-35%), anorexia and nausea (45%). Less frequent signs are intra-abdominal haemorrhage (4-20%), jaundice (17-50%), or a palpable mass (7-40%) (10, 11, 27, 29, 54). 

Laboratory

There are generally two types of laboratory analyses. First, measurement of specific hormones in the case of functioning-pNEN, which include glucagon, somatostatin, serotonin, gastrin, insulin, etc. in baseline conditions and after stimulation using respective clinical-laboratory tests (12, 48). Secondly, dosage of generic neuroendocrine markers such as chromogranin A (CgA) and neuron-specific enolase (NSE). The former is a glycoprotein localized inside secretory vesicles of endocrine cells as well as neuronal cells of the central and peripheral nervous system. It is the most important generic neuroendocrine marker with sensitivity between 60-90% (39). Nevertheless, plasma levels of CgA can be falsely positive in the presence of altered renal function, atrophic chronic gastritis, and during therapy with proton pump inhibitors. The latter (NSE) is found at the cytoplasmic level in endocrine cells and neurons (37).

Imaging

Radiological examinations such as abdominal computed tomography (CT) and ultrasound, especially with contrast medium, are able to localize the neoplasm and obtain complete preoperative assessment in about 60% of cases (46). Hyper vascularization of endocrine tumors, present in 60-70% of patients (30, 35), can be observed by both CT (Figure 3) and ultrasound with contrast medium, and can localize the lesion and provide other information regarding its nature. Recently, there has been a dramatic technical improvement in MRI assessment of pancreatic disease using diffusion-weighted imaging (DWI) (14). DWI has been used for years in brain MRI but it has only recently been extended to abdominal imaging. Recent reports have suggested that high b-value DWI may be helpful in the detection and characterization of pancreatic tumors (25, 28). 


Figure 3. CT scan demonstrating a hypervascularized lesion of the pancreatic tail associated with multiple liver metastase

It has been reported that a reduced apparent diffusion coefficient (ADC) is observed in most malignant lesions related to the histopathological features of the tumor (33). Endoscopic ultrasound (EUS) can be useful when it is difficult to localize the neoplasm as well as other techniques such as venous sampling during angiography. As EUS offers excellent visualization of the pancreas from the duodenum or stomach and can produce high-resolution images of the pancreas. It has been considered one of the most accurate methods for the detection of a pancreatic focal lesion, especially in patients with small tumors of 3 cm or less, but is operator dependant (41, 53). EUS also has the unique ability to obtain specimens for histopathological diagnosis using EUS-guided fine-needle tissue acquisition (EUS-FNTA).  Ki-67 determination can be assessed in the 87% of patients with a concordance of 83% with postoperative Ki-67 (22). 

Nuclear Medicine

The presence of prominent molecular biomarkers makes pNENs attractive for functional imaging with PET and SPECT. Over the past few years the gold standard for functional imaging of pNENs has been somatostatin receptor scintigraphy (SRS) with 111In-diethylenetriaminepentaacetic acid octreotide (6). More recently the introduction of 68Ga-DOTA-peptide PET/CT has significantly improved the diagnostic work-up in the evaluation of pNENs (3) (Figure 4). In comparison to scintigraphy, positron emission tomography (PET) has a 2- to 3-fold higher spatial resolution (3–6 vs. 10–15 mm) and facilities quantification of tracer uptake.


Figure 4. 68Gallium PET/CT of a pNET with liver metastases

Several different DOTA-peptides (DOTATOC, DOTANOC, and DOTATATE) have been used in the clinical setting for both pNENs diagnosis and peptide receptor radionuclide therapy (PRRT). The major difference among these compounds relies on a slightly different affinity to somatostatin receptor subtypes (3). The sensitivity and specificity of 68Ga PET/CT in detecting NET is 93 and 91%, respectively (49). Moreover, this technique provides relevant information for pNET patients’ clinical management (3). 18F-FDG PET/CT is of value for tumors with a high proliferation index (pNEC), whereas the diagnostic sensitivity seems to be low for pNETs with a low proliferation index, slow growth rate, and low glucose consumption (6). Usually, 18F-FDG PET/CT had a sensitivity of 92% for tumors with a proliferation index of above 15% (6). Dual tracer

PET/CT can be performed on a single day owing to the short half-life of 68Ga and can be superior to histopathology by demonstrating tumor heterogeneity especially for patients with multiple metastases (18).

6. Treatment

Surgery

Incidental diagnosis of pancreatic NENs is associated with a significantly better survival after curative resection, compared to patients with symptoms (9). Moreover, Bettini et al. (4) demonstrated that patients with an incidental diagnosis associated with a tumor size < 2 cm, had a 5-year overall survival of 100% with a minimal risk of recurrence. On the basis of these experiences, the European Neuroendocrine Tumor Society (ENETS) guidelines now recommend a wait and see policy in selected patients with asymptomatic sporadic pancreatic NENs, when the possibility of surgical cure has to be weighed against the operative morbidity, mortality and long-term complications associated with pancreatic surgery (15). Preliminary reports have demonstrated the safety of this conservative approach (9, 17, 24). On the other hand, surgery still represents the treatment of choice for pancreatic NENs > 2 cm and/or for symptomatic forms.  Pancreatic resections differ according to tumor site: lesions of the pancreatic head are treated with a pancreaticoduodenectomy while lesions of the body and tail with a distal pancreatectomy. Role of lymphadenectomy for patients with a pancreatic NEN is still unclear (40). Lymph node metastases occur only in the 30% of patients affected by these tumors (40) but the association between node metastases and poorer survival is still debated (5). Laparoscopic procedures play an important role in the treatment of pancreatic endocrine tumors. It was demonstrated that laparoscopic distal pancreatectomy and enucleation are safe and feasible in patients with pancreatic endocrine tumors (16). Whenever a resection leaves no residual disease, an aggressive approach, including liver resection, is recommended. The conditions that have to be assessed preoperatively are (23) the absence of extra-abdominal disease, (30) the presence of a low proliferative index (Ki67), and (34) the existence of somatostatin receptors in order to deliver radiolabelled therapies, as they have been effective after cytoreductive surgery (15). The type of hepatic resection depends on the number of liver metastases, site and hepatic reserve itself. It can range from simple enucleation to segmental resection or to hepatectomy. In those patients with bilobar metastases or more than 75% of liver involvement, radical surgery can be rarely performed. In this light, medical, ablative and embolizing techniques can be provided in order to allow radical resection. There are valid palliative options in patients with pancreatic NETs with liver metastases which are not candidates for surgical resection. These mainly include trans arterial embolization (TAE), trans arterial chemo-embolization (TACE) and radiofrequency ablation (RFA). Such procedures can be used as loco-regional ablative therapy per se or as an adjunct to palliative surgery. Liver transplantation may be an option in a patient without extrahepatic metastases, and low proliferation rate when all other therapeutic options have failed (15).

Anti-tumoral treatments

A number of potential therapeutic targets have been identified and investigated for treating advanced pNENs. Recently, three large randomized clinical trials have been published (Table 3).

The mTOR pathway

The mammalian target of rapamycin (mTOR) is a serine-threonine kinase that stimulated cell growth, proliferation, and angiogenesis. Autocrine activation of the mTOR signaling pathway mediated through insulin-like growth factor 1, has been implicated in the proliferation of pancreatic neuroendocrine tumor cells. Consistently with this observation is the finding that inhibition of mTOR has a significant antiproliferative effect on PNET (56).

RADIANT-3 compared the efficacy of daily everolimus 10 mg versus placebo in 410 patients with advanced low- or intermediate-grade PNET (56). Everolimus significantly increased median PFS (progression-free survival) by 6.4 months compared with placebo (11 vs. 4.6 months, respectively; HR 0.35; 95% CI 0.27-0.45, p<0.001). 

VEGF pathway

Vascular endothelial growth factor (VEGF) is a key driver of angiogenesis in PNET. Tissue from pancreatic neuroendocrine tumors shows widespread expression of platelet-derived growth factor receptors (PDGFRs) α and β, stem-cell factor receptor (c-kit), and VEGF receptor (VEGFR)-2 and VEGFR-3 (42). Sunitinib malate inhibits these kinases. In a phase III, double-blind, placebo-controlled trial, sunitinib was compared with placebo in patients with advanced, well-differentiated PNET (42). Patients in the sunitinib group (37.5 mg/d) had a median PFS of 11.4 months compared with 5.5 months for the placebo group (HR 0.42; 95% CI 0.26-0.66; np<0.001).

Somatostatin Receptor Pathway

Somatostatin is a hormone that targets transmembrane G-protein-coupled somatostatin receptors. There are 5 known types of somatostatin receptors (SST1, SST2, SST3, SST4, and SST5). The use of somatostatin analogues (SSAs) in the treatment of patients with neuroendocrine neoplasms is a well-established practice (38). SSAs include octreotide/octreotide LAR, Lanreotide, and Pasireotide. Lanreotide was compared with placebo in a recent randomized, double-blind, multinational study (8). This study included neuroendocrine tumors originating in the pancreas, midgut, or hindgut or of unknown origin. Lanreotide, as compared with placebo, was associated with significantly prolonged PFS (HR 0.47; 95% CI 0.30-0.73, P<0.001). Nevertheless, therapeutic effect in the subgroup of PNET was not demonstrated (HR 0.58; 95% CI 0.32-1.04) (8). Peptide receptor radionuclide therapy (PRRT) is a new modality that uses radiolabelled peptides for treating unresectable or metastasized NENs. The rationale for such therapy is to convey radioactivity inside the tumor cells, where the sensitive targets, such as DNA, can be hit as a result of internalization of the somatostatin receptor and radiolabelled analogue complex (21). However, the exact role of this treatment in the management of NET remains to be defined. One trial comparing PRRT with high-dose SSA therapy has been launched (1).

Chemotherapy

Chemotherapy is recommended in PNET-G2 when other treatments failed and in PNEC-G3 (15). Most important regimens include the combination of streptozotocin (STZ) + doxorubicin or 5-fluorouracil (5-FU), cisplatin + etoposide and dacarbazine (13). The main indication for the use of cisplatin+etoposide is treatment of poorly differentiated neuroendocrine tumors. The combination has been reported to produce objective response in about 50% of anaplastic or poorly differentiated neuroendocrine tumors (31).

7. Functioning PNET

Insulinoma

Insulinomas are the most common functional pNETs with an incidence of 4 cases in one million people per year. Insulinoma should be suspected in the presence of Whipple's triad (symptoms or signs consistent with hypoglycemia; glucose level <50 mg/dL; relief of symptoms after administration of glucose). Insulinomas usually appear as small neoplasms, and are smaller than 1 cm in about 85% of cases (51); in 10-20% of cases they cannot be localized. In the majority of cases, however, ultrasound with contrast medium, abdominal CT and EUS are sufficient to localize the neoplasm. Surgery is the treatment of choice for localized lesions. A laparotomy is preferable because it allows meticulous manual exploration of the pancreas (36).

Gastrinoma

Gastrinomas are the second most frequent functioning endocrine tumor of the pancreatic area. Gastrinomas are malignant in 60-90% of cases and have a very aggressive behaviour in about 25% of cases. Similar to other functioning endocrine neoplasms, a gastrinoma is suspected when patients exhibit clinical symptoms, known as Zollinger-Ellison syndrome.   The diagnosis is confirmed based on laboratory tests of acid secretion and gastrin levels before and after secretin stimulation. Gastrinomas are generally malignant tumors and are often associated with unrecognized nodal metastases (52). A pancreaticoduodenectomy is the treatment of choice (26).

8. Conclusion

PNENs are rare neoplasms of the pancreas with a disease course considerably different from pancreatic ductal adenocarcinoma. Aggressive and extensive surgery may be an option for some patients suffering from locally advanced and even metastatic disease. In select cases this may involve resection of multiple organs to achieve a significant reduction of the tumor mass. The multidisciplinary approach is always mandatory in the management of these lesions. Since most pNENs exhibit indolent behavior, both the preservation of quality of life of the patient and the personalization of the therapy according to tumor’s and patient’s features are needed.

9. References

  1. A Study Comparing Treatment With 177Lu-DOTA0-Tyr3-Octreotate to Octreotide LAR in Patients With Inoperable, Progressive, Somatostatin Receptor Positive Midgut Carcinoid Tumours (NETTER-1), available on-line at: www.clinicaltrials.gov .
  2. Aldo Scarpa, Vincenzo Corbo, PATHOLOGY AND GENETICS, in: “Uncommon pancreatic neoplasms”, Paolo Pederzoli and Claudio Bassi editors, ISBN: 9788847026728, Springer-Verlag, 2013.
  3. Ambrosini V, Campana D, Bodei L et al. 68Ga-DOTANOC PET/CT clinical impact in patients with neuroendocrine tumors. J Nucl Med. 51: 669-673, 2010. PMID: 20395323
  4. Bettini R, Partelli S, Boninsegna L et al. Tumor size correlates with malignancy in nonfunctioning pancreatic endocrine tumor. Surgery 150: 75-82, 2011. PMID: 21683859
  5. Bilimoria KY, Talamonti MS, Tomlinson JS et al. Prognostic score predicting survival after resection of pancreatic neuroendocrine tumors: analysis of 3851 patients. Ann Surg 247: 490-500, 2008. PMID: 18376195
  6. Binderup T, Knigge U, Loft A et al. Functional imaging of neuroendocrine tumors: a head-to-head comparison of somatostatin receptor scintigraphy, 123I-MIBG scintigraphy, and 18F-FDG PET. J Nucl Med. 51: 704-712, 2010. PMID: 20395333
  7. Bosman FT: WHO Classification of Tumor of the Digestive System. Lyon, IARC Press, 2010.
  8. Caplin ME, Pavel M, Cwikla JB, Phan AT, Raderer M, Sedlackova E, et al. Lanreotide in metastatic enteropancreatic neuroendocrine tumors. New Engl J Med. 371(3):224-33, 2014. PMID 25014687
  9. Cheema A, Weber J, Strosberg JR. Incidental detection of pancreatic neuroendocrine tumors: an analysis of incidence and outcomes. Ann Surg Oncol 19: 2932-2936, 2012. PMID: 22350605
  10. Cheslyn-Curtis S, Sitaram V, Williamson RC. Management of non-functioning neuroendocrine tumours of the pancreas. Br J Surg 80: 625–627, 1993. PMID: 7686077
  11. Chu QD, Hill HC, Douglass HO Jr, et al. Predictive factors associated with long-term survival in patients with neuroendocrine tumors of the pancreas. Ann Surg Oncol 9: 855–862, 2002. PMID: 12417506
  12. 12.  Doppman JL, Miller DL, Chang R, et al. Intraarterial calcium stimulation test for detection of insulinomas. World J Surg 17(4): 439-443, 1993. PMID: 8362527
  13. Eriksson B1, Annibale B, Bajetta E, Mitry E, Pavel M, Platania M, Salazar R, Plöckinger U; Mallorca Consensus Conference participants; European Neuroendocrine Tumor Society. ENETS Consensus Guidelines for the Standards of Care in Neuroendocrine Tumors: chemotherapy in patients with neuroendocrine tumors. Neuroendocrinology 90(2):214-9, 2009. PMID: 19713713
  14. Erturk SM, Ichikawa T, Motosugi U, Sou H, Araki T. Diffusion-weighted MR imaging in the evaluation of pancreatic exocrine function before and after secretin stimulation. Am J Gastroenterol 101(1):133-6, 2006. PMID: 16405545
  15. Falconi M, Bartsch D, Eriksson B et al. ENETS Consensus Guidelines for the Management of Patients with Digestive Neuroendocrine Neoplasms of the Digestive System: Well-Differentiated Pancreatic Non-Functioning Tumors. Neuroendocrinology 95:120-134, 2012. PMID: 22261872
  16. Fernandez-Cruz L, Blanco L, Cosa R, Rendon H. Is laparoscopic resection adequate in patients with neuroendocrine pancreatic tumors? World J Surg 32: 904–917, 2008. PMID: 18264824
  17. Gaujoux S, Partelli S, Maire F et al. Observational study of natural history of small sporadic nonfunctioning pancreatic neuroendocrine tumors. J Clin Endocrinol Metab 98: 4784-4789, 2013. PMID: 24057286
  18. Kayani I, Bomanji JB, Groves A et al. Functional imaging of neuroendocrine tumors with combined PET/CT using 68Ga-DOTATATE (DOTA-DPhe1,Tyr3-octreotate) and 18F-FDG. Cancer 112: 2447-2455, 2008. PMID: 18383518
  19. Kloppel G, Perren A, Heitz PU. The gastroenteropancreatic neuroendocrine cell system and its tumors: the WHO classification. Ann N Y Acd Sci. 1014: 13-27, 2004. PMID: 15153416
  20. Kuo JH, Lee JA, Chabot JA. Nonfunctional pancreatic neuroendocrine tumors. Surg Clin North Am. 94(3):689-708, 2014. PMID: 24857584
  21. Kwekkeboom DJ, de Herder WW, Kam BL, van Eijck CH, van Essen M, Kooij PP, Feelders RA, van Aken MO, Krenning EP. Treatment with the radiolabeled somatostatin analog [177 Lu-DOTA 0,Tyr3]octreotate: toxicity, efficacy, and survival., J Clin Oncol. 26(13):2124-30, 2008. PMID: 18445841
  22. Larghi A, Capurso G, Carnuccio A, Ricci R, Alfieri S, Galasso D, Lugli F, Bianchi A, Panzuto F, De Marinis L, Falconi M, Delle Fave G, Doglietto GB, Costamagna G, Rindi G. Ki-67 grading of nonfunctioning pancreatic neuroendocrine tumors on histologic samples obtained by EUS-guided fine-needle tissue acquisition: a prospective study. Gastrointest Endosc. 76(3):570-7, 2012. PMID: 22898415
  23. Lawrence B, Gustafsson BI, Chan A et al. The epidemiology of gastroenteropancreatic neuroendocrine tumors. Endocrinol Metab Clin North Am 40: 1-18, vii, 2011. PMID: 21349409
  24. Lee LC, Grant CS, Salomao DR et al. Small, nonfunctioning, asymptomatic pancreatic neuroendocrine tumors (PNETs): role for nonoperative management. Surgery 152: 965-974, 2012. PMID: 23102679
  25. Lee SS, Byun JH, Park BJ, Park SH, Kim N, Park B, Kim JK, Lee MG. Quantitative analysis of diffusion-weighted magnetic resonance imaging of the pancreas: usefulness in characterizing solid pancreatic masses. J Magn Reson Imaging 28:928-36, 2008. PMID: 18821618
  26. Lopez CL, Falconi M, Waldmann J, Boninsegna L, Fendrich V, Goretzki PK, Langer P, Kahn PH, Partelli S, Bartsch DK. Partial pancreaticoduodenectomy can provide cure for duodenal gastrinoma associated with multiple endocrine neoplasia type 1. Ann Surg. 257:308-14, 2013.  PMID: 22580937
  27. Madura JA, Cummings OW, Wiebke EA, et al. Nonfunctioning islet cell tumors of the pancreas: a difficult diagnosis but one worth the effort. Am Surg 63: 573–577; discussion 577–578, 1997. PMID: 9202529
  28. Matsuki M, Inada Y, Nakai G, Tatsugami F, Tanikake M, Narabayashi I, Masuda D, Arisaka Y, Takaori K, Tanigawa N. Diffusion-weighed MR imaging of pancreatic carcinoma. Abdom Imaging 32:481-3, 2007. PMID: 17431713
  29. Matthews BD, Heniford BT, Reardon PR, et al. Surgical experience with nonfunctioning neuroendocrine tumors of the pancreas. Am Surg 66: 1116–1122;discussion 1122– 1113, 2000. PMID: 11149582
  30. Modlin IM, Tang LH. Approaches to the diagnosis of the gut neuroendocrine tumours: the last word. Gastroenterology 112: 583-590, 1997. PMID: 9024313
  31. Moertel CG1, Lefkopoulo M, Lipsitz S, Hahn RG, Klaassen D. Streptozocin-doxorubicin, streptozocin-fluorouracil or chlorozotocin in the treatment of advanced islet-cell carcinoma. N Engl J Med. 326(8):519-23, 1992. PMID: 1310159
  32. Moore FD, Scoinski MA, Joste NE. Endocrine tumors and malignancies. In : Skarin A, ed. Atlas of Diagnostic Oncology (ed 3rd). Philadelphia: Elsevier science limited; 2003
  33. Muraoka N, Uematsu H, Kimura H, Imamura Y, Fujiwara Y, Murakami M, Yamaguchi A, Itoh H. Apparent diffusion coefficient in pancreatic cancer: characterization and histopathological correlations. J Magn Reson Imaging 27(6):1302-8, 2008. PMID: 18504750
  34. Niederle MB, Hackl M, Kaserer K, Niederle B. Gastroenteropancreatic neuroendocrine tumours: the current incidence and staging based on the WHO and European Neuroendocrine Tumour Society classification: an analysis based on prospectively collected parameters. Endocr Relat Cancer 17: 909-918, 2010. PMID: 20702725
  35. Nojima T, Kojima T, Kato H, et al. Cystic endocrine tumour of the pancreas. Int J Pancreatol 10(1): 65-72, 1991. PMID: 1757731
  36. Norton JA. Intraoperativemethods to stage and localize pancreatic and duodenal tumors. An Oncol 10(suppl 4): 182-184, 1999. PMID: 10436817
  37. Oberg K. Diagnostic pathways. In: Kaplin M, Kvols L, Handbook of neuroendocrine tumours, their current and future management. Bioscientifica p103, 2006.
  38. Oberg KE, Reubi JC, Kwekkeboom DJ, Krenning EP. Role of somatostatins in gastroenteropancreatic neuroendocrine tumor development and therapy Gastroenterology. 139(3):742-53, 753, 2010. PMID: 20637207
  39. Panzuto F, Severi C, Cannizzaro R, et al. Utility of combined use of plasma levels of chromogranin A and pancreatic polypeptide in the diagnosis of gastrointestinal and pancreatic endocrine tumors. J Endocrinol  Invest 27: 6–11, 2004. PMID: 15053236
  40. Partelli S, Gaujoux S, Boninsegna L et al. Pattern and Clinical Predictors of Lymph Node Involvement in Nonfunctioning Pancreatic Neuroendocrine Tumors (NF-PanNETs). JAMA Surg, 2013. PMID: 23986355
  41. Rösch T, Lorenz R, Braig C, Feuerbach S, Siewert JR, Schusdziarra V, Classen M. Endoscopic ultrasound in pancreatic tumor diagnosis. Gastrointest Endosc. 37:347–352, 1991. PMID: 2070987
  42. Raymond E, Faivre S, Hammel P, Ruszniewski P. Sunitinib paves the way for targeted therapies in neuroendocrine tumors. Target Oncol. 4(4):253-4, 2009. PMID: 19911111
  43. Rindi G, Falconi M, Klersy C, et al. TNM staging of neoplasms of the endocrine pancreas: results from a large international cohort study. J Natl Cancer Inst. 104:764-77, 2012. PMID: 22525418
  44. Rindi G, Klöppel G, Alhman H, et al. TNM staging of foregut (neuro)endocrine tumors a consensus proposal including a grading system. Virchows Arch. 449:395-401, 2006. PMID: 16967267
  45. Rindi, G., R. Arnold, F. T. Bosman, C. Capella, D. S., Klimstra, G. Klo¨ppel, et al. Nomenclature and classification of neuroendocrine neoplasms of the digestive system, p. 13–14, in: F. T. Bosman, F. Carneiro, R. H. Hruban and N. Theise, eds. WHO, Classification of tumors of the digestive system. 4th ed. International Agency for Research on Cancer, 2010, Lyon.
  46. Rothmund M. Localization of endocrine pancreatic tumours. Br J Surg, 81(2): 164-166, 1994. PMID: 8156325
  47. Sobin LH, Gospodarowicz MK, Wittekind C (2009) UICC: TNM classification of malignant tumours, 7th edn. Wiley-Blackwell Oxford
  48. Thom AK, Norton JA, Doppman JL, et al. Prospective study of the use of intraarterial secretin injection and portal venous sampling tool localized duodenal gastrinomas, Surgery 112(6); 1002-1009, 1992. PMID: 1455303
  49. Treglia G, Castaldi P, Rindi G et al. Diagnostic performance of Gallium-68 somatostatin receptor PET and PET/CT in patients with thoracic and gastroenteropancreatic neuroendocrine tumours: a meta-analysis. Endocrine 42: 80-87, 2012. PMID: 22350660
  50. Vagefi PA, Razo O, Deshpande V, McGrath DJ, Lauwers GY, Thayer SP, Warshaw AL, Fernández-Del Castillo C. Evolving patterns in the detection and outcomes of pancreatic neuroendocrine neoplasms: the Massachusetts General Hospital experience from 1977 to 2005. Arch Surg. 142(4):347-54, 2007. PMID: 17438169
  51. Van Heerden J. The surgical treatment of insulinomas. In: Beger HG, Warshaw AL et al., The pancreas, 3rd edition, Oxford: Blackwell sciences. 1998. Vol. 77 pp 717-732.
  52. Volker F, Waldmann J, Bartsch DK. Langer P. Surgical management of pancreatic endocrine tumors Fendrich, V. et al. Nat. Rev. Clin. Oncol. 6, 419–428, 2009
  53. Volmar KE, Vollmer RT, Jowell PS, Nelson RC, Xie HB. Pancreatic FNA in 1000 cases: a comparison of imaging modalities. Gastrointest Endosc. 61:854–861, 2005. PMID: 15933687
  54. White TJ, Edney JA, Thompson JS, et al. Is there a prognostic difference between functional and nonfunctional islet cell tumors? Am J Surg 168: 627–629; discussion 629–630, 1994. PMID: 7978008
  55. Yao JC, Eisner MP, Leary C, et al. Population-based study of islet cell carcinoma. Ann Surg Oncol 14: 3492-3500, 2007. PMID: 17896148
  56. Yao JC, Shah MH, Ito T, Bohas CL, Wolin EM, Van Cutsem E, Hobday TJ, Okusaka T, Capdevila J, de Vries EG, Tomassetti P, Pavel ME, Hoosen S, Haas T, Lincy J, Lebwohl D, Öberg K. RAD001 in Advanced Neuroendocrine Tumors, Third Trial (RADIANT-3) Study Group. Everolimus for advanced pancreatic neuroendocrine tumors. N Engl J Med. 364(6):514-23, 2011. PMID: 21306238