Insulinoma

Earn CME/CE in your profession:


Continuing Education Activity

Insulinoma is a type of functional neuroendocrine tumor characterized by hypersecretion of insulin, causing hypoglycemia. It typically presents as a solitary benign tumor but can be associated with multiple endocrine neoplasia type 1 (MEN1). Patients with insulinoma have hypoglycemic episodes, more characteristically as fasting hypoglycemia. Insulinoma is usually diagnosed by biochemical testing. Surgical resection is the preferred treatment choice. This activity reviews the manifestations, evaluation, and treatment of insulinoma and highlights the importance of an interprofessional team in managing patients with this condition.

Objectives:

  • Describe the typical presenting symptoms of insulinoma.
  • Review the evaluation of insulinoma, including appropriate biochemical tests and localization modalities.
  • Summarize the management of insulinoma.
  • Outline the importance of collaboration of an interprofessional team in managing insulinoma to improve the patient quality of care.

Introduction

Insulinoma is a type of functional neuroendocrine tumor (NET) that manifests with hypoglycemia caused by inappropriately high insulin secretion. It more commonly presents as a solitary benign tumor, but it can sometimes be associated with multiple endocrine neoplasia type 1 (MEN1). Patients with insulinoma have hypoglycemic episodes, more characteristically as fasting hypoglycemia. Insulinoma is usually diagnosed by biochemical testing when there is high clinical suspicion. Surgical resection is the preferred treatment choice. Localization with imaging studies is fundamental to characterize the tumor better before surgical resection. Other treatment options are also available depending on the stage and grade of the tumor.

Etiology

The etiology of solitary insulinoma is not clearly understood. Normally high plasma glucose concentration would trigger the secretion of insulin from the pancreatic beta cells and vice versa. In patients with insulinoma, insulin still gets secreted, even when plasma glucose concentration is low. Hyperinsulinemia could result from an upregulated translation process due to a variant of insulin splice.[1] When the intracellular insulin store has been filled up, insulin gets secreted into the blood. Hyperinsulinemia causes increased glycogen synthesis, decreased gluconeogenesis, and glycogenolysis, all of which attribute to hypoglycemia.

MEN1 is a disorder characterized by a predisposition to the pituitary, parathyroid, and enteropancreatic tumors. When insulinomas are associated with MEN1, a mutation in the MEN1 gene at chromosome 11q13 is typically present in most cases.[2]

Epidemiology

The incidence of insulinoma is around 1 to 4 per million per year.[3][4] More commonly, insulinoma is a single benign tumor; in 5.8% of cases, it can be malignant.[3] It can coexist with other benign tumors in 7% of the cases, and in 6% to 7.6% of the cases, it is associated with MEN1 syndrome.[3][5] Surgical diagnosis happens at a median age of 47 to 50 years old.[3][5]

History and Physical

Fasting hypoglycemia is the most characteristic finding of insulinoma, reported in 73% of the population.[5] Around 20% of patients have both fasting and postprandial hypoglycemic symptoms.[5] There have been increasing numbers of patients reporting only postprandial hypoglycemic symptoms, which is more prominent in men.[5] Weight gain occurs in most of these patients.

Hypoglycemia presents in different ways. Sympathoadrenal activation symptoms can be seen, including palpitations, tremulousness, and diaphoresis. Severe hypoglycemia can cause neuroglycopenic symptoms, including blurry vision, confusion, seizures, or a behavioral change. Amnesia of the hypoglycemic event is common.[6]

Insulinoma should be suspected when a patient presents with the Whipple’s triad, which includes: symptoms of hypoglycemia, a documented low plasma glucose concentration, and subsequent improvement of the symptoms with the use of glucose treatment. 

In rare cases reported in the literature, the insulinoma may present with psychiatric symptoms, especially panic attacks.[7]

Evaluation

There are two crucial steps in evaluating an insulinoma. The first is to establish the diagnosis with biochemical tests when there is high clinical suspicion, and the second is to localize the tumor.

A 72-hour fast is the gold-standard test for diagnosing insulinoma.[4][8] It is useful when the Whipple triad is not observable and when there were no biochemical tests during spontaneous hypoglycemic episodes. [9] The combination of plasma glucose concentration under 55 mg/dL, insulin level greater than or equal to 3 microUnits/mL, C peptide level greater than or equal to 0.6 ng/mL, proinsulin level greater than or equal to 5 pmol/L, beta-hydroxybutyrate level less than or equal to 2.7 mmol/L and a simultaneous negative sulfonylurea level indicates that the hypoglycemia induction is by hyperinsulinemia.

A successful preoperative localization by non-invasive modalities remains around 75%.[5] Computed tomography (CT) with contrast is the most frequently used initial non-invasive testing.[8] CT detects 70 to 80% of the tumors where the insulinoma usually presents as a small solid mass, enhancing after contrast.[10][11] Abdominal ultrasound (US) focusing on the pancreas is sometimes preferred, given its convenience and low cost. Magnetic resonance image (MRI) detects about 85% of the insulinomas that seem to enhance homogeneously after gadolinium administration.[10][11] Glucagon-like peptide-1 receptor (GLP-1) imaging is a promising new modality test and has been studied to localize insulinoma with a successful detection rate of 66 to 97% in some small studies.[12][13][14][15] In animal models, PET and SPECT scans using radiolabeled excedin-4 helped localize the insulinoma in mice.[16]

When there is a high suspicion of insulinoma with negative non-invasive studies, invasive tests such as endoscopic ultrasonography (EUS) or a selective arterial calcium stimulation test (SACST) with hepatic venous sampling could be considered. EUS has a sensitivity of 70 to 95% and is superior to CT for localization. [10][17][18] This is the preferred test of choice if non-invasive studies are negative. It also allows tissue acquisition which can be very helpful, especially in small tumors.[17] SACST has a sensitivity of 93% for detecting insulinoma and could be performed when other imaging studies are negative or in patients with MEN1 syndrome.[5][19]

Histology examination and immunostaining for chromogranin A, synaptophysin, and insulin should be done to support the diagnosis of insulinoma.[4] A Ki67 index should also be obtained for grading, especially when a patient demonstrates an aggressive clinical course.[4]

Tumor size of greater than or equal to 2 cm, Ki67 more than 2%, and molecular features including chromosomal alteration such as 3p and 6q loss and 12 q gain or chromosomal instability, are possible predictors for metastatic disease and poor progression-free survival (PFS).[20]

Malignant insulinomas have evidence of extrapancreatic regional lymph nodes or even remote extension. Certain factors include fasting time for hypoglycemia being under 8 hours, higher blood insulin level (greater than or equal to 28 microUnits/mL), and higher C-peptide level (greater than or equal to 4.0 ng/mL) at the glycemic nadir. These factors, as well as larger tumor size (greater than or equal to 2.5 cm), all correlate with an increased risk of malignant disease.[21] The extent of liver metastases and the presence of lymph node metastases are important prognostic factors.[19][22][23]

The recommendation is that all young patients with insulinoma undergo genetic testing. MEN1 syndrome should be considered in patients with family and/or personal histories of other endocrinopathies.[24]

Treatment / Management

Surgical resection is recommended for local disease but should also be considered for advanced disease.[20] Single sporadic insulinoma is curable by surgical intervention.[4] Surgeons have described several different surgical approaches. The most common of these is enucleation of the insulinoma and partial distal pancreatectomy.[3] Nowadays, laparoscopic resection of insulinoma is gaining in popularity with a shorter hospital stay and at least similar favorable outcomes.[25] Preoperatively the patients can be treated with diazoxide or somatostatin analogs as tolerated.[26][25] Robotic enucleation of insulinoma is being reported in some cases with good short and long-term outcomes compared to the laparoscopic approach.[27][28] Patients with MEN1 might need a different surgical approach, including subtotal pancreatectomy in addition to enucleation of the tumors. EUS-guided ethanol ablation has been attempted in poor surgical candidates and has been successful in some case series.[19][29][30][31]

Clinicians can attempt medical treatment before surgery in recurrent or malignant cases.[19] For initial insulinomas, dietary modification, diazoxide, everolimus, and MedicAlert bracelets are prudent steps.[24] Other strategies, including glucagon pen, somatostatin analogs, and steroids, are also considerations.[24]

If the tumor is unresectable, if the patient has metastatic disease, or if he is a poor surgical candidate, diazoxide is an option.[9] Diazoxide can decrease insulin release and enhance glycogenolysis. Around 60% of the patients become symptom-free.[32] The efficacy of octreotide in patients with refractory hypoglycemia is less predictable but remains an option for patients who do not respond to diazoxide. For patients with refractory symptoms and stable tumor volume, adjusting the dose of octreotide or getting surgical resection could be considered.[24]

For patients with refractory symptoms and increasing tumor volume, oncologic control with referral to specialty centers is recommended.[24] For patients with well-differentiated pancreatic neuroendocrine tumors (pNET), targeted therapy (everolimus or sunitinib) is a pharmaceutical option.[33] Everolimus inhibits the mammalian target of rapamycin.[34] Everolimus was found to improve median PFS in patients with low or intermediate-grade, advanced progressive pNET and a subgroup of functional pNET, including but not limited to insulinomas.[35][36] It can also help to control hypoglycemia in patients with malignant insulinoma.[19] Last, sunitinib is also recommended for progressive metastatic pNET, although only a small number of patients with insulinoma participated in these studies.[24][37]

For advanced disease, surgical resection could still be an option if most (90% or more) of the tumor is safely resectable or if the liver metastases seem to be resectable.[24] There has not been sufficient research on the different regimens of therapies to determine established choices.[24] Cytotoxic therapies such as 5-FU or temozolomide could be possibilities in patients with advanced pNET as palliative therapy.[24]

Some research suggests neoadjuvant therapy involving peptide receptor radionuclide therapy (PRRT) in unresectable or borderline resectable pNET because of survival benefits.[38][39] PRRT can also control hypoglycemia in malignant insulinomas, but its role in insulinoma treatment still needs more investigation.[19][38][40]

Differential Diagnosis

  • Persistent hyperinsulinemic hypoglycemia of infancy (PHHI)
  • Noninsulinoma pancreatogenous hypoglycemia syndrome (NIPHS)
  • Post-gastric bypass hypoglycemia
  • Factitious use of insulin
  • Sulfonylurea-induced hypoglycemia
  • Insulin autoimmune hypoglycemia
  • Non-islet-cell tumors that secrete insulin-like growth factors (IGF)
  • Nesidioblastosis[41]

Staging

Researchers have developed several classification and grading systems (WHO2010, ENETs, AJCC) for pNET. Most of them provide essential prognostic values.[19]

Newer classification systems include WHO2017 and AJCC 8 edition. AJCC (8th edition, 2017) includes ENETs definitions for T stage and prognostic stage grouping. Further modification of this staging system is available, which might provide better prognostic information.[42]

TNM Staging

  • TX - Tumor cannot be assessed
  • T1 - Tumor limited to the pancreas, less than 2 cm
  • T2 - Tumor limited to the pancreas, 2 to 4 cm
  • T3 - Tumor limited to the pancreas, greater than 4 cm; or tumor invading the duodenum or common bile duct
  • T4 - Tumor invasion of adjacent organs (e.g., stomach, spleen, colon, adrenal gland) or the walls of large vessels (celiac axis or the superior mesenteric artery)
  • NX - Regional lymph nodes cannot be assessed
  • N0 - No regional lymph node involvement
  • N1 - Regional lymph node involvement
  • M0 - No distant metastasis 
  • M1 - Distant metastasis 
  • M1a - Metastasis confined to the liver
  • M1b - Metastasis in at least one extrahepatic site (e.g., lung, ovary, nonregional lymph node, peritoneum, bone)
  • M1c - Both hepatic and extrahepatic metastases

Prognosis

In insulinoma patients, the 10-year survival rate is 88% after successful surgical removal, and 87.5% of patients get cured (being free of symptoms for at least six months) after surgery.[3][43][3] The 10-year survival in malignant insulinomas was 29% after successful surgical removal in one study, and the 5-year survival was 24% in another.[3][5] Patients with MEN1 syndrome or malignant insulinomas have a higher rate of failing initial surgery or experiencing recurrent disease.[3] The recurrence rate among patients with MEN1 is around 21% at 10 and 20 years, while those without MEN 1 are reported at 5% at ten years and 7% at 20 years.[3]

Complications

The complications associated with insulinoma include persistent hypoglycemia causing interference with daily activities, irreversible neurocognitive impairment, and even death.

Postoperative and Rehabilitation Care

Insulinoma patients without MEN1 should have a follow-up at 3 to 6 months post-resection.[4] Biochemical testing and imaging studies should be repeated at the follow-up visit. If cured, patients can be followed as needed if they experience recurrence of symptoms. Patients with persistent symptoms after surgery might have multiple tumors or MEN1 syndrome and should receive repeat imaging studies.[24] Patients with multiple insulinomas or MEN1 should be followed yearly, even if they continue to be cured post-resection.[4]

Deterrence and Patient Education

Patients and family members should receive education about recognizing symptoms associated with hypoglycemia and also be trained to use convenient ways to correct hypoglycemia quickly. Patients should follow up with endocrinology specialists for evaluation and treatment of insulinoma. Genetic testing is sometimes necessary.

Enhancing Healthcare Team Outcomes

The evaluation of insulinoma includes biochemical testing and localization with imaging studies. An interprofessional healthcare team, including primary care clinicians (MDs, DOs, NPs, and PAs), specialists (endocrinologists and oncologists), radiologists, surgeons, specialty-trained nurses, pharmacists, and sometimes genetic specialists, could facilitate better management of insulinomas. 

Patient education is paramount; therefore, every interprofessional team member must educate the patient whenever possible and is also responsible for tracking patient progress or lack thereof. Any changes in status should be recorded in the patient's health record and immediately reported to other team members so therapeutic changes can be implemented, if necessary. The length of follow-up required post-resection varies, and a timely treatment plan adjustment is more accessible if patients follow up as instructed. This kind of interprofessional teamwork and case management will improve outcomes for patients with insulinomas. [Level 5]


Details

Author

Fenghao Zhuo

Updated:

6/20/2023 10:35:41 PM

References


[1]

Minn AH, Kayton M, Lorang D, Hoffmann SC, Harlan DM, Libutti SK, Shalev A. Insulinomas and expression of an insulin splice variant. Lancet (London, England). 2004 Jan 31:363(9406):363-7     [PubMed PMID: 15070567]


[2]

Shin JJ, Gorden P, Libutti SK. Insulinoma: pathophysiology, localization and management. Future oncology (London, England). 2010 Feb:6(2):229-37. doi: 10.2217/fon.09.165. Epub     [PubMed PMID: 20146582]


[3]

Service FJ, McMahon MM, O'Brien PC, Ballard DJ. Functioning insulinoma--incidence, recurrence, and long-term survival of patients: a 60-year study. Mayo Clinic proceedings. 1991 Jul:66(7):711-9     [PubMed PMID: 1677058]


[4]

Salazar R, Wiedenmann B, Rindi G, Ruszniewski P. ENETS 2011 Consensus Guidelines for the Management of Patients with Digestive Neuroendocrine Tumors: an update. Neuroendocrinology. 2012:95(2):71-3. doi: 10.1159/000335600. Epub 2012 Feb 15     [PubMed PMID: 22262042]

Level 3 (low-level) evidence

[5]

Placzkowski KA,Vella A,Thompson GB,Grant CS,Reading CC,Charboneau JW,Andrews JC,Lloyd RV,Service FJ, Secular trends in the presentation and management of functioning insulinoma at the Mayo Clinic, 1987-2007. The Journal of clinical endocrinology and metabolism. 2009 Apr;     [PubMed PMID: 19141587]


[6]

Pratò S, Didonna V, Garletti F, Marfia G, Barbaresi A, Palumbo F, Garzia E, Ciniglio Appiani G, Riboldi L, Vigna L. Loss of consciousness in a helicopter pilot as plausible first sign of insulinoma: a case report. La Medicina del lavoro. 2022 Feb 22:113(1):e2022007. doi: 10.23749/mdl.v113i1.12037. Epub 2022 Feb 22     [PubMed PMID: 35226655]

Level 3 (low-level) evidence

[7]

Karanth JB, Pai V, Maribashetti K. Pancreatic neuroendocrine tumour-insulinoma masquerading as a psychiatric illness. BMJ case reports. 2022 Jun 14:15(6):. doi: 10.1136/bcr-2022-249698. Epub 2022 Jun 14     [PubMed PMID: 35701018]

Level 3 (low-level) evidence

[8]

Vinik AI, Woltering EA, Warner RR, Caplin M, O'Dorisio TM, Wiseman GA, Coppola D, Go VL, North American Neuroendocrine Tumor Society (NANETS). NANETS consensus guidelines for the diagnosis of neuroendocrine tumor. Pancreas. 2010 Aug:39(6):713-34. doi: 10.1097/MPA.0b013e3181ebaffd. Epub     [PubMed PMID: 20664471]

Level 3 (low-level) evidence

[9]

Cryer PE, Axelrod L, Grossman AB, Heller SR, Montori VM, Seaquist ER, Service FJ, Endocrine Society. Evaluation and management of adult hypoglycemic disorders: an Endocrine Society Clinical Practice Guideline. The Journal of clinical endocrinology and metabolism. 2009 Mar:94(3):709-28. doi: 10.1210/jc.2008-1410. Epub 2008 Dec 16     [PubMed PMID: 19088155]

Level 1 (high-level) evidence

[10]

ASGE Standards of Practice Committee, Eloubeidi MA, Decker GA, Chandrasekhara V, Chathadi KV, Early DS, Evans JA, Fanelli RD, Fisher DA, Foley K, Hwang JH, Jue TL, Lightdale JR, Pasha SF, Saltzman JR, Sharaf R, Shergill AK, Cash BD, DeWitt JM. The role of endoscopy in the evaluation and management of patients with solid pancreatic neoplasia. Gastrointestinal endoscopy. 2016 Jan:83(1):17-28. doi: 10.1016/j.gie.2015.09.009. Epub     [PubMed PMID: 26706297]


[11]

Virgolini I, Traub-Weidinger T, Decristoforo C. Nuclear medicine in the detection and management of pancreatic islet-cell tumours. Best practice & research. Clinical endocrinology & metabolism. 2005 Jun:19(2):213-27     [PubMed PMID: 15763696]


[12]

Christ E, Wild D, Forrer F, Brändle M, Sahli R, Clerici T, Gloor B, Martius F, Maecke H, Reubi JC. Glucagon-like peptide-1 receptor imaging for localization of insulinomas. The Journal of clinical endocrinology and metabolism. 2009 Nov:94(11):4398-405. doi: 10.1210/jc.2009-1082. Epub 2009 Oct 9     [PubMed PMID: 19820010]


[13]

Luo Y, Pan Q, Yao S, Yu M, Wu W, Xue H, Kiesewetter DO, Zhu Z, Li F, Zhao Y, Chen X. Glucagon-Like Peptide-1 Receptor PET/CT with 68Ga-NOTA-Exendin-4 for Detecting Localized Insulinoma: A Prospective Cohort Study. Journal of nuclear medicine : official publication, Society of Nuclear Medicine. 2016 May:57(5):715-20. doi: 10.2967/jnumed.115.167445. Epub 2016 Jan 21     [PubMed PMID: 26795291]


[14]

Shah R, Garg R, Majmundar M, Purandare N, Malhotra G, Patil V, Ramteke-Jadhav S, Lila A, Shah N, Bandgar T. Exendin-4-based imaging in insulinoma localization: Systematic review and meta-analysis. Clinical endocrinology. 2021 Aug:95(2):354-364. doi: 10.1111/cen.14406. Epub 2021 Jan 21     [PubMed PMID: 33386617]

Level 1 (high-level) evidence

[15]

Imperiale A, Boursier C, Sahakian N, Ouvrard E, Chevalier E, Sebag F, Addeo P, Taïeb D. Value of (68)Ga-DOTATOC and Carbidopa-Assisted (18)F-DOPA PET/CT for Insulinoma Localization. Journal of nuclear medicine : official publication, Society of Nuclear Medicine. 2022 Mar:63(3):384-388. doi: 10.2967/jnumed.121.262401. Epub 2021 Jul 16     [PubMed PMID: 34272321]


[16]

Murakami T, Fujimoto H, Hamamatsu K, Yamauchi Y, Kodama Y, Fujita N, Fujikura J, Shimizu Y, Nakamoto Y, Kimura H, Saji H, Inagaki N. Distinctive detection of insulinoma using [(18)F]FB(ePEG12)12-exendin-4 PET/CT. Scientific reports. 2021 Jul 22:11(1):15014. doi: 10.1038/s41598-021-94595-6. Epub 2021 Jul 22     [PubMed PMID: 34294854]


[17]

Khashab MA, Yong E, Lennon AM, Shin EJ, Amateau S, Hruban RH, Olino K, Giday S, Fishman EK, Wolfgang CL, Edil BH, Makary M, Canto MI. EUS is still superior to multidetector computerized tomography for detection of pancreatic neuroendocrine tumors. Gastrointestinal endoscopy. 2011 Apr:73(4):691-6. doi: 10.1016/j.gie.2010.08.030. Epub 2010 Nov 10     [PubMed PMID: 21067742]


[18]

Marx M, Trosic-Ivanisevic T, Caillol F, Demartines N, Schoepfer A, Pesenti C, Ratone JP, Robert M, Giovannini M, Godat S. EUS-guided radiofrequency ablation for pancreatic insulinoma: experience in 2 tertiary centers. Gastrointestinal endoscopy. 2022 Jun:95(6):1256-1263. doi: 10.1016/j.gie.2021.11.045. Epub 2021 Dec 10     [PubMed PMID: 34902374]


[19]

Falconi M, Eriksson B, Kaltsas G, Bartsch DK, Capdevila J, Caplin M, Kos-Kudla B, Kwekkeboom D, Rindi G, Klöppel G, Reed N, Kianmanesh R, Jensen RT, Vienna Consensus Conference participants. ENETS Consensus Guidelines Update for the Management of Patients with Functional Pancreatic Neuroendocrine Tumors and Non-Functional Pancreatic Neuroendocrine Tumors. Neuroendocrinology. 2016:103(2):153-71. doi: 10.1159/000443171. Epub 2016 Jan 5     [PubMed PMID: 26742109]

Level 3 (low-level) evidence

[20]

Jonkers YM, Claessen SM, Perren A, Schmitt AM, Hofland LJ, de Herder W, de Krijger RR, Verhofstad AA, Hermus AR, Kummer JA, Skogseid B, Volante M, Voogd AC, Ramaekers FC, Speel EJ. DNA copy number status is a powerful predictor of poor survival in endocrine pancreatic tumor patients. Endocrine-related cancer. 2007 Sep:14(3):769-79     [PubMed PMID: 17914106]


[21]

Câmara-de-Souza AB,Toyoshima MTK,Giannella ML,Freire DS,Camacho CP,Lourenço DM Jr,Rocha MS,Bacchella T,Jureidini R,Machado MCC,Almeida MQ,Pereira MAA, Insulinoma: A retrospective study analyzing the differences between benign and malignant tumors. Pancreatology : official journal of the International Association of Pancreatology (IAP) ... [et al.]. 2018 Apr;     [PubMed PMID: 29452754]

Level 2 (mid-level) evidence

[22]

Kurakawa KI, Okada A, Manaka K, Konishi T, Jo T, Ono S, Uda K, Michihata N, Matsui H, Fushimi K, Yamaguchi S, Yamauchi T, Nangaku M, Yasunaga H, Kadowaki T. Clinical Characteristics and Incidences of Benign and Malignant Insulinoma Using a National Inpatient Database in Japan. The Journal of clinical endocrinology and metabolism. 2021 Nov 19:106(12):3477-3486. doi: 10.1210/clinem/dgab559. Epub     [PubMed PMID: 34343300]


[23]

Sada A, Yamashita TS, Glasgow AE, Habermann EB, Thompson GB, Lyden ML, Dy BM, Halfdanarson TR, Vella A, McKenzie TJ. Comparison of benign and malignant insulinoma. American journal of surgery. 2021 Feb:221(2):437-447. doi: 10.1016/j.amjsurg.2020.08.003. Epub 2020 Aug 16     [PubMed PMID: 32873371]


[24]

Kunz PL, Reidy-Lagunes D, Anthony LB, Bertino EM, Brendtro K, Chan JA, Chen H, Jensen RT, Kim MK, Klimstra DS, Kulke MH, Liu EH, Metz DC, Phan AT, Sippel RS, Strosberg JR, Yao JC, North American Neuroendocrine Tumor Society. Consensus guidelines for the management and treatment of neuroendocrine tumors. Pancreas. 2013 May:42(4):557-77. doi: 10.1097/MPA.0b013e31828e34a4. Epub     [PubMed PMID: 23591432]

Level 3 (low-level) evidence

[25]

Su AP, Ke NW, Zhang Y, Liu XB, Hu WM, Tian BL, Zhang ZD. Is laparoscopic approach for pancreatic insulinomas safe? Results of a systematic review and meta-analysis. The Journal of surgical research. 2014 Jan:186(1):126-34. doi: 10.1016/j.jss.2013.07.051. Epub 2013 Aug 19     [PubMed PMID: 23992857]

Level 1 (high-level) evidence

[26]

Gilliaux Q, Bertrand C, Hanon F, Donckier JE. Preoperative treatment of benign insulinoma: diazoxide or somatostatin analogues? Acta chirurgica Belgica. 2022 Apr:122(2):136-139. doi: 10.1080/00015458.2020.1765676. Epub 2020 May 18     [PubMed PMID: 32375590]


[27]

Liang M, Jiang J, Dai H, Hong X, Han X, Cong L, Tong A, Li F, Luo Y, Liu W, Zhou L, Di W, Wu W, Zhao Y. Robotic enucleation for pediatric insulinoma with MEN1 syndrome: a case report and literature review. BMC surgery. 2018 Jun 19:18(1):44. doi: 10.1186/s12893-018-0376-5. Epub 2018 Jun 19     [PubMed PMID: 29921249]

Level 3 (low-level) evidence

[28]

Belfiori G, Wiese D, Partelli S, Wächter S, Maurer E, Crippa S, Falconi M, Bartsch DK. Minimally Invasive Versus Open Treatment for Benign Sporadic Insulinoma Comparison of Short-Term and Long-Term Outcomes. World journal of surgery. 2018 Oct:42(10):3223-3230. doi: 10.1007/s00268-018-4628-4. Epub     [PubMed PMID: 29691623]


[29]

Dąbkowski K, Gajewska P, Walter K, Londzin-Olesik M, Białek A, Andrysiak-Mammos E, Kos-Kudła B, Starzyńska T. Successful EUS-guided ethanol ablation of insulinoma, four-year follow-up. Case report and literature review. Endokrynologia Polska. 2017:68(4):472-479. doi: 10.5603/EP.2017.0053. Epub     [PubMed PMID: 28819950]

Level 3 (low-level) evidence

[30]

Alyusuf EY, Ekhzaimy AA, Rivera JA. Radiofrequency Ablation as a Primary Therapy for Benign Functioning Insulinoma. AACE clinical case reports. 2021 Mar-Apr:7(2):153-157. doi: 10.1016/j.aace.2020.12.003. Epub 2020 Dec 28     [PubMed PMID: 34095476]

Level 3 (low-level) evidence

[31]

Jayawickreme KP, Muthukuda DT, Kariyawasam C, Piyarisi L, Abeywickrama BA. A rare case of insulinoma presenting with deep vein thrombosis, successfully treated with minimally invasive procedures including microwave ablation. Endocrinology, diabetes & metabolism case reports. 2022 May 1:2022():. pii: 21-0127. doi: 10.1530/EDM-21-0127. Epub 2022 May 1     [PubMed PMID: 35510504]

Level 3 (low-level) evidence

[32]

Gill GV, Rauf O, MacFarlane IA. Diazoxide treatment for insulinoma: a national UK survey. Postgraduate medical journal. 1997 Oct:73(864):640-1     [PubMed PMID: 9497974]

Level 3 (low-level) evidence

[33]

Singh S, Sivajohanathan D, Asmis T, Cho C, Hammad N, Law C, Wong R, Zbuk K. Systemic therapy in incurable gastroenteropancreatic neuroendocrine tumours: a clinical practice guideline. Current oncology (Toronto, Ont.). 2017 Aug:24(4):249-255. doi: 10.3747/co.24.3634. Epub 2017 Aug 31     [PubMed PMID: 28874893]

Level 1 (high-level) evidence

[34]

Mujica-Mota R, Varley-Campbell J, Tikhonova I, Cooper C, Griffin E, Haasova M, Peters J, Lucherini S, Talens-Bou J, Long L, Sherriff D, Napier M, Ramage J, Hoyle M. Everolimus, lutetium-177 DOTATATE and sunitinib for advanced, unresectable or metastatic neuroendocrine tumours with disease progression: a systematic review and cost-effectiveness analysis. Health technology assessment (Winchester, England). 2018 Sep:22(49):1-326. doi: 10.3310/hta22490. Epub     [PubMed PMID: 30209002]

Level 1 (high-level) evidence

[35]

Lombard-Bohas C, Yao JC, Hobday T, Van Cutsem E, Wolin EM, Panneerselvam A, Stergiopoulos S, Shah MH, Capdevila J, Pommier R. Impact of prior chemotherapy use on the efficacy of everolimus in patients with advanced pancreatic neuroendocrine tumors: a subgroup analysis of the phase III RADIANT-3 trial. Pancreas. 2015 Mar:44(2):181-9. doi: 10.1097/MPA.0000000000000262. Epub     [PubMed PMID: 25479584]


[36]

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. The New England journal of medicine. 2011 Feb 10:364(6):514-23. doi: 10.1056/NEJMoa1009290. Epub     [PubMed PMID: 21306238]


[37]

Raymond E, Dahan L, Raoul JL, Bang YJ, Borbath I, Lombard-Bohas C, Valle J, Metrakos P, Smith D, Vinik A, Chen JS, Hörsch D, Hammel P, Wiedenmann B, Van Cutsem E, Patyna S, Lu DR, Blanckmeister C, Chao R, Ruszniewski P. Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. The New England journal of medicine. 2011 Feb 10:364(6):501-13. doi: 10.1056/NEJMoa1003825. Epub     [PubMed PMID: 21306237]


[38]

Hicks RJ, Kwekkeboom DJ, Krenning E, Bodei L, Grozinsky-Glasberg S, Arnold R, Borbath I, Cwikla J, Toumpanakis C, Kaltsas G, Davies P, Hörsch D, Tiensuu Janson E, Ramage J, Antibes Consensus Conference participants. ENETS Consensus Guidelines for the Standards of Care in Neuroendocrine Neoplasia: Peptide Receptor Radionuclide Therapy with Radiolabeled Somatostatin Analogues. Neuroendocrinology. 2017:105(3):295-309. doi: 10.1159/000475526. Epub 2017 Apr 13     [PubMed PMID: 28402980]

Level 3 (low-level) evidence

[39]

Ezziddin S, Khalaf F, Vanezi M, Haslerud T, Mayer K, Al Zreiqat A, Willinek W, Biersack HJ, Sabet A. Outcome of peptide receptor radionuclide therapy with 177Lu-octreotate in advanced grade 1/2 pancreatic neuroendocrine tumours. European journal of nuclear medicine and molecular imaging. 2014 May:41(5):925-33. doi: 10.1007/s00259-013-2677-3. Epub 2014 Feb 7     [PubMed PMID: 24504504]


[40]

Sansovini M, Severi S, Ambrosetti A, Monti M, Nanni O, Sarnelli A, Bodei L, Garaboldi L, Bartolomei M, Paganelli G. Treatment with the radiolabelled somatostatin analog Lu-DOTATATE for advanced pancreatic neuroendocrine tumors. Neuroendocrinology. 2013:97(4):347-54. doi: 10.1159/000348394. Epub 2013 May 22     [PubMed PMID: 23392072]


[41]

Doi S, Yamada T, Kito Y, Obara S, Fujii Y, Nishimura T, Kato T, Nakayama H, Tsutsumi M, Okamura R. Adult-Onset Focal Nesidioblastosis With Nodular Formation Mimicking Insulinoma. Journal of the Endocrine Society. 2022 Jan 1:6(1):bvab185. doi: 10.1210/jendso/bvab185. Epub 2021 Dec 11     [PubMed PMID: 35024540]


[42]

Luo G,Javed A,Strosberg JR,Jin K,Zhang Y,Liu C,Xu J,Soares K,Weiss MJ,Zheng L,Wolfgang CL,Cives M,Wong J,Wang W,Sun J,Shao C,Wang W,Tan H,Li J,Ni Q,Shen L,Chen M,He J,Chen J,Yu X, Modified Staging Classification for Pancreatic Neuroendocrine Tumors on the Basis of the American Joint Committee on Cancer and European Neuroendocrine Tumor Society Systems. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2017 Jan 20;     [PubMed PMID: 27646952]


[43]

Peltola E, Hannula P, Huhtala H, Metso S, Sand J, Laukkarinen J, Tiikkainen M, Sirén J, Soinio M, Nuutila P, Moilanen L, Laaksonen DE, Ebeling T, Arola J, Schalin-Jäntti C, Jaatinen P. Long-term morbidity and mortality in patients diagnosed with an insulinoma. European journal of endocrinology. 2021 Sep 1:185(4):577-586. doi: 10.1530/EJE-21-0230. Epub 2021 Sep 1     [PubMed PMID: 34374651]