Insulinoma

Back

Practice Essentials

Insulinomas are the most common cause of hypoglycemia resulting from endogenous hyperinsulinism. Approximately 90-95% of insulinomas are benign, and long-term cure with total resolution of preoperative symptoms is expected after complete resection.[1] See the image below.



View Image

CT scan image with oral and intravenous contrast in a patient with biochemical evidence of insulinoma. The 3-cm contrast-enhancing neoplasm (arrow) is....

Signs and symptoms

Insulinomas are characterized clinically by the Whipple triad, as follows:

About 85% of patients with insulinoma present with one of the following symptoms of hypoglycemia:

Hypoglycemia can also result in the following:

See Clinical Presentation for more detail.

Diagnosis

Lab studies

Failure of endogenous insulin secretion to be suppressed by hypoglycemia is the hallmark of an insulinoma. Thus, the finding of inappropriately elevated levels of insulin in the face of hypoglycemia is the key to diagnosis.

The biochemical diagnosis of insulinoma is established in 95% of patients during prolonged fasting (up to 72 h) when the following results are found:

Imaging studies

Insulinomas can be located with the following imaging modalities:

See Workup for more detail.

Management

Pharmacologic therapy

Pharmacologic treatment is designed to prevent hypoglycemia and, in patients with malignant tumors, to reduce the tumor burden. Agents used in this therapy include the following:

Surgery

Even when metastases are found, surgical excision is often feasible before any medical, chemotherapeutic, or other interventional therapy is considered. Resect all gross disease; this would include performing wedge resections of hepatic metastases

See Treatment and Medication for more detail.

Background

Insulinomas are the most common cause of hypoglycemia resulting from endogenous hyperinsulinism. In a large single-center series of 125 patients with neuroendocrine tumors, insulinomas constituted the majority of cases (55%), followed by gastrinomas (36%), VIPomas (vasoactive intestinal polypeptide tumor) (5%), and glucagonomas (3%).[8]

In 1927, Wilder established the association between hyperinsulinism and a functional islet cell tumor.[9] In 1929, Graham achieved the first surgical cure of an islet cell adenoma. Insulinomas can be difficult to diagnose. It was not uncommon for patients to have been misdiagnosed with psychiatric illnesses or seizure disorders before insulinoma was recognized.

Pathophysiology

An insulinoma is a neuroendocrine tumor, deriving mainly from pancreatic islet cells, that secretes insulin. Some insulinomas also secrete other hormones, such as gastrin, 5-hydroxyindolic acid, adrenocorticotropic hormone (ACTH), glucagon, human chorionic gonadotropin, and somatostatin. The tumor may secrete insulin in short bursts, causing wide fluctuations in blood levels.

About 90% of insulinomas are benign. Approximately 10% of insulinomas are malignant (metastases are present). Approximately 10% of patients have multiple insulinomas; of patients with multiple insulinomas, 50% have multiple endocrine neoplasia type 1 (MEN 1). Insulinomas are associated with MEN 1 in 5% of patients. On the other hand, 21% of patients with MEN 1 develop insulinomas. Because of the association of insulinomas with MEN 1, consideration should be given to screening family members of insulinoma patients for MEN 1.

An in vitro study by Henquin et al identified three distinct patterns of insulin secretion by insulinomas, as follows[10] :

Increased expression of the phosphorylated mechanistic target of rapamycin (p-mTOR) signaling pathway and its downstream serine/threonine kinase p70S6k has been observed in insulinoma tumor specimens.[11] This discovery has led to studies exploring mTOR inhibitors as new therapeutic options.

Epidemiology

United States

Insulinomas are the most common pancreatic endocrine tumors. The incidence is 3-10 cases per million people per year.[12] These make up 55% of neuroendocrine tumors.

International

Exact data for international incidence of insulinomas are not available. One source from Northern Ireland reported an annual incidence of 1 case per million persons. A study from Iran found 68 cases in a time span of 20 years in a university in Tehran.[13] A 10-year single-institution study from Spain of 49 consecutive patients who underwent laparoscopic surgery for neuroendocrine pancreatic tumors included 23 cases of insulinoma.[7] These reports may be an underestimate.

Mortality/Morbidity

Pancreatic fistula is the most common complication of surgery for insulinoma,[14] with a Other complications associated with surgery for insulinoma surgery include pseudocyst, intra-abdominal abscess, pancreatitis, hemorrhage, and diabetes. The postoperative mortality rate in another series of 117 insulinoma patients was 7.7%.[15] The median survival in metastatic disease to the liver ranges from 16-26 months.

Race-, Sex-, and Age-related Demographics

Insulinomas have been reported in persons of all races. No racial predilection appears to exist.

The male-to-female ratio for insulinomas is 2:3.

The median age at diagnosis is about 47 years, except in insulinoma patients with MEN 1, in whom the median age is the mid 20s. In one series, patients with benign disease were younger (mean age of 38 y) than those with metastases (mean age of 52 y). The age range for peak incidence of insulinoma is between 30 and 60 years.

History and Physical Examination

Features of the history in patients with insulinoma are as follows:

Most patients with insulinoma have normal physical examination findings.

Causes

The genetic changes in neuroendocrine tumors are under investigation.[19] The gene of multiple endocrine neoplasia type 1, an autosomal dominant disease, is called MEN1 and maps to band 11q13. MEN1 is thought to function as a tumor suppressor gene. Data suggest that the MEN1 gene also is involved in the pathogenesis of at least one third of sporadic neuroendocrine tumors. Researchers were able to detect loss of heterozygosity in band 11q13 in DNA samples from resected insulinoma tissue by using fluorescent microsatellite analysis.

In a study of 12 children with insulinoma, four cases showed heterozygous mutations of MEN1 on 11q. Aneuploidy of chromosome 11 and other chromosomes was common in both MEN1 and non-MEN1 insulinomas.[20]

One study showed k-ras mutation to be present in 23% of insulinomas.

Approach Considerations

The classic hallmark of insulinomas is the Whipple triad, which consists of the following:[14]

Common diagnostic criteria include documentation of blood glucose level < 50 mg/dl with hypoglycemic symptoms, with relief of symptoms after eating, and absence of sulfonylurea on plasma assays. The classic diagnostic criteria include demonstration of the following during a supervised fast:

Once the diagnosis of insulinoma has been established biochemically, imaging studies are used to localize the tumor.

Laboratory Studies

Failure of endogenous insulin secretion to be suppressed by hypoglycemia is the hallmark of an insulinoma. Thus, the finding of inappropriately elevated levels of insulin in the face of hypoglycemia is the key to diagnosis. Considering the reference range, the fasting plasma levels of insulin, C-peptide, and, to a lesser degree, proinsulin need not be elevated in insulinoma patients in absolute terms.

A prolonged supervised fast in hospitalized patients provides the most reliable results. During this test, levels of plasma glucose, insulin, C peptide, and proinsulin are measured every 6 hours until the plasma glucose level falls to 60 mg/dl or less, at which point the testing interval is reduced to every 1–2 h. The fast is terminated when the plasma glucose level is 45 mg/dl or less and the patient has symptoms and signs of hypoglycemia

Traditionally, a 72-hour fast has been used. However, with current assays, 90–95% of insulinomas can be diagnosed with a supervised fast of 48 hours.[14]

The calculation of ratios of insulin (µU/mL) to plasma glucose (mg/dL) during the fast is diagnostic: Healthy patients maintain a ratio of less than 0.25 (obese patients may have a slightly higher rate), whereas in patients with insulinoma, the ratio rises during fasting.

In a study from the Netherlands, a positive Whipple triad on a prolonged fasting test, in combination with an insulin/C-peptide ratio < 1, had a sensitivity of 88.9% and a specificity of 100% for the diagnosis of insulinoma.[25]

The biochemical diagnosis is established in 95% of patients during prolonged fasting when the following parameters are found:

Stimulation tests are no longer recommended. The intravenous administration of tolbutamide, glucagon, or calcium can be hazardous, as it may induce prolonged and refractory hypoglycemia.

The presence of multiple endocrine neoplasia type 1 (MEN 1) must be evaluated by excluding the following:

Imaging Studies

Start imaging studies only after the diagnosis has been confirmed biochemically, because 80% of insulinomas are less than 2 cm in size and may not be visible by computed tomography (CT) scanning or transabdominal ultrasonography.

Successful preoperative tumor localization is achieved in about 60% of patients.[27] Some experienced surgeons perform only transabdominal ultrasound preoperatively. Other surgeons argue that the preoperative localization of insulinomas is not necessary at all because surgical exploration and intraoperative ultrasonography identify more than 90% of tumors.[28] Thus, the extent to which one attempts to define the anatomy of the beta cell lesion before surgery is a matter of judgment.

Computed tomography and magnetic resonance imaging

Helical or multislice CT has 82-94% sensitivity. In one study, dual-phase helical CT proved more sensitive than single-phase for detection of insulinoma; in addition, image acquisition in the arterial phase proved more helpful than acquisition during the portal-venous phase.[29] See the image below.



View Image

CT scan image with oral and intravenous contrast in a patient with biochemical evidence of insulinoma. The 3-cm contrast-enhancing neoplasm (arrow) is....

Zhu et al reported that the addition of whole-pancreas perfusion to biphasic contrast-enhanced CT (CECT) may improve the detection of insulinomas. In their study of 79 consecutive cases, mean blood flow values of both insulinomas and insulinoma-harboring regions were statistically significantly higher (P < 0.01, for both) than those of tumor-free pancreatic parenchyma. The addition of perfusion allowed correct diagnosis of nine of 46 tumors (19.6%) in which biphasic CT was negative or indeterminate.[30]

In a subsequent prospective study, however, Zhu et al reported that volume perfusion CT (VPCT) is significantly more accurate than CECT for insulinoma detection, and 3 Tesla magnetic resonance imaging (3T MRI) with diffusion-weighted imaging (DWI) is better still. Lesions were more conspicuous on MRI than on VPCT, and conspicuity with both was better than with CECT. Tumor-to–pancreatic duct distance was better appreciated on MRI than on VPCT and CECT.[5]

Arteriography

The accuracy of selective arteriography is 82%, although affected by a false-positive rate of 5%. Many experts see it as the best overall preoperative localization procedure.

Arteriography with catheterization of small arterial branches of the celiac system combined with calcium injections (which stimulate insulin release from neoplastic tissue but not from normal islets), and simultaneous measurements of hepatic vein insulin during each selective calcium injection localizes tumors in 47% of patients.

Nuclear medicine

Insulinomas have been shown to have a very high density of glucagon-like peptide–1 (GLP-1) receptors, and radiolabeling GLP-1 analogs (eg, exendin-4) with appropriate radioisotopes can provide noninvasive localization of benign insulinomas.[31, 32] For example,indium-111(111In)–labeled exendin-4 (111In-DOTA-exendin-4) has successfully been used to localize small insulinomas both preoperatively and, with the use of a gamma probe, intraoperatively.[33] Luo et al reported that PET/CT scanning with 68Ga-NOTA–exendin-4 correctly detected insulinomas in 42 of 43 patients.[34]

Somatostatin receptor target imaging can diagnose malignant insulinomas missed by GLP-1 target imaging because these tumors often lack GLP-1 receptors.[35] Nockel et al studied the accuracy of galium68 (68Ga)–DOTA-(Tyr3)-octreotate (Ga-DOTATATE) PET/CT for localizing insulinoma and suggested that this technique may be considered as an adjunct imaging study when all other imaging studies are negative and when a minimally invasive surgical approach is planned.[6]

Ultrasonography

Endoscopic ultrasonography detects 77% of insulinomas in the pancreas.[2, 3, 4] The yield can be higher if it is done in combination with CT scan. A majority of sporadic insulinomas will be detected and localized by a combination of these two investigative means. Note the following:

See the image below.



View Image

Endoscopic ultrasonography in a patient with an insulinoma. The hypoechoic neoplasm (arrows) is seen in the body of the pancreas anterior to the splen....

Other Tests

Other tests used in the localization of insulinoma are as follows:

Histologic Findings

Insulinomas are solitary tumors in 90% of patients. In MEN 1 syndrome, multiple microinsulinomas and macroinsulinomas are found, although hypoglycemia may be caused by a single tumor. The tumors are distributed evenly throughout the pancreas. Tumor size does not relate to the severity of clinical symptoms. Ectopic insulinomas may be found in the ligament of Treitz.

No histologic criteria are available to distinguish benign from malignant insulinomas. Malignant tumors are usually larger (average size 6.2 cm), and a third of them have metastasized to the liver. Insulinoma tumor cells contain less insulin and secretory granules than normal B cells but higher levels of proinsulin. Atypical granules, or even agranular cells, are frequent. The clinical response to diazoxide and somatostatin correlates with the frequency and type of granules.

Staging

A tumor-node-metastasis (TNM) staging system has not been validated yet for insulinoma tumors. The staging used is similar for exocrine and endocrine tumors of pancreas.

Medical Care

Medical therapy is indicated in patients with malignant insulinomas and in those who will not or cannot undergo surgery. These measures are designed to prevent hypoglycemia and, in patients with malignant tumors, to reduce the tumor burden. In malignant insulinomas, dietary therapy with frequent oral feedings or enteral feedings may control mild symptoms of hypoglycemia. A trial of glucagon may be attempted to control hypoglycemia.

Diazoxide is related to the thiazide diuretics and reduces insulin secretion. Adverse effects include sodium retention, a tendency to congestive cardiac failure, and hirsutism. Prescribe hydrochlorothiazide to counteract the edema and hyperkalemia secondary to diazoxide and to potentiate its hyperglycemic effect.

Of patients with insulinoma, 50% may benefit from the somatostatin analog octreotide to prevent hypoglycemia.[36] The effect of the therapy depends on the presence of somatostatin receptor subtype 2 on insulinoma tumor cells. Use of the somatostatin analogs lanreotide and pasireotide have also been reported.[1, 37]

As studies have shown, an OctreoScan is not a prerequisite before starting octreotide treatment. In patients with insulinoma and a negative scan finding, somatostatin decreased insulin levels significantly and lowered the incidence of hypoglycemic events.[38]  

Indications for chemotherapy include progressive disease with an increase of greater than 25% of the main tumor masses in a follow-up period of 12 months, or tumor symptoms not treatable with other methods. Combination regimens have achieved better results than single agents.

The current medical treatment choices are those used for any metastatic neuroendocrine gastroenteric pancreatic tumor. Options are as follows:

There is a single case report of successful control of intractable hypoglycemia in an elderly man with metastatic insulinoma through the use of oral rapamycin (sirolimus), 2 mg/d.[41]

The mTOR inhibitor everolimus is approved for treatment of locally advanced or metastatic neuroendocrine tumors of pancreatic origin. In a French study, everolimus therapy normalized blood glucose levels in 11 of 12 patients with metastatic insulinoma and refractory hypoglycemia, with the therapeutic effect maintained for a median duration of 6.5 months (range 1-35+ months). However, three patients discontinued everolimus because of cardiac and/or pulmonary adverse events, which led to two deaths.[42]

Ablation

Ablation is a nonsurgical option that has had some long-term success.CT-guided radiofrequency ablation has been used successfully to treat insulinoma in an elderly patient whose hypoglycemia that was refractory to diazoxide, and who was not a candidate for surgery because of comorbidities and poor physical condition.[43] In addition to radiofrequency ablation, successful results have been attained with embolization and with ethanol, among other techniques.[44, 45]

Surgical Care

Because insulinoma resection achieves cure in 90% of patients, it is currently the therapy of choice. Enucleation is the preferred technique.

Preoperative management

Preoperative management is as follows:

Tumor location

If an open procedure is selected, fully expose the pancreas, including a wide Kocher maneuver to allow complete bimanual palpation.

Compared with open procedures, laparoscopy has been found to permit equivalent oncologic resection, along with decreased postoperative pain, better cosmetic results, shorter hospital stay, and a shorter postoperative recovery period.[47] A large study from Spain showed laparoscopic surgery to be safe and effective in benign and malignant tumor resection.[7]

Laparoscopic enucleation techniques, also in combination with preservation of the spleen for distal pancreatic tumors, have been described.[48] Because of the small likelihood that a tumor that presents without metastatic spread is malignant, insulinomas may be removed by enucleation.[49] Care should be taken to achieve total tumor capsule removal to prevent tumor recurrence.

If enucleation is not possible, a larger pancreatic resection including pancreaticoduodenectomy may be necessary. This should only rarely be required. When metastatic insulinoma is found on a patient's initial presentation, the organ spread is to liver and sometimes to bone.

Other surgical considerations are as follows:

Metastatic disease

Insulinomas are found to be metastatic at surgery in about 5-10% of patients. It would be extremely uncommon for metastases to develop in a case in which only a solitary lesion was found on initial presentation.

Patients who are responsive to diazoxide should be continued on it while more invasive imaging studies are performed, before repetitive surgery is considered. If the patient is not responsive (5-10%) or if drug intolerance is present and ectopic disease is excluded, a blind distal two-thirds pancreatectomy may be performed. However, this procedure has only a 25% success rate.

Most authorities recommend serial sectioning during resection. Tumors that are not found at surgery normally are located in the pancreatic head (54%), body (20%), and tail (14%).

Even when metastases are found, surgical excision is often feasible before any medical, chemotherapeutic, or other interventional therapy is considered. Resect all gross disease, including wedge resections of hepatic metastases. Avoid ligation of the hepatic artery in case further regional infusion therapy becomes necessary.

Consultations

Consult with the anesthetist to plan for precise preoperative and intraoperative blood glucose monitoring. The approach should be multidisciplinary, with an endocrinologist, surgeon, and anesthesiologist.

Diet and Activity

Considerations include the following:

Guidelines Summary

Guidelines contributor: Evan S Ong, MD, MS Assistant Professor of Surgery, Section of Surgical Oncology, University of Arizona College of Medicine

The following organizations have issued clinical guidelines for the management of gastroenteropancreatic neuroendocrine tumors (GEP-NETs):

Grading and Staging

Grading schemes for neuroendocrine tumors (NETs) use mitotic count; the level of the nuclear protein Ki-67, which is associated with cellular proliferation; and assessment of necrosis. The World Health Organization (WHO) and the European Neuroendocrine Tumor Society (ENETS)  both incorporate mitotic count and Ki-67 proliferation for the classification of gastroenteropancreatic NETs (GEP-NETs).[50, 51, 52]

Tumors fall into one of the following three grades:

The National Comprehensive Cancer Network (NCCN) recommends that tumor differentiation, mitotic rate, and Ki-67 rate be included in the pathology report and that the specific classification and grading scheme be noted to avoid confusion. Clinicians are advised to view histologic grade as a general guide and use clinical judgment to make treatment decisions, particularly in cases of discordance between differentiation and Ki-67 proliferation results.[53]

NCCN guidelines recommend staging according to the 7th edition of the American Joint Committee on Cancer's AJCC Cancer Staging Manual.[54]

For staging of GEP-NETs, the European Society for Medical Oncology (ESMO) guidelines, updated in 2012, utilize the tumor-node-metastasis (TNM) classification created by the ENETS and the 2010 WHO grading system.[55]

In 2012, the UK and Ireland Neuroendocrine Tumour Society (UKI NETS) released updated guidelines for the management of GEP-NETs. Recommendations for grading and staging are as follows[56] :

In 2013, the North American Neuroendocrine Tumor Society (NANETS) concluded that while the criteria differ among the various classification systems, the underlying data are similar and pathology reports should include notation of the systems and parameters used to assign the grade and stage.[57]

Treatment

Treatment for locoregional disease

National Comprehensive Cancer Network (NCCN) guidelines recommend resection as the primary treatment for most localized pancreatic neuroendocrine tumors (NETs). Exceptions include patients with life-limiting comorbidities or high surgical risk. In addition, observation may be appropriate for incidentally discovered tumors < 1 cm, depending on the site.[53]

Prior to surgery, NCCN recommends that symptoms of hormonal excess be treated with octreotide or lanreotide; however, such treatment is contraindicated in patients with insulinoma because of the potential for fatal complications. Specific recommendations vary by tumor subtype.Cholecystectomy is recommended during surgical resection if treatment with octreotide or lanreotide is planned, due to the increased rate of biliary problems associated with long-term use of these agents.[53]

Additional NCCN recommendations for insulinoma treatment include the following[53] :

Medication Summary

Diazoxide is the drug of choice because it inhibits insulin release from the tumor. Adverse effects must be treated with hydrochlorothiazide. In patients not responsive to or intolerant of diazoxide (10%), a somatostatin analog may be indicated to prevent hypoglycemia.

Experience with systemic chemotherapy is limited. The traditional regimen of choice has been streptozocin and doxorubicin. Response rates as high as 69% have been reported but radiologic response is lower and due to uncertainty about efficacy and increased toxicity, this regimen is not accepted as a standard first-line therapy.

Diazoxide (Proglycem, Hyperstat)

Clinical Context:  Produces an increase in blood glucose within 1 h by inhibition of insulin release from the insulinoma.

Class Summary

Inhibit insulin release from the tumor.

Hydrochlorothiazide (Microzide, HydroDIURIL, Esidrix)

Clinical Context:  Inhibits reabsorption of sodium in distal tubules, causing increased excretion of sodium and water as well as potassium and hydrogen ions.

Class Summary

Used to counteract edema and hyperkalemia secondary to diazoxide and to potentiate its hyperglycemic effect.

Lanreotide (Somatuline Depot)

Clinical Context:  Long-acting analog of somatostatin, administered q4wk.

Octreotide acetate (Sandostatin)

Clinical Context:  Acts similarly to the natural hormone somatostatin and can suppress secretion of gastroenteropancreatic peptides, including insulin.

Class Summary

May control symptoms by suppressing secretion of gastroenteropancreatic peptides including insulin. High-dose treatment also may lead to additional antiproliferative effects. However, long-term application of somatostatin may down-regulate receptor expression levels, resulting in decreased efficiency despite increasing doses. Both short- and long-acting depot preparations are available.

Lutetium Lu 177-dota-tate (Lutathera)

Clinical Context:  Binds to somatostatin receptors with highest affinity for subtype 2 receptors (SSRT2). Upon binding to somatostatin receptor-expressing cells, including malignant somatostatin receptor-positive tumors, the compound is internalized. Beta emission from Lu 177 induces cellular damage by forming free radicals in somatostatin receptor-positive cells and in neighboring cells. It is indicated for somatostatin receptor-positive gastroenteropancreatic neuroendocrine tumors (GEP-NETs), including foregut, midgut, and hindgut neuroendocrine tumors in adults.

Class Summary

Lutetium Lu 177-dota-tate (Lu 177), the first peptide receptor radionuclide therapy (PRRT), was approved by the FDA in January 2018. Approval was based on the NETTER-1 clinical trial. The trial was a single-institution, single-arm, open-label trial conducted by Erasmus Medical Center in Rotterdam, Netherlands in more than 1,200 patients with somatostatin receptor positive tumors. Results showed a 79% reduction in risk of disease progression or death in the Lu 177 arm compared with octreotide LAR 60 mg arm (95% CI: 0.13-0.32; p<0.0001). Median PFS was not reached in the Lu 177 arm compared with 8.5 months for octreotide LAR. An interim overall survival analysis determined that Lu 177 treatment lead to a 48% reduction in the estimated risk of death (hazard ratio 0.52, 95% CI: 0.32-0.84) compared with octreotide LAR.[40]

Streptozocin (Zanosar)

Clinical Context:  Has high affinity for neuroendocrine cells, inhibits cell proliferation, and is cytolytic. Interferes with normal function of DNA by alkylation and protein modification.

Temozolomide (Temodar)

Clinical Context:  Orally active alkylating agent; may be considered in the treatment of metastatic pancreatic neuroendocrine tumors. 

Class Summary

These agents inhibit cell growth and proliferation.

Everolimus (Afinitor)

Clinical Context:  Everolimus is indicated for progressive neuroendocrine tumors (PNET) located in the pancreas that are not surgically resectable or are metastatic. It is also indicated for well-differentiated, nonfunctional neuroendocrine tumors (NET) of gastrointestinal (GI) or lung.

Class Summary

Mammalian target of rapamycin (mTOr), a serine-threonine kinase is dysregulated in several human cancers. Inhibitors of mTOR reduces cell proliferation, angiogenesis, and glucose uptake.

Further Inpatient Care

After insulinoma resection, hyperglycemia may persist for 48-72 hours because of chronic down-regulation of insulin-receptors by the previously high circulating insulin levels secreted by the tumor and the suppression of normal pancreatic B cells. Small subcutaneous doses of insulin every 3-6 hours may be necessary if plasma glucose level exceeds 300 mg/dL (16.7 mmol/L).

Patients with major pancreatic resections may develop diabetes mellitus.

Streptozocin chemotherapy appears to be toxic to cells that produce insulin. For insulinomas, some cases of sustained improvement in hypoglycemic attacks have been reported, particularly when streptozotocin has been used.

Short-acting somatostatin analogues may be tried to control insulin release. In patients with unresectable metastatic disease to the liver, when systemic chemotherapy was unsuccessful, embolization of the hepatic artery and intraarterial chemotherapy may be indicated to control symptoms and hormone release, to inhibit tumor growth, and to improve survival.[58]

New therapy is currently under investigation. OctreoTher consists of a somatostatin peptide analogue, labeled with a beta-emitter (yttrium-90). OctreoTher binds to somatostatin receptor 2 and 3, has a mean path length of 5 mm, and a physical half-life of 64.1 hours. By targeting somatostatin-receptor–positive tumors (imaged by scan), it may deliver a local tumoricidal dose of radiation.[59]  

Inpatient & Outpatient Medications

Continue diazoxide and hydrochlorothiazide in patients who are not fit for surgery or when tumor resection was unsuccessful.

Transfer

In advanced metastatic disease, the indications for chemotherapy or other interventional treatments must be emphasized in a multidisciplinary way and discussed with surgeons, specialists in chemoembolization, gastroenterologists, endocrinologists, and medical oncologists.

Complications

See the list below:

Prognosis

Approximately 90-95% of insulinomas are benign. Long-term cure with complete resolution of preoperative symptoms is expected after complete resection.

Recurrence of benign insulinomas was observed in 5.4% of patients in a series of 120 patients over a period of 4-17 years. The same diagnostic and therapeutic approach was recommended, including surgical exploration and tumor resection.

Patients may develop nonfunctioning metastatic disease to the liver up to 14 years after insulinoma resection.[60] Note that some insulinomas are indolent (depending on the tumor biology), resulting in prolonged survival.

What are insulinomas?How are insulinomas clinically characterized?What are symptoms of hypoglycemia in patients with insulinoma?What causes hypoglycemia in patients with insulinoma?What is the hallmark of an insulinoma?How is a biochemical diagnosis of insulinoma established?Which imaging modalities are used in the diagnosis of insulinomas?Which medications are used in the treatment of insulinoma?What are the surgical options for treatment of insulinoma?What are insulinomas?What is the pathophysiology of insulinoma?What are patterns of insulin secretion by insulinomas?What is the incidence of insulinoma in the US?What is the global incidence of insulinoma?What is the mortality and morbidity associated to insulinoma?What are the racial predilections in the prevalence of insulinoma?What are the sexual predilections in the prevalence of insulinoma?Which age groups have the highest incidence of insulinoma?Which clinical history findings are characteristic of insulinoma?What causes insulinoma?How is insulinoma differentiated from other causes of hypoglycemia?What is the classic hallmark of insulinomas?What are the diagnostic criteria for insulinoma?What is the role of lab testing in the diagnosis of insulinoma?How is a biochemical diagnosis of insulinoma made?How is multiple endocrine neoplasia type 1 (MEN 1) assessed during the evaluation for insulinoma?What is the role of imaging studies in the diagnosis of insulinoma?What is the role of CT scanning and MRI in the diagnosis of insulinoma?What is the role of arteriography in the diagnosis of insulinoma?What is the role of nuclear medicine in the diagnosis of insulinoma?What is the role of ultrasonography in the diagnosis of insulinoma?What is the role of portal venous sampling in the evaluation of insulinoma?What is the role of anti-insulin labeled with iodine-131 in the evaluation of insulinoma?What is the role of biopsy in the evaluation of insulinoma?Which histologic findings are characteristic of insulinoma?How is insulinoma staged?What are the medical treatment options for insulinoma?What are the medical treatment options for metastatic insulinoma?What is the role of mammalian target of rapamycin (mTOR) inhibitors in the treatment of insulinoma?What is the role of ablation in the treatment of insulinoma?What is the treatment of choice for insulinoma?What is included in preoperative management of insulinoma?What are the surgical options for the treatment of insulinoma?What is the role of surgery in the treatment of metastatic insulinoma?Which specialist consultations are beneficial for patients with insulinoma?Which diet and activity modifications are used in the treatment of insulinoma?Which organizations have issued clinical treatment guidelines for insulinoma?What are grading schemes for insulinoma?What are the NCCN guidelines for grading and staging insulinoma?What the UK and Ireland Neuroendocrine Tumour Society (UKI NETS) guidelines for grading and staging of insulinoma?What are the NCCN treatment guidelines for locoregional insulinoma?What is the role of diazoxide in the treatment of insulinoma?Which medications in the drug class Antineoplastic, mTOr Kinase Inhibitor are used in the treatment of Insulinoma?Which medications in the drug class Antineoplastic Agents are used in the treatment of Insulinoma?Which medications in the drug class Radiopharmaceuticals are used in the treatment of Insulinoma?Which medications in the drug class Somatostatin Analogs are used in the treatment of Insulinoma?Which medications in the drug class Diuretics are used in the treatment of Insulinoma?Which medications in the drug class Hyperglycemic agents are used in the treatment of Insulinoma?What is included in inpatient care for insulinoma?What are the indications for discontinuation of diazoxide and hydrochlorothiazide in the treatment of insulinoma?Which specialists should be included in a multidisciplinary treatment team for the management of insulinoma?What are complications of insulinoma?What is the prognosis of insulinoma?

Author

Zonera Ashraf Ali, MBBS, Consulting Staff, Main Line Oncology Hematology Associates, Lankenau Cancer Center

Disclosure: Nothing to disclose.

Specialty Editors

Francisco Talavera, PharmD, PhD, Adjunct Assistant Professor, University of Nebraska Medical Center College of Pharmacy; Editor-in-Chief, Medscape Drug Reference

Disclosure: Received salary from Medscape for employment. for: Medscape.

Chief Editor

Neetu Radhakrishnan, MD, Associate Professor (Adjunct) of Medicine, Division of Hematology/Oncology, University of Cincinnati Medical Center; Hematology/Oncology Medical Director, West Chester Outpatient Clinics

Disclosure: Nothing to disclose.

Additional Contributors

Pradyumna D Phatak, MBBS, MD, Chair, Division of Hematology and Medical Oncology, Rochester General Hospital; Clinical Professor of Oncology, Roswell Park Cancer Institute

Disclosure: Received honoraria from Novartis for speaking and teaching.

Acknowledgements

Klaus Radebold, MD, PhD Former Research Associate, Department of Surgery, Yale University School of Medicine

Disclosure: Nothing to disclose.

References

  1. Vinik A, Feliberti E, Perry RR, De Groot LJ, Chrousos G, Dungan K, et al. Insulinomas. July 3, 2017. [View Abstract]
  2. Kirkeby H, Vilmann P, Burcharth F. Insulinoma diagnosed by endoscopic ultrasonography-guided biopsy. J Laparoendosc Adv Surg Tech A. 1999 Jun. 9(3):295-8. [View Abstract]
  3. Proye C, Malvaux P, Pattou F, et al. Noninvasive imaging of insulinomas and gastrinomas with endoscopic ultrasonography and somatostatin receptor scintigraphy. Surgery. 1998 Dec. 124(6):1134-43; discussion 1143-4. [View Abstract]
  4. McLean A. Endoscopic ultrasound in the detection of pancreatic islet cell tumours. Cancer Imaging. 2004 Mar 29. 4(2):84-91. [View Abstract]
  5. Zhu L, Xue H, Sun Z, Li P, Qian T, Xing X, et al. Prospective comparison of biphasic contrast-enhanced CT, volume perfusion CT, and 3 Tesla MRI with diffusion-weighted imaging for insulinoma detection. J Magn Reson Imaging. 2017 Apr 17. [View Abstract]
  6. Nockel P, Babic B, Millo C, Herscovitch P, Patel D, Nilubol N, et al. Localization of Insulinoma Using 68Ga-DOTATATE PET/CT Scan. J Clin Endocrinol Metab. 2017 Jan 1. 102 (1):195-199. [View Abstract]
  7. Fernandez-Cruz L, Blanco L, Cosa R, Rendon H. Is laparoscopic resection adequate in patients with neuroendocrine pancreatic tumors?. World J Surg. 2008 May. 32(5):904-17. [View Abstract]
  8. Phan GQ, Yeo CJ, Hruban RH, et al. Surgical experience with pancreatic and peripancreatic neuroendocrine tumors: review of 125 patients. J Gastrointest Surg. 1998 Sep-Oct. 2(5):472-82. [View Abstract]
  9. Mathur A, Gorden P, Libutti SK. Insulinoma. Surg Clin North Am. 2009 Oct. 89(5):1105-21. [View Abstract]
  10. Henquin JC, Nenquin M, Guiot Y, Rahier J, Sempoux C. Human Insulinomas Show Distinct Patterns of Insulin Secretion In Vitro. Diabetes. 2015 Oct. 64 (10):3543-53. [View Abstract]
  11. Zhan HX, Cong L, Zhao YP, Zhang TP, Chen G, Zhou L, et al. Activated mTOR/P70S6K signaling pathway is involved in insulinoma tumorigenesis. J Surg Oncol. 2012 Dec. 106(8):972-80. [View Abstract]
  12. Dadan J, Wojskowicz P, Wojskowicz A. Neuroendocrine tumors of the pancreas. Wiad Lek. 2008. 61(1-3):43-7. [View Abstract]
  13. Larijani B, Aghakhani S, Lor SS, Farzaneh Z, Pajouhi M, Bastanhagh MH. Insulinoma in Iran: a 20-year review. Ann Saudi Med. 2005 Nov-Dec. 25(6):477-80. [View Abstract]
  14. Shin JJ, Gorden P, Libutti SK. Insulinoma: pathophysiology, localization and management. Future Oncol. 2010 Feb. 6 (2):229-37. [View Abstract]
  15. Kuzin NM, Egorov AV, Kondrashin SA, Lotov AN, Kuznetzov NS, Majorova JB. Preoperative and intraoperative topographic diagnosis of insulinomas. World J Surg. 1998 Jun. 22 (6):593-7; discussion 597-8. [View Abstract]
  16. Dizon AM, Kowalyk S, Hoogwerf BJ. Neuroglycopenic and other symptoms in patients with insulinomas. Am J Med. 1999 Mar. 106(3):307-10. [View Abstract]
  17. Schmitt J, Boullu-Sanchis S, Moreau F, Drui S, Louis B, Chabrier G, et al. Association of malignant insulinoma and type 2 diabetes mellitus: a case report. Ann Endocrinol (Paris). 2008 Feb. 69(1):69-72. [View Abstract]
  18. Ruby RJ, Armato JP, Pyke C, Peters AL. GLP-1 provoked severe hypoglycemia in an individual with type 2 diabetes and a benign insulinoma. Diabetes Care. 2014 Aug. 37 (8):e177-8. [View Abstract]
  19. Hrascan R, Pecina-Slaus N, Martic TN, Colic JF, Gall-Troselj K, Pavelic K. Analysis of selected genes in neuroendocrine tumours: insulinomas and phaeochromocytomas. J Neuroendocrinol. 2008 Aug. 20(8):1015-22. [View Abstract]
  20. Bhatti TR, Ganapathy K, Huppmann AR, Conlin L, Boodhansingh KE, MacMullen C, et al. Histologic and Molecular Profile of Pediatric Insulinomas: Evidence of a Paternal Parent-of-Origin Effect. J Clin Endocrinol Metab. 2016 Jan 12. 161 (3):jc20152914. [View Abstract]
  21. Waickus CM, de Bustros A, Shakil A. Recognizing factitious hypoglycemia in the family practice setting. J Am Board Fam Pract. 1999 Mar-Apr. 12(2):133-6. [View Abstract]
  22. Redmon JB, Nuttall FQ. Autoimmune hypoglycemia. Endocrinol Metab Clin North Am. 1999 Sep. 28(3):603-18, vii. [View Abstract]
  23. Eriguchi N, Aoyagi S, Hara M, et al. Nesidioblastosis with hyperinsulinemic hypoglycemia in adults: report of two cases. Surg Today. 1999. 29(4):361-3. [View Abstract]
  24. Starke A, Saddig C, Kirch B, Tschahargane C, Goretzki P. Islet hyperplasia in adults: challenge to preoperatively diagnose non-insulinoma pancreatogenic hypoglycemia syndrome. World J Surg. 2006 May. 30(5):670-9. [View Abstract]
  25. van Bon AC, Benhadi N, Endert E, Fliers E, Wiersinga WM. Evaluation of endocrine tests. D: the prolonged fasting test for insulinoma. Neth J Med. 2009 Jul-Aug. 67(7):274-8. [View Abstract]
  26. Guettier JM, Lungu A, Goodling A, Cochran C, Gorden P. The Role of Proinsulin and Insulin in the Diagnosis of Insulinoma: A Critical Evaluation of the Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab. 2013 Sep 30. [View Abstract]
  27. Boukhman MP, Karam JM, Shaver J, et al. Localization of insulinomas. Arch Surg. 1999 Aug. 134(8):818-22; discussion 822-3. [View Abstract]
  28. Hashimoto LA, Walsh RM. Preoperative localization of insulinomas is not necessary. J Am Coll Surg. 1999 Oct. 189(4):368-73. [View Abstract]
  29. Liu Y, Song Q, Jin HT, Lin XZ, Chen KM. The value of multidetector-row CT in the preoperative detection of pancreatic insulinomas. Radiol Med. 2009 Sep 30. [View Abstract]
  30. Zhu L, Xue H, Sun H, Wang X, Wu W, Jin Z, et al. Insulinoma Detection With MDCT: Is There a Role for Whole-Pancreas Perfusion?. AJR Am J Roentgenol. 2017 Feb. 208 (2):306-314. [View Abstract]
  31. Hubalewska-Dydejczyk A, Sowa-Staszczak A, Tomaszuk M, Stefańska A. GLP-1 and exendin-4 for imaging endocrine pancreas. A review. Labelled glucagon-like peptide-1 analogues: past, present and future. Q J Nucl Med Mol Imaging. 2015 Jun. 59 (2):152-60. [View Abstract]
  32. Sowa-Staszczak A, Trofimiuk-Müldner M, Stefańska A, Tomaszuk M, Buziak-Bereza M, Gilis-Januszewska A, et al. 99mTc Labeled Glucagon-Like Peptide-1-Analogue (99mTc-GLP1) Scintigraphy in the Management of Patients with Occult Insulinoma. PLoS One. 2016. 11 (8):e0160714. [View Abstract]
  33. Christ E, Wild D, Forrer F, Brändle M, Sahli R, Clerici T, et al. Glucagon-Like Peptide-1 Receptor Imaging for Localization of Insulinomas. J Clin Endocrinol Metab. 2009 Oct 9. [View Abstract]
  34. Luo Y, Pan Q, Shao Y, Yu M, Wu W, Xue H, et al. Glucagon-like Peptide-1 Receptor PET/CT with 68Ga-NOTA-exendin-4 for Detecting Localized Insulinoma: a Prospective Cohort Study. J Nucl Med. 2016 Jan 21. [View Abstract]
  35. Wild D, Christ E, Caplin ME, et al. Glucagon-like peptide-1 versus somatostatin receptor targeting reveals 2 distinct forms of malignant insulinomas. J Nucl Med. 2011 Jul. 52(7):1073-8. [View Abstract]
  36. Arnold R, Simon B, Wied M. Treatment of neuroendocrine GEP tumours with somatostatin analogues: a review. Digestion. 2000. 62 Suppl 1:84-91. [View Abstract]
  37. Tirosh A, Stemmer SM, Solomonov E, Elnekave E, Saeger W, Ravkin Y, et al. Pasireotide for malignant insulinoma. Hormones (Athens). 2016 Apr. 15 (2):271-6. [View Abstract]
  38. Keymeulen B, Bossuyt A, Peeters TL, Somers G. 111In-octreotide scintigraphy: a tool to select patients with endocrine pancreatic tumors for octreotide treatment?. Ann Nucl Med. 1995 Aug. 9(3):149-52. [View Abstract]
  39. FDA approves new treatment for certain digestive tract cancers. U.S. Food & Drug Administration. Available at https://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm594043.htm. January 26, 2018; Accessed: January 30, 2018.
  40. Strosberg J, El-Haddad G, Wolin E, Hendifar A, Yao J, Chasen B, et al. Phase 3 Trial of 177Lu-Dotatate for Midgut Neuroendocrine Tumors. N Engl J Med. 2017 Jan 12. 376 (2):125-135. [View Abstract]
  41. Bourcier ME, Sherrod A, DiGuardo M, Vinik AI. Successful control of intractable hypoglycemia using rapamycin in an 86-year-old man with a pancreatic insulin-secreting islet cell tumor and metastases. J Clin Endocrinol Metab. 2009 Sep. 94(9):3157-62. [View Abstract]
  42. Bernard V, Lombard-Bohas C, Taquet MC, Caroli-Bosc FX, Ruszniewski P, Niccoli-Sire P, et al. Efficacy of Everolimus in Patients with Metastatic Insulinoma and Refractory Hypoglycemia. Eur J Endocrinol. 2013 Feb 7. [View Abstract]
  43. Limmer S, Huppert PE, Juette V, Lenhart A, Welte M, Wietholtz H. Radiofrequency ablation of solitary pancreatic insulinoma in a patient with episodes of severe hypoglycemia. Eur J Gastroenterol Hepatol. 2009 Sep. 21(9):1097-101. [View Abstract]
  44. Mele C, Brunani A, Damascelli B, Tichà V, Castello L, Aimaretti G, et al. Non-surgical ablative therapies for inoperable benign insulinoma. J Endocrinol Invest. 2017 Jul 28. [View Abstract]
  45. Dąbkowski K, Gajewska P, Walter K, Londzin-Olesik M, Białek A, Andrysiak-Mammos E, et al. Successful EUS-guided ethanol ablation of insulinoma, four-year follow-up. Case report and literature review. Endokrynol Pol. 2017. 68 (4):472-479. [View Abstract]
  46. Lo CY, Lam KY, Fan ST. Surgical strategy for insulinomas in multiple endocrine neoplasia type I. Am J Surg. 1998 Apr. 175(4):305-7. [View Abstract]
  47. Fernandez Ranvier GG, Shouhed D, Inabnet WB 3rd. Minimally Invasive Techniques for Resection of Pancreatic Neuroendocrine Tumors. Surg Oncol Clin N Am. 2016 Jan. 25 (1):195-215. [View Abstract]
  48. Dexter SP, Martin IG, Leindler L, et al. Laparoscopic enucleation of a solitary pancreatic insulinoma. Surg Endosc. 1999 Apr. 13(4):406-8. [View Abstract]
  49. Grant CS. Surgical aspects of hyperinsulinemic hypoglycemia. Endocrinol Metab Clin North Am. 1999 Sep. 28(3):533-54. [View Abstract]
  50. Bosman FT, Carneiro F, Hruban RH, Theise ND. World Health Organization Classification of Tumours of the Digestive System. Lyons, France: IARC; 2010.
  51. Rindi G, Klöppel G, Alhman H, et al., European Neuroendocrine Tumor Society (ENETS). TNM staging of foregut (neuro)endocrine tumors: a consensus proposal including a grading system. Virchows Arch. 2006 Oct. 449 (4):395-401. [View Abstract]
  52. Rindi G, Klöppel G, Couvelard A, Komminoth P, Körner M, Lopes JM, et al. TNM staging of midgut and hindgut (neuro) endocrine tumors: a consensus proposal including a grading system. Virchows Arch. 2007 Oct. 451 (4):757-62. [View Abstract]
  53. [Guideline] National Comprehensive Cancer Network. NCCN Clinical Practice Guidelines in Oncology: Neuroendocrine Tumors. NCCN. Available at http://www.nccn.org/professionals/physician_gls/pdf/neuroendocrine.pdf. Version 3.2017 — June 13, 2017; Accessed: October 3, 2017.
  54. Edge SB, Byrd DR, Compton CC, et al. AJCC Cancer Staging Manual. 7th Ed. New York: Springer; 2010.
  55. [Guideline] Öberg K, Knigge U, Kwekkeboom D, Perren A, ESMO Guidelines Working Group. Neuroendocrine gastro-entero-pancreatic tumors: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2012 Oct. 23 Suppl 7:vii124-30. [View Abstract]
  56. [Guideline] Ramage JK, Ahmed A, Ardill J, et al, UK and Ireland Neuroendocrine Tumour Society. Guidelines for the management of gastroenteropancreatic neuroendocrine (including carcinoid) tumours (NETs). Gut. 2012 Jan. 61 (1):6-32. [View Abstract]
  57. [Guideline] Kunz PL, Reidy-Lagunes D, Anthony LB, Bertino EM, Brendtro K, Chan JA, et al. Consensus guidelines for the management and treatment of neuroendocrine tumors. Pancreas. 2013 May. 42 (4):557-77. [View Abstract]
  58. Moscetti L, Saltarelli R, Giuliani R, et al. Intra-arterial liver chemotherapy and hormone therapy in malignant insulinoma: case report and review of the literature. Tumori. 2000 Nov-Dec. 86(6):475-9. [View Abstract]
  59. Smith MC, Liu J, Chen T, et al. OctreoTher: ongoing early clinical development of a somatostatin- receptor-targeted radionuclide antineoplastic therapy. Digestion. 2000. 62 Suppl 1:69-72. [View Abstract]
  60. Gonzalez-Gonzalez A, Recio-Cordova JM. Liver metastases 9 years after removal of a malignant insulinoma which was initially considered benign. JOP. 2006. 7(2):226-9.
  61. Wiesli P, Uthoff H, Perren A, et al. Are biochemical markers of neuroendocrine tumors coreleased with insulin following local calcium stimulation in patients with insulinomas?. Pancreas. 2011 Oct. 40(7):995-9. [View Abstract]
  62. Begu-Le Corroller A, Valero R, Moutardier V, Henry JF, Le Treut YP, Gueydan M. Aggressive multimodal therapy of sporadic malignant insulinoma can improve survival: A retrospective 35-year study of 12 patients. Diabetes Metab. 2008 Jun 13. [View Abstract]
  63. Yao JC, Evans DB. Pancreatic endocrine tumors. De Vita V, Lawrence T, Rosenberg S. De Vita, Hellman, and Rosenberg's Cancer. Principles and Practice of Oncology. 10th ed. Philadelphia, PA: Wolters Kluwer Health; 2015. 1205-17.
  64. Faggiano A, Mansueto G, Ferolla P, Milone F, del Basso de Caro ML, Lombardi G, et al. Diagnostic and prognostic implications of the World Health Organization classification of neuroendocrine tumors. J Endocrinol Invest. 2008 Mar. 31(3):216-23. [View Abstract]
  65. Crippa S, Zerbi A, Boninsegna L, Capitanio V, Partelli S, Balzano G, et al. Surgical management of insulinomas: short- and long-term outcomes after enucleations and pancreatic resections. Arch Surg. 2012 Mar. 147 (3):261-6. [View Abstract]

CT scan image with oral and intravenous contrast in a patient with biochemical evidence of insulinoma. The 3-cm contrast-enhancing neoplasm (arrow) is seen in the tail of the pancreas (P) posterior to the stomach (S) (Yeo, 1993).

CT scan image with oral and intravenous contrast in a patient with biochemical evidence of insulinoma. The 3-cm contrast-enhancing neoplasm (arrow) is seen in the tail of the pancreas (P) posterior to the stomach (S) (Yeo, 1993).

Endoscopic ultrasonography in a patient with an insulinoma. The hypoechoic neoplasm (arrows) is seen in the body of the pancreas anterior to the splenic vein (SV) (Rosch, 1992).

CT scan image with oral and intravenous contrast in a patient with biochemical evidence of insulinoma. The 3-cm contrast-enhancing neoplasm (arrow) is seen in the tail of the pancreas (P) posterior to the stomach (S) (Yeo, 1993).

Endoscopic ultrasonography in a patient with an insulinoma. The hypoechoic neoplasm (arrows) is seen in the body of the pancreas anterior to the splenic vein (SV) (Rosch, 1992).