VIPomas

Back

Background

VIPomas are neuroendocrine tumors that secrete vasoactive intestinal peptide (VIP) autonomously.[1, 2, 3] They originate in amine precursor uptake and decarboxylation (APUD) cells of the gastroenteropancreatic endocrine system and in adrenal or extra-adrenal neurogenic sites. Neural crest cells are precursors of APUDoma and neurogenic cells.

The symptoms of VIPoma were initially described in 1958, when Verner and Morrison described a syndrome of watery diarrhea, hypokalemia, and achlorhydria (WDHA).[4] In 1970, Said and Nutt extracted the responsible hormone from animal gut[5] ; in 1973, Bloom causally linked this hormone to WDHA syndrome. In 1975, Swift et al was the first to report a child who had watery diarrhea and a ganglioneuroma with secretion of VIP.[6]

VIPomas arise from the pancreas in 90% of cases, but they may also be found in periganglionic tissue or at other sites (eg, colon, bronchus, adrenal glands, and liver), especially in children.[7] They are almost always solitary, with fewer than 5% being multicentric. The tumors are usually greater than 3 cm in diameter at the time of diagnosis and are found primarily in the body and tail of the pancreas.

In children, it is extremely rare for a VIPoma to originate in the pancreas; instead, WDHA syndrome is usually associated with VIP-secreting neurogenic tumors involving the retroperitoneum and mediastinum. Pancreatic non–beta-cell hyperplasia is rare but has been reported in children. Clinical experience is based mainly on case reports (~85 cases before 2005).

Approximately 60-80% of VIPomas are malignant and have metastasized at the time of diagnosis. Metastasis occurs most frequently in the liver but may also occur in the lymph nodes, lungs, or kidneys.[8] Approximately 5% of VIPomas are associated with multiple endocrine neoplasia (MEN) type 1 syndrome. Conversely, 17% of patients with MEN 1 develop VIPomas at some stage of their disease. Approximately 10% of neuroendocrine tumors of the gastrointestinal (GI) tract (except carcinoids) are VIPomas.

Pathophysiology and Etiology

VIP has a molecular weight of 3381, consists of 28 amino acids, and belongs to the secretin-glucagon family. The VIP gene is located on chromosome 6. VIP is normally expressed in the central nervous system (CNS) and in the neurons of the GI, respiratory, and urogenital tracts, where it functions as a neurotransmitter.

VIP regulates the synthesis, secretion, and action of other neuroendocrine hormones; it also regulates cytokines and chemokines. VIP deficiency leads to developmental and behavioral abnormalities (eg, impaired circadian rhythms) in animal models.[9] Overexpression of VIP causes diarrhea and cancer, and overexpression of VIP receptors promotes cancerous growth. In the GI tract, VIP is responsible for relaxation of vascular and nonvascular smooth muscle and secretion of water and electrolytes. It is released in response to distention of the gut by food.

VIP is a potent stimulator of gut cyclic adenosine monophosphate (cAMP) production, which leads to massive secretion of water and electrolytes (mainly potassium). VIP resembles secretin, which stimulates the secretion of alkaline pancreatic juices. In the stomach, VIP inhibits histamine- and pentagastrin-stimulated acid secretion. Like glucagon, VIP stimulates lipolysis and glycogenolysis and has an inotropic effect on the myocardium. It also has anti-inflammatory properties and modulates the immune system.

VIPomas in adults are usually neuroendocrine islet cell tumors of the pancreas that produce high amounts of VIP; other secreted hormones may include secreted gastrin and pancreatic polypeptide. In children and adolescents, VIP is produced mainly by ganglioneuromas, ganglioneuroblastomas, neurofibromas, or other tumors in the adrenal area (the most common location). Only a small fraction of neuroblastomas and ganglioneuroblastomas produce VIP, but VIP production indicates a more favorable prognosis.

Ganglioneuromatosis that affects the entire colon and rectum has been reported in a 7-year-old boy.[10] In contrast to individuals with VIP-secreting pancreatic tumors, patients with neurogenic lesions generally have normal serum levels of pancreatic polypeptide, gastrin, insulin, and somatostatin.

VIPomas can be part of MEN 1 syndrome. This relationship has not been observed with extrapancreatic VIP-secreting tumors of childhood. Somatic point mutations on chromosome 11 of the MEN1 gene have been discovered in sporadic VIPomas and VIPoma cases associated with MEN type 1.

Epidemiology

United States statistics

VIPomas are the third most common neuroendocrine tumor of the pancreas (15%), after insulinomas (50%) and gastrinomas (30%). Annually, 0.05-0.5 new cases per million adults have been reported. No data are available for the incidence of this condition in children.

Age- and sex-related demographics

Peak incidence occurs in the fifth decade of life, but VIPomas may occur in any age group, including young children and elderly persons. In a series of 19 childhood cases, the mean age of onset was 2.5 years.[11] In another series of 10 cases, the mean age of onset was 4 years. The earliest age of onset ever reported is 2 weeks. The male-to-female ratio in children is approximately 1:1, compared with 1:3 in adults.

Prognosis

Approximately 50% of surgical patients with VIPoma are cured after tumor resection. In a large series of 241 adult patients with VIPomas, the 5-year survival rate was 89% among those with pancreatic VIPomas and 68.5% among those with neurogenic VIP-producing tumors.[12] In metastatic disease, the 5-year survival rate was 59.6%. Children with neuroblastomas have a relatively poor overall survival rate (30-40%); children with VIP-secreting ganglioneuroblastomas have a considerably higher survival rate (>90%).

A study by Keutgen et al that evaluated factors affecting survival in malignant-functioning pancreatic neuroendocrine tumors found that the median survival time for VIPomas was 7.9 years. The study also found that primary tumor resection is associated with longer survival in stages I-III as well as stage IV malignant-functioning pancreatic neuroendocrine tumors.[13]

Morbidity from untreated WDHA syndrome is associated with long-standing dehydration and with electrolyte and acid-base disturbances, which may cause chronic renal failure.[14] Death results from renal failure or cardiac arrest caused by volume depletion, hypokalemia, and severe acid-base disturbances.

History

The onset of VIPoma is insidious. The dominant symptom is profuse diarrhea despite fasting; this symptom may persist for years before the diagnosis is established. Diarrhea may be episodic initially, but it becomes continuous as the tumor progresses. The stool is typically odorless and tea-colored, without blood or mucus.

Clinical diagnosis is based on a history of approximately 10 watery stools per day. Fecal losses during fasting are at least 20 mL/kg/day but exceed 50 mL/kg/day in most cases. Fecal osmolality is entirely accounted for by twice the sum of the concentrations of sodium and potassium, indicating the electrolyte loss.

The loss of water, sodium, and chloride may lead to volume depletion, dehydration, and exhaustion among patients who are unable to replace the lost fluid and electrolytes. Weight loss and even renal failure have been reported in some patients. Excretion of large amounts of potassium and bicarbonate in the stool causes hypokalemia and non–anion gap acidosis. Hypokalemia may present as muscle cramps or weakness.

Abdominal discomfort or bloating has been reported. In a 31-case series from China, facial flushing was observed in one third of patients.[15] Other studies have also reported facial flushing, but without specifying its frequency.

One patient in China reportedly suffered from periodic backache and a rash involving the chest, back, and upper limb. These 2 symptoms occurred before or after the diarrhea, worsened over 6 years, and resolved after surgical resection.

Physical Examination

Aside from a mildly extended abdomen, no relevant abnormalities may be apparent on physical evaluation of patients with VIPomas. However, physical examination may also reveal one or more of the following:

Electrolyte abnormalities (see Workup) are commonly noted.

Approach Considerations

Plasma vasoactive intestinal peptide (VIP) levels are determined by radioimmunoassay. In cases of VIPoma, VIP levels are usually 2-10 times the normal range (20-30 pmol/L). VIP levels must be determined when the patient is symptomatic because VIP release from the tumor fluctuates.

Imaging studies focus primarily on the pancreas, where 90% of VIPomas are located. Tumor localization normally is not difficult, because at the time of diagnosis, these tumors are generally larger than 3 cm in their longest dimension. Computed tomography (CT), magnetic resonance imaging (MRI), and somatostatin receptor scintigraphy are imaging modalities that can be used in the diagnosis of VIPoma. Reports have demonstrated successful VIPoma localization with99m Tc sestamibi.[16]

No formal staging criteria for VIPoma have been generally accepted. Metastasis most often occurs to the liver or regional lymph nodes. Rarely, metastasis to skin has been reported.

Laboratory Studies

A diagnosis of VIPoma is made when watery diarrhea, hypokalemia, and achlorhydria are present in the setting of elevated serum VIP concentrations. Stool volumes of less than 700 mL/day virtually exclude the diagnosis; typical stool volumes in the presence of a VIPoma exceed 3 L/day.

A normal plasma VIP level is 20-30 pmol/L or less, as determined by radioimmunoassay. VIP levels in patients with VIPoma often reach 160-250 pmol/L or higher. VIP levels should be drawn after fasting.[17] Because VIP is degraded rapidly, a protease inhibitor, such as aprotinin, is added to the blood sample, which must be kept frozen at −70°C until processed. Because VIP secretion from the tumor may be episodic, serum VIP levels should be collected during bouts of severe diarrhea.

Hypokalemia and non–anion gap acidosis are the main diagnostic features of VIPomas. Hypokalemia may necessitate aggressive potassium replacement.

Hypercalcemia may occur in the absence of multiple endocrine neoplasia (MEN) type 1 syndrome or elevated parathyroid hormone levels. The mechanism of action is not clear but is believed to involve increased bone resorption. The dehydration from severe diarrhea certainly may exacerbate the hypercalcemia.

Hyperglycemia may be caused by the direct glycogenolytic effect of VIP on the liver and by the inhibitory effect of hypokalemia on pancreatic islet cell insulin release.

Hypochlorhydria or achlorhydria is seen in at least 75% of patients with VIPoma because VIP inhibits histamine- and pentagastrin-stimulated gastric acid secretion. This abnormality can be evaluated by measuring gastric pH or basal gastric acid output.

Renal function should be assessed by measuring blood urea nitrogen (BUN) and serum creatinine levels. Other electrolytes, including magnesium, should be checked and replaced. Stool weight with potassium measurements verifies high gastrointestinal (GI) potassium losses.

VIPomas may cosecrete other hormones, including pancreatic polypeptide, calcitonin, and neurotensin.

Computed Tomography

CT is indicated to search for neck, mediastinal, or retroperitoneal masses. No calcifications or bony infiltrations should be found. CT will successfully identify the primary tumor in most cases; it also assists in including or excluding liver metastasis.

In a series of 31 patients from China, Peng et al reported that CT successfully identified all VIPomas in the pancreatic body and tail but only 71% of VIPomas in the pancreatic head.[15]

Magnetic Resonance Imaging

MRI may be used if CT is contraindicated (eg, if the patient is allergic to iodine contrast dyes or is in a state of renal failure). VIPomas are best observed on T1-weighted, fat-suppressed images as low-signal-intensity masses. Liver metastases may demonstrate intensive peripheral ring enhancement on immediate postgadolinium spoiled gradient-echo images.[18]

Somatostatin Receptor Scintigraphy

Somatostatin receptor scintigraphy using radionuclide-labeled octreotide (111 In-pentetreotide (ie, DTPA-D-Phe-1-octreotide),111 In-DOTA-DPhe1 -Tyr3 -octreotide), or lanreotide (111 In -DOTA-lanreotide) may be useful for characterizing an abnormality found on a CT scan or for identifying occult or distant metastatic disease. It may also be used if postoperative changes diminish the clarity of a CT scan. Sensitivity for localization of all pancreatic endocrine tumors has been reported at 80-90%, with 92% sensitivity for tumors larger than 1 cm.

Other previously employed techniques include technetium-99m scintigraphy, 123-iodine-VIP receptor scintigraphy, and single-photon emission CT (SPECT). Investigations have suggested that in the future, the use of SPECT scanning may improve the value of somatostatin receptor scintigraphy for the localization of neuroendocrine tumors, including VIPomas.[19]

Other Studies

18 F-deoxyglucose (FDG)-PET scan has also been used to diagnose neuroendocrine tumors. However, it may not be as sensitive as somatostatin receptor scintigraphy.[20]

Chest radiography may reveal a paravertebral mass. Endoscopic retrograde cholangiopancreatography may demonstrate occlusion of the major pancreatic duct. It may also reveal calcifications in the body of the pancreas. Transabdominal ultrasonography may be used for early screening to exclude liver metastases, which may be present as hepatic calcifications.

Electrocardiography may reveal QRS widening and T-wave flattening if hypokalemia is severe. Colonoscopy may be useful as a means of evaluating for a villous adenoma as an alternative cause of potassium-losing diarrhea.

Histology

VIPomas, like other pancreatic endocrine tumors, are thought to arise from the pluripotent cells in ductal epithelium. Histologic examination usually reveals, as is typical for neuroendocrine tumors, sheets of nested, uniform-appearing cells with round nuclei and a low mitotic rate.

Immunohistochemical staining is positive for chromogranin A and VIP. Under electron microscopy, neurosecretory granules may be seen clustering around Golgi complexes and the endoplasmic reticulum. Classifying a tumor as malignant or benign on the basis of microscopic appearance alone is difficult.

Approach Considerations

Initial treatment of VIPomas is directed toward correcting volume and electrolyte abnormalities. Octreotide acetate controls diarrhea in up to 90% of patients with VIPomas. Glucocorticoids reduce symptoms in 50%.

Systemic chemotherapy may be needed in cases of unresectable or progressive disease. Streptozocin, doxorubicin, fluorouracil, or a combination of these appears to be beneficial, though the number of treated cases has been limited.

Local tumor resection is the treatment of choice. In advanced disease, tumor debulking may relieve symptoms, but it is not effective in all cases. Transarterial chemoembolization (TACE) with chemotherapy-loaded materials may provide palliation in patients with extensive hepatic disease.[21] This treatment may be repeated several times, depending on tumor growth and symptoms. External radiation therapy may be indicated in unresectable tumors. Laparoscopic surgical resection has been successful in at least one adult with hepatic metastasis. Irreversible electroporation (IRE) for the management of unresectable pancreatic VIPoma has been reported.[22]

In patients with VIPoma, parathyroidectomy does not correct hypercalcemia. (VIP and its parathyroid hormone–like action cause hypercalcemia.)

Pharmacologic Therapy

Initial treatment for VIPoma is aimed at correcting any volume, electrolyte, and acid-base abnormalities with intravenous (IV) normal saline, potassium chloride, and, if acidosis is severe, sodium bicarbonate. In many cases, these abnormalities are pronounced enough to necessitate hospital admission.

In more than 90% of patients, somatostatin effectively reduces serum vasoactive intestinal peptide (VIP) levels and promptly controls diarrhea.[15] To circumvent the short serum half-life of somatostatin, the derivative octreotide is used. A long-acting octreotide formulation is available that allows once-monthly intragluteal administration. (Patients may have to make monthly clinic visits to receive the injections.) Diarrhea recurs when treatment is discontinued. It is currently debated whether somatostatin analogues also diminish tumor size.

Unless a surgical cure has been achieved (see Surgical Intervention), octreotide dosing is continued in most patients. Long-term octreotide treatment frequently results in gradually increasing resistance to this therapy. When the highest tolerable dosages of octreotide are unable to control symptoms, interferon alfa may be added to control diarrhea (and, possibly, achieve a modest reduction in tumor size).

Glucocorticoids are less effective, reducing symptoms in approximately 50% of patients. However, they are also less expensive.

In patients with advanced disease, tumors may respond to treatment with streptozocin-based chemotherapy (eg, streptozocin combined with 5-fluorouracil [5-FU]). If conventional chemotherapy and somatostatin are not effective, 5-FU may be combined with interferon alfa. Other chemotherapy agents that may be considered include dacarbazine and doxorubicin HCl.

Radiation Therapy

The use of radiolabeled octreotide to target radiation treatment toward a VIPoma is based on the affinity of octreotide for the somatostatin receptors on the VIPoma cells. In one trial, half of the patients achieved stabilization of previously progressive tumors, with minimal bone marrow toxicity.[23] This therapeutic approach may be applied to advanced neuroendocrine tumors in general.

Occasionally, external radiation therapy may be indicated for locally advanced unresectable tumors; however, experience with this treatment is limited.

Surgical Intervention

Surgical exploration with tumor resection (see the image below) leads to cure in 50% of patients. After appropriate fluid and electrolyte replacement, all operable patients with apparently resectable disease should receive abdominal exploration with careful staging. Intraoperative ultrasonography of the pancreas may aid in locating an otherwise unidentified tumor. For patients without nodal or distant metastasis, complete surgical resection offers the only chance for a cure.



View Image

Patient with a large VIPoma. (A) Arteriogram showing vascularity of a large VIPoma preoperatively. (B) Large mass seen intraoperatively. (C) Gross pat....

Local tumor resection is the treatment of choice.[15, 24, 25, 26] Pancreatoduodenectomy is indicated when the tumor is in the pancreatic head or processus uncinatus. If no tumor is found at surgery, a blind pancreatic tail resection may be performed. A total pancreatectomy no longer is recommended. Serum VIP levels may normalize within an hour after curative tumor resection. Preoperative treatment with a proton pump inhibitor is advisable to prevent rebound gastric acid hypersecretion after surgical tumor removal.

Severe hypotension may develop temporarily during and after tumor removal as a consequence of the vasodilatory effect of the VIP released during manipulation of the tumor. Severe hypertension may develop temporarily after tumor removal. Postoperatively, octreotide therapy will usually be needed indefinitely to control symptoms of VIP hypersecretion from residual tumor.[25]

In most cases, at the time of diagnosis of VIPoma, metastatic disease is already present. For these patients, tumor debulking may reduce clinical symptoms,[15, 26] but for substantial clinical benefit to be achieved, operative planning ought to include resection of more than 90% of tumor volume.

Unresectable liver metastases may be treated with hepatic artery radioembolization or transcatheter chemoembolization with doxorubicin or cisplatin.[27] When embolization is unsuccessful or not feasible for liver metastases, percutaneous or intraoperative radiofrequency tumor ablation may be attempted, though it is not ideal for large metastatic tumors.

All patients being surgically treated for a VIPoma should undergo a cholecystectomy to alleviate concerns of gallstones with somatostatin analogue therapy, in case such therapy is needed in the future. In patients with VIPomas, a parathyroidectomy does not correct hypercalcemia unless the VIPoma is resected.

Orthotopic liver transplantation has been performed in a small number of adult patients with pancreatic endocrine tumors; the 5-year survival rate has been approximately 50%.[28]

Consultations

Consultations with the following specialists may be warranted:

Long-Term Monitoring

Patients with VIPomas need frequent outpatient follow-up to monitor hydration status and serum electrolyte levels. In patients with continuing fluid loss that is not controlled effectively by medical and surgical treatment options, a long-term central venous access device may be implanted; the patient may be trained to replace fluid and electrolytes at home or in an ambulatory setting.

Posttreatment VIP levels may serve as a tumor marker for recurrent disease.

Medication Summary

Somatostatin reduces serum vasoactive intestinal polypeptide (VIP) levels and controls diarrhea in patients with VIPomas. To circumvent the short serum half-life of somatostatin, the derivative octreotide is used. An available long-acting formulation of octreotide allows once-monthly intragluteal administration. Long-term treatment with octreotide often causes resistance to the drug. When maximum tolerable octreotide doses cannot control symptoms, interferon alfa may be added to control diarrhea.

Glucocorticoids are less effective but also less expensive.

Octreotide acetate (Sandostatin, Sandostatin LAR)

Clinical Context:  Octreotide acetate acts similarly to the natural hormone somatostatin and has the ability to suppress the secretion of gastroenteropancreatic peptides, including VIP. Octreotide treatment in patients with VIPomas should start with low dosages, which are then titrated on the basis of patient response. Sandostatin LAR is a long-acting depot dosage formulation intended for intramuscular injection.

Lanreotide (Somatuline)

Clinical Context:  Lanreotide is a long-acting synthetic analogue of somatostatin. It binds to the same receptors as somatostatin but with higher affinity to peripheral receptors. It has a much longer biological half-life than octreotide, and its pharmacological effects are longer than octreotide.

Lanreotide's action is similar to the natural hormone somatostatin, but it is long-acting. It suppresses peptide secretion, including VIP, from gastroenteropancreatic tumors. It controls diarrhea in 80% of patients with unresectable or metastatic tumors. It is available in two formulations: sustained-release Somatuline La for IM injection every 10-14 days, and a depot formulation Somatuline Depot injected SC monthly.

Class Summary

Somatostatin analogues may control diarrheal symptoms in as many as 80% of patients with unresectable or metastatic tumors. High-dose treatment may lead to additional, antiproliferative effects. However, long-term application of somatostatin may downregulate receptor expression levels, resulting in decreased efficiency despite increasing doses. Short-acting and long-acting depot preparations are available.

Prednisone (Rayos)

Clinical Context:  Prednisone is an immunosuppressant used to treat autoimmune disorders. It may decrease inflammation by reversing increased capillary permeability and suppressing polymorphonuclear leukocyte (PMN) activity. It stabilizes lysosomal membranes and also suppresses lymphocytes and antibody production.

Prednisolone (Pediapred, Prelone, Orapred, Millipred, Flo-Pred)

Clinical Context:  Prednisolone may decrease inflammation by reversing increased capillary permeability and suppressing PMN activity. It is a commonly used oral agent.

Class Summary

Glucocorticoids have anti-inflammatory properties and cause profound and varied metabolic effects, modifying the body’s immune response to diverse stimuli. In the treatment of VIPomas, these agents are less expensive than octreotide but also less effective; they reduce symptoms in approximately 50% of patients.

Author

Sai-Ching Jim Yeung, MD, PhD, FACP, Professor of Medicine, Department of Emergency Medicine, Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center

Disclosure: Serve(d) as a director, officer, partner, employee, advisor, consultant or trustee for: Celgene, Inc.<br/>Received research grant from: DepoMed and Bristol-Myer-Squibb.

Coauthor(s)

Daniel S Tung, MD, Fellow in Endocrinology, Department of Internal Medicine, Baylor College of Medicine

Disclosure: Nothing to disclose.

Chief Editor

George T Griffing, MD, Professor Emeritus of Medicine, St Louis University School of Medicine

Disclosure: Nothing to disclose.

Acknowledgements

Arthur B Chausmer, MD, PhD, FACP, FACE, FACN, CNS Professor of Medicine (Endocrinology, Adj), Johns Hopkins School of Medicine; Affiliate Research Professor, Bioinformatics and Computational Biology Program, School of Computational Sciences, George Mason University; Principal, C/A Informatics, LLC

Arthur B Chausmer, MD, PhD, FACP, FACE, FACN, CNS is a member of the following medical societies: American Association of Clinical Endocrinologists, American College of Endocrinology, American College of Nutrition, American College of Physicians, American College of Physicians-American Society of Internal Medicine, American Medical Informatics Association, American Society for Bone and Mineral Research, Endocrine Society, and International Society for Clinical Densitometry

Disclosure: Nothing to disclose.

Alicia Diaz-Thomas, MD, MPH Assistant Professor of Pediatrics, University of Tennessee Health Science Center

Alicia Diaz-Thomas, MD, MPH is a member of the following medical societies: American Academy of Clinical Endocrinology, Tennessee Medical Association, and The Endocrine Society

Disclosure: Nothing to disclose.

Robert J Ferry Jr, MD Le Bonheur Chair of Excellence in Endocrinology, Professor and Chief, Division of Pediatric Endocrinology and Metabolism, Department of Pediatrics, University of Tennessee Health Science Center

Robert J Ferry Jr, MD is a member of the following medical societies: American Academy of Pediatrics, American Diabetes Association, American Medical Association, Endocrine Society, Pediatric Endocrine Society, Society for Pediatric Research, and Texas Pediatric Society

Disclosure: Eli Lilly & Co Grant/research funds Investigator; MacroGenics, Inc Grant/research funds Investigator; Ipsen, SA (formerly Tercica, Inc) Grant/research funds Investigator; NovoNordisk SA Grant/research funds Investigator; Diamyd Grant/research funds Investigator; Bristol-Myers-Squibb Grant/research funds Other; Amylin Other; Pfizer Grant/research funds Other; Takeda Grant/research funds Other

Stephen Kemp, MD, PhD Professor, Department of Pediatrics, Section of Pediatric Endocrinology, University of Arkansas for Medical Sciences College of Medicine, Arkansas Children's Hospital

Stephen Kemp, MD, PhD is a member of the following medical societies: American Academy of Pediatrics, American Association of Clinical Endocrinologists, American Pediatric Society, Phi Beta Kappa, Southern Medical Association, Southern Society for Pediatric Research, and The Endocrine Society

Disclosure: Nothing to disclose.

Lynne Lipton Levitsky, MD Chief, Pediatric Endocrine Unit, Massachusetts General Hospital; Associate Professor of Pediatrics, Harvard Medical School

Lynne Lipton Levitsky, MD is a member of the following medical societies: Alpha Omega Alpha, American Academy of Pediatrics, American Diabetes Association, American Pediatric Society, Pediatric Endocrine Society, Society for Pediatric Research, and The Endocrine Society

Disclosure: Pfizer Grant/research funds P.I.; Tercica Grant/research funds Other; Eli Lily Grant/research funds PI; NovoNordisk Grant/research funds PI; NovoNordisk Consulting fee Consulting; Onyx Heart Valve Consulting fee Consulting

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

Disclosure: Nothing to disclose.

Arlan L Rosenbloom, MD Adjunct Distinguished Service Professor Emeritus of Pediatrics, University of Florida College of Medicine; Fellow of the American Academy of Pediatrics; Fellow of the American College of Epidemiology

Arlan L Rosenbloom, MD is a member of the following medical societies: American Academy of Pediatrics, American College of Epidemiology, American Pediatric Society, Florida Pediatric Society, Pediatric Endocrine Society, Society for Pediatric Research, and The Endocrine Society

Disclosure: Nothing to disclose.

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

Disclosure: Medscape Salary Employment

Mary L Windle, PharmD Adjunct Associate Professor, University of Nebraska Medical Center College of Pharmacy; Editor-in-Chief, Medscape Drug Reference

Disclosure: Nothing to disclose.

Frederick H Ziel, MD Associate Professor of Medicine, University of California, Los Angeles, David Geffen School of Medicine; Physician-In-Charge, Endocrinology/Diabetes Center, Director of Medical Education, Kaiser Permanente Woodland Hills; Chair of Endocrinology, Co-Chair of Diabetes Complete Care Program, Southern California Permanente Medical Group

Frederick H Ziel, MD is a member of the following medical societies: American Association of Clinical Endocrinologists, American College of Endocrinology, American College of Physicians, American College of Physicians-American Society of Internal Medicine, American Diabetes Association, American Federation for Medical Research, American Medical Association, American Society for Bone and Mineral Research, California Medical Association, Endocrine Society, and InternationalSociety for Clinical Densitometry

Disclosure: Nothing to disclose.

References

  1. Batcher E, Madaj P, Gianoukakis AG. Pancreatic neuroendocrine tumors. Endocr Res. 2011. 36(1):35-43. [View Abstract]
  2. Alvite-Canosa M, Alonso-Fernández L, Seoane-López M, Pérz-Grobas J, Berdeal-Díaz M, Carral-Freire M, et al. Benign pancreatic vipoma. Rev Esp Enferm Dig. 2011 Mar. 103(4):224-5. [View Abstract]
  3. Strosberg JR, Cheema A, Weber J, Han G, Coppola D, Kvols LK. Prognostic validity of a novel American Joint Committee on Cancer Staging Classification for pancreatic neuroendocrine tumors. J Clin Oncol. 2011 Aug 1. 29(22):3044-9. [View Abstract]
  4. Verner JV, Morrison AB. Islet cell tumor and a syndrome of refractory watery diarrhea and hypokalemia. Am J Med. 1958 Sep. 25(3):374-80. [View Abstract]
  5. Said SI, Mutt V. Polypeptide with broad biological activity: isolation from small intestine. Science. 1970 Sep 18. 169(951):1217-8. [View Abstract]
  6. Swift PG, Bloom SR, Harris F. Watery diarrhoea and ganglioneuroma with secretion of vasoactive intestinal peptide. Arch Dis Child. 1975 Nov. 50(11):896-9. [View Abstract]
  7. Doherty GM. Rare endocrine tumours of the GI tract. Best Pract Res Clin Gastroenterol. 2005 Oct. 19(5):807-17. [View Abstract]
  8. Ayub A, Zafar M, Abdulkareem A, et al. Primary hepatic vipoma. Am J Gastroenterol. 1993 Jun. 88(6):958-61. [View Abstract]
  9. Aton SJ, Colwell CS, Harmar AJ, et al. Vasoactive intestinal polypeptide mediates circadian rhythmicity and synchrony in mammalian clock neurons. Nat Neurosci. 2005 Apr. 8(4):476-83. [View Abstract]
  10. Rescorla FJ, Vane DW, Fitzgerald JF, West KW, Grosfeld JL. Vasoactive intestinal polypeptide-secreting ganglioneuromatosis affecting the entire colon and rectum. J Pediatr Surg. 1988 Jul. 23(7):635-7. [View Abstract]
  11. Quak SH, Prabhakaran K, Kwok R, O'Reilly AP. Vasoactive intestinal peptide secreting tumours in children: a case report with literature review. Aust Paediatr J. 1988 Feb. 24(1):55-8. [View Abstract]
  12. Müller S, Kupka S, Königsrainer I, Northoff H, Sotlar K, Bock T. MSH2 and CXCR4 involvement in malignant VIPoma. World J Surg Oncol. 2012. 10:264. [View Abstract]
  13. Keutgen XM, Nilubol N, Kebebew E. Malignant-functioning neuroendocrine tumors of the pancreas: A survival analysis. Surgery. 2016 May. 159 (5):1382-9. [View Abstract]
  14. Grier JF. WDHA (watery diarrhea, hypokalemia, achlorhydria) syndrome: clinical features, diagnosis, and treatment. South Med J. 1995 Jan. 88(1):22-4. [View Abstract]
  15. Peng SY, Li JT, Liu YB, Fang HQ, Wu YL, Peng CH, et al. Diagnosis and treatment of VIPoma in China: (case report and 31 cases review) diagnosis and treatment of VIPoma. Pancreas. 2004 Jan. 28(1):93-7. [View Abstract]
  16. Cesani F, Ernst R, Walser E, Villanueva-Meyer J. Tc-99m sestamibi imaging of a pancreatic VIPoma and parathyroid adenoma in a patient with multiple type I endocrine neoplasia. Clin Nucl Med. 1994 Jun. 19(6):532-4. [View Abstract]
  17. de Herder WW. Biochemistry of neuroendocrine tumours. Best Pract Res Clin Endocrinol Metab. 2007 Mar. 21(1):33-41. [View Abstract]
  18. Sofka CM, Semelka RC, Marcos HB, Woosley JT. MR imaging of metastatic pancreatic VIPoma. Magn Reson Imaging. 1997. 15(10):1205-8. [View Abstract]
  19. Schillaci O, Corleto VD, Annibale B, Scopinaro F, Delle Fave G. Single photon emission computed tomography procedure improves accuracy of somatostatin receptor scintigraphy in gastro-entero pancreatic tumours. Ital J Gastroenterol Hepatol. 1999 Oct. 31 Suppl 2:S186-9. [View Abstract]
  20. Virgolini I, Patri P, Novotny C, et al. Comparative somatostatin receptor scintigraphy using in-111-DOTA-lanreotide and in-111-DOTA-Tyr3-octreotide versus F-18-FDG-PET for evaluation of somatostatin receptor-mediated radionuclide therapy. Ann Oncol. 2001. 12 Suppl 2:S41-5. [View Abstract]
  21. Dréanic J, Lepère C, El Hajjam M, Gouya H, Rougier P, Coriat R. Emergency therapy for liver metastases from advanced VIPoma: surgery or transarterial chemoembolization?. Ther Adv Med Oncol. 2016 Sep. 8 (5):383-7. [View Abstract]
  22. Niu L, Li J, Zeng J, Fang G, Zhou L, Xu K, et al. Percutaneous Irreversible Electroporation for Pancreatic VIPoma: A Case Report. Pancreas. 2017 Jan. 46 (1):135-137. [View Abstract]
  23. Khasraw M, Gill A, Harrington T, Pavlakis N, Modlin I. Management of advanced neuroendocrine tumors with hepatic metastasis. J Clin Gastroenterol. 2009 Oct. 43(9):838-47. [View Abstract]
  24. Ruiz-Tovar J, Priego P, Martínez-Molina E, Morales V, Sanjuanbenito A, Lobo E. Pancreatic neuroendocrine tumours. Clin Transl Oncol. 2008 Aug. 10(8):493-7. [View Abstract]
  25. Ghaferi AA, Chojnacki KA, Long WD, Cameron JL, Yeo CJ. Pancreatic VIPomas: subject review and one institutional experience. J Gastrointest Surg. 2008 Feb. 12(2):382-93. [View Abstract]
  26. Akerström G, Hellman P. Surgery on neuroendocrine tumours. Best Pract Res Clin Endocrinol Metab. 2007 Mar. 21(1):87-109. [View Abstract]
  27. King J, Quinn R, Glenn DM, Janssen J, Tong D, Liaw W, et al. Radioembolization with selective internal radiation microspheres for neuroendocrine liver metastases. Cancer. 2008 Sep 1. 113(5):921-9. [View Abstract]
  28. Bramley PN, Lodge JP, Losowsky MS, Giles GR. Treatment of metastatic Vipoma by liver transplantation. Clin Transplant. 1990 Oct. 4(5 part 1):276-8; discussion 279. [View Abstract]

Patient with a large VIPoma. (A) Arteriogram showing vascularity of a large VIPoma preoperatively. (B) Large mass seen intraoperatively. (C) Gross pathologic specimen. Patient subsequently developed liver metastases; he was treated with chemoembolization of liver masses multiple times and finally succumbed to disease 20 years after initial surgical treatment.

Patient with a large VIPoma. (A) Arteriogram showing vascularity of a large VIPoma preoperatively. (B) Large mass seen intraoperatively. (C) Gross pathologic specimen. Patient subsequently developed liver metastases; he was treated with chemoembolization of liver masses multiple times and finally succumbed to disease 20 years after initial surgical treatment.