U.S. flag

An official website of the United States government

NCBI Bookshelf. A service of the National Library of Medicine, National Institutes of Health.

Feingold KR, Anawalt B, Blackman MR, et al., editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000-.

Cover of Endotext

Endotext [Internet].

Show details

Vasoactive Intestinal Peptide-Secreting Tumor (VIPoma)

, MD, PhD and , MD, PhD.

Author Information and Affiliations

Last Update: April 5, 2023.

ABSTRACT

A VIPoma is a neuroendocrine neoplasm secreting vasoactive intestinal peptide (VIP), usually presenting with severe watery secretory diarrhea, which can result in hypokalemia and metabolic acidosis and with flushes. Hypochlorhydria, stimulation of glycogenolysis, and hypercalcemia can be also found in VIPoma patients. Plasma VIP levels are elevated in all patients with the VIPoma syndrome, which is also known as “watery diarrhea, hypokalemia, achlorhydria (WDHA)-syndrome”, or “Verner-Morrison syndrome”. The majority of VIPomas are located in the pancreas (75%) and (usually young) patients can present with VIP-producing neuroblastoma, ganglioneuroblastoma, ganglioneuroma, pheochromocytoma and paraganglioma, or neoplasms of the retroperitoneum and mediastinum. The first treatment aim of a VIPoma patient is to correct the fluid and electrolyte deficits. Administration of a somatostatin analog (SSA) can decrease flushing and diarrhea, further aiding in the restoration of fluid and electrolyte imbalances. Surgical resection should be considered in patients with a locoregionally confined VIPoma. In patients with a metastatic or unresectable VIPoma, SSAs likely prolong progression-free survival. Other treatment options include peptide receptor radionuclide therapy (PRRT) with radiolabeled SSAs, interferon alpha, everolimus, sunitinib, cytotoxic chemotherapy, or liver-directed therapies. For complete coverage of all related areas of Endocrinology, please visit our on-line FREE web-text, WWW.ENDOTEXT.ORG.

INTRODUCTION

Vasoactive intestinal polypeptide (VIP) is a neurotransmitter found in the central nervous system, in neurons in the intestine, lungs, adrenals, pancreas and liver and in neuroendocrine cells in the pancreas (1). In the gastrointestinal tract, VIP stimulates contraction of enteric smooth muscle cells, secretion from the exocrine pancreas, gastrointestinal blood flow, and inhibits gastric acid secretion (2-4). A VIPoma is a neuroendocrine neoplasm (NEN) secreting VIP. VIP hypersecretion causes severe watery secretory diarrhea, which can result in hypokalemia and metabolic acidosis (VIPoma syndrome) (5).

HISTORY

In 1958 the US physician John V. Verner Jr. (1927-2022) and the Irish-US pathologist Ashton B. Morrison (1922-2008) reported on two patients with a VIPoma syndrome (6). Both patients presented with watery diarrhea and severe refractory hypokalemia and subsequently died of cardiac arrhythmias. Autopsy revealed pancreatic “islet cell” tumors in both patients (6). One of their patients was a 19-year-old male who also developed hypercalcemia and at autopsy hyperplasia of one of the parathyroid glands was found. The pituitary was not examined (6, 7). The publication by Verner and Morrison further cites 7 similar cases already published in the literature at that time (6) Thereafter, the VIPoma syndrome was also named “watery diarrhea, hypokalemia, achlorhydria (WDHA)-syndrome”, or “Verner-Morrison syndrome”. In the late 1960s and early 1970s, VIP was first isolated from the lungs and small intestine of experimental animals by the group of the Estonian scientist Viktor Mutt (1923-1998) in Sweden (8-10). In 1973, a radioimmunoassay for VIP became available and subsequently the British physician Stephen R. Bloom and colleagues could for the first time measure elevated VIP levels in the blood of a patient with the VIPoma syndrome (11). In 1983, the US gastroenterologist Mary G. Kane and colleagues injected five healthy subjects with porcine VIP, which resulted within 4 hours in high plasma VIP levels and was followed by secretory diarrhea in all patients (12).

CLINICAL PRESENTATION

Secretory diarrhea is the most characteristic symptom of a VIPoma. In severe cases, patients can produce up to 6-8L of watery stools per day. The stool is rich in electrolytes like potassium and bicarbonate, resulting in hypokalemia and metabolic acidosis in the VIPoma patient (13, 14). Another VIPoma symptom is facial flushing (occurring in 15-30% of patients). Hypochlorhydria, stimulation of glycogenolysis, and hypercalcemia can be diagnosed in patients with a VIPoma (5, 14-19). VIP has a structural homology with secretin, glucagon, and GIP which may account for the enhanced secretion of pancreatic enzymes, inhibition of gastric acid secretion, and glycogenolysis (9). The cause of the patchy erythematous flushing is not clear, but the flushing has been attributed to VIP, or to prostaglandins co-secreted by the tumor. Approximately 50% of patients have hypercalcemia, but again the mechanism of action is unknown. Hypercalcemia might be related to the co-secretion of parathyroid hormone related peptide (PTHrp) by the tumor (20, 21), or in specific cases coexisting primary hyperparathyroidism in the spectrum of the multiple endocrine neoplasia 1 (MEN1) syndrome (7).

Pancreatic VIPomas account for only 0.6–1.5% of all pancreatic neuroendocrine neoplasms (panNENs) (17) and approximately 2–6% of all functioning panNENs (17). The incidence is 0.05–0.2 cases per 1 million person-years with no gender predilection (15, 17, 18, 22). The mean age of these patients is 50.5 years (17). Pancreatic VIPomas can be associated with the MEN1 syndrome, but they are present in less than 1% of MEN1 patients (7, 23, 24). Around 75-90% of WDHA syndrome originates from a VIP-secreting panNEN. Approximately 70% of these pancreatic VIPomas are located in the body or tail and 30% in the head (18, 19, 25, 26). 10-25% of the WDHA syndrome derives from extra-pancreatic sources and can be found in patients with neuroblastoma, ganglioneuroblastoma, ganglioneuroma, pheochromocytoma and paraganglioma, and neoplasms of the retroperitoneum and mediastinum (5, 19, 27-30). The neurogenic tumors are more commonly found in the pediatric population (mean age 7.3 years). VIP-production from medullary thyroid carcinoma and lung neoplasms can also occur but this generally does not lead to the VIPoma / WDHA syndrome (31-33).

DIAGNOSIS

In the circulation, VIP has a very short half-life of less than 1 minute and, normally, plasma levels of VIP are low (below 20 pmol/L = 70 pg/mL) (34, 35). In the absence of a VIPoma, plasma VIP levels reflect the overflow of VIP from VIP-containing vascular nerves. By definition, plasma VIP levels should be elevated in all patients with the VIPoma syndrome. Bloom and colleagues measured plasma VIP levels in nearly 1000 patients with diarrhea and the diagnosis of VIPoma could be confirmed in all patients with plasma VIP levels greater than 60 pmol/L (= 203 pg/mL) (13, 34). In another series of 52 pancreatic VIPoma patients, elevated VIP levels were also measured with a median of 188 pmol/L (= 630 pg/mL - range 30-2131 pmol/L) (14). Moderately elevated plasma VIP levels can also be caused by gastrointestinal ischemia, renal insufficiency, or congestive heart failure (36-38).

The diameter of the primary pancreatic VIPoma is on average larger than 2 cm in 80% of patients (19). Therefore, these tumors can be easily detected with abdominal MRI, 3 phase CT, or endoscopic ultrasound (EUS). Additionally, a positron emission tomography (PET)-CT/MRI with 68Ga-labelled somatostatin analogs (DOTATATE, DOTANOC, DOTATOC) should be performed to determine, or exclude metastatic spread. In most centers, somatostatin receptor scintigraphy and SPECT using 111In-pentetreotide (OctreoScan) has become obsolete. In a small case series, 111In-pentetreotide scintigraphy proved to be superior to conventional radiological imaging for localizing the VIPoma and its metastases (39).

Similar to work-up for all NENs, a biopsy of the primary tumor or its metastases is recommended to confirm the diagnosis and for grading (Ki67 index), since the tumor grade can influence treatment decisions (17). An overview of the current panNEN staging and grading systems is provided in the chapter “Insulinoma” (40). Pancreatic VIPoma tumor cells usually express neuroendocrine differentiation markers (chromogranin-A, synaptophysin, INSM1), keratins, transcription factors, and somatostatin receptor subtype 2 (17). The extent of VIP expression can be variable given the rapid turnover of the protein synthesis. Secondary, or metachronous insulin secretion and/or positive insulin immunohistochemistry on the tumor specimen is generally associated with poor survival (41-43).

In patients with metastatic VIPoma, the 5-years survival is 60% (14, 16). Patients with high circulating VIP levels (plasma VIP ≥ 5xULN) have a poorer prognosis than those with moderately elevated levels (plasma VIP <5xULN) (16).

TREATMENT

Correction of Fluid and Electrolyte Deficits

The first treatment aim in a patient with a VIPoma is to correct the fluid and electrolyte deficits. In the majority of severe cases, intravenous resuscitation with saline, potassium and bicarbonate is required. Administration of a somatostatin analog (SSA) can decrease the secretory diarrhea, further aiding in the restoration of fluid and electrolyte imbalances (13, 44, 45). In the acute setting, the SSA octreotide can be administered subcutaneously, or via continuous intravenous infusion (46).

Surgery

After initial stabilization, a surgical resection should be performed in patients with a locoregionally confined VIPoma. The 5-year overall survival after surgery of patients with a localized VIPoma is >90% (14, 16, 19). In these patients the symptomatology of the VIPoma syndrome also completely resolved after surgery (16). Extended surgical resection, also involving the liver, can be considered in selected patients with limited liver metastases (47).

In case of an unresectable VIPoma, treatment is focused on tumor stabilization and control of VIP hypersecretion and symptoms (16). In general, anti-tumor therapy is similar to that used for other non-functioning and functioning panNENs and described in the guidelines by ENETS, NANETS and ESMO (48-50).

Somatostatin Analogs

Somatostatin analogs (SSAs) represent the first-line palliative treatment for metastatic or unresectable VIPomas. SSAs can have an antiproliferative effect, based on randomized trials with low grade (G1-G2) panNEN. In the CLARINET trial, including grade 1-2 panNENs, treatment with lanreotide autogel (120 mg every 4 weeks) prolonged median progression-free survival (PFS) from 18 to 38 months as compared to placebo by slowing tumor growth (51, 52). Treatment with SSAs results in a reduction of diarrhea episodes and volume in approximately 65-85% of VIPoma patients (15, 16, 45, 53, 54). It is, therefore, recommended to continue SSAs for symptom control when further lines of treatment are instituted for the control of tumor progression.

Everolimus

Everolimus is registered for the second-line treatment of G1-2 panNENs based on the result of the RADIANT-3 trial. In this study, 24% of patients had a functioning (= hormone-secreting) panNEN and treatment with everolimus (10 mg / day) improved median progression-free survival by 6.4 months compared with placebo. Everolimus treatment was associated with a (statistically not significant) overall survival benefit of 6.3 months (55, 56). Only a few VIPoma patients treated with Everolimus have been reported. In these patients, a symptomatic response was found in less than 10% of patients (15).

Sunitinib

In a randomized controlled trial in patients with G1-2 panNENs, second-line sunitinib treatment (37.5 mg/day) resulted in an increased progression-free survival by 5.9 months compared to placebo (57, 58). Two patients with a VIPoma were included in this trial, but they were both treated with placebo (57). In case series, a symptomatic response rate of 30-100% has been described for VIPoma patients treated with sunitinib (15, 16, 59, 60).

Other Medical Options

Next to SSAs, interferon-alpha is an established first-line antiproliferative and anti-secretory therapy for NENs of the gastrointestinal tract and pancreas either as monotherapy, or in combination with an SSA. However, the many side-effects mainly preclude its widespread use. Variable symptomatic responses with this therapy in VIPoma patients have been reported (61, 62). Prednisone has also been occasionally used to control the diarrhea frequency and stool volume in selected cases (45, 63).

Peptide Receptor Radionuclide Therapy

Peptide receptor radionuclide therapy (PRRT) with 177Lu-DOTATATE results in a response rate of 55% for panNENs, with a median PFS of 30 months and median overall survival (OS) of 71 months (64). PRRT with 177Lu-DOTATATE has only been reported in a limited number of patients with a VIPoma. In case series, the symptomatic response rate of VIPomas to this therapy was approximately 80% and disease control rate was 67% (15, 65, 66). Withdrawal from non-radioactive SSAs can lead to swift recurrence of severe watery diarrhea, providing rationale to limit the time for SSA withdrawal before PRRT cycles with 177Lu-DOTATATE to a very minimum e.g., by continued use of short-acting octreotide until shortly before the administration of this therapy (64).

Liver Directed Therapy

In patients with liver-dominant disease, liver metastases can be resected or treated by bland embolization, radioembolization (SIRT), radiofrequency ablation (RFA), microwave and cryoablation, high-intensity focused ultrasound (HIFU), laser, brachytherapy and irreversible electroporation (IRE) depending on local availability (47). Reduction of liver tumor burden was associated with a symptomatic response of VIPomas in small series (15, 16, 67). Orthotopic liver transplantation with removal of the diseased liver in VIPoma patients preoperatively diagnosed with “liver-only” disease can result in an improved disease course, or even complete cure (68-70).

Chemotherapy

Chemotherapy is also effective for the treatment of panNEN with symptomatic and tumor growth control achieved in a significant proportion of VIPoma patients (42, 55, 56)(15, 16, 43).

REFERENCES

1.
Larsson LI, Fahrenkrug J, Schaffalitzky De Muckadell O, Sundler F, Håkanson R, Rehfeld JR. Localization of vasoactive intestinal polypeptide (VIP) to central and peripheral neurons. Proc Natl Acad Sci U S A. 1976;73(9):3197–200. [PMC free article: PMC430979] [PubMed: 787988]
2.
Holst JJ, Fahrenkrug J, Knuhtsen S, Jensen SL, Poulsen SS, Nielsen OV. Vasoactive intestinal polypeptide (VIP) in the pig pancreas: role of VIPergic nerves in control of fluid and bicarbonate secretion. Regul Pept. 1984;8(3):245–59. [PubMed: 6379759]
3.
Robberecht P, Conlon TP, Gardner JD. Interaction of porcine vasoactive intestinal peptide with dispersed pancreatic acinar cells from the guinea pig. Structural requirements for effects of vasoactive intestinal peptide and secretin on cellular adenosine 3':5'-monophosphate. J Biol Chem. 1976;251(15):4635–9. [PubMed: 181379]
4.
Barbezat GO, Grossman MI. Intestinal secretion: stimulation by peptides. Science. 1971;174(4007):422–4. [PubMed: 5111998]
5.
Krejs GJ. VIPoma syndrome. Am J Med. 1987;82(5b):37–48. [PubMed: 3035922]
6.
Verner JV, Morrison AB. Islet cell tumor and a syndrome of refractory watery diarrhea and hypokalemia. Am J Med. 1958;25(3):374–80. [PubMed: 13571250]
7.
Pieterman CRC, van Leeuwaarde RS, van den Broek MFM, van Nesselrooij BPM, Valk GD. Multiple Endocrine Neoplasia Type 1. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, et al., editors. Endotext. South Dartmouth (MA): MDText.com, Inc.; 2000.
8.
Said SI, Mutt V. Potent peripheral and splanchnic vasodilator peptide from normal gut. Nature. 1970;225(5235):863–4. [PubMed: 5415118]
9.
Mutt V, Said SI. Structure of the porcine vasoactive intestinal octacosapeptide. The amino-acid sequence. Use of kallikrein in its determination. Eur J Biochem. 1974;42(2):581–9. [PubMed: 4829446]
10.
Said SI, Mutt V. A peptide fraction from lung tissue with prolonged peripheral vasodilator activity. Scand J Clin Lab Invest Suppl. 1969;107:51–6. [PubMed: 5375727]
11.
Bloom SR, Polak JM, Pearse AG. Vasoactive intestinal peptide and watery-diarrhoea syndrome. Lancet. 1973;2(7819):14–6. [PubMed: 4123289]
12.
Kane MG, O'Dorisio TM, Krejs GJ. Production of secretory diarrhea by intravenous infusion of vasoactive intestinal polypeptide. N Engl J Med. 1983;309(24):1482–5. [PubMed: 6316136]
13.
Kapoor R, Moseley RH, Kapoor JR, Crapo LM, Saint S. Clinical problem-solving. Needle in a haystack. N Engl J Med. 2009;360(6):616–21. [PubMed: 19196679]
14.
Schizas D, Mastoraki A, Bagias G, Patras R, Moris D, Lazaridis II, et al. Clinicopathological data and treatment modalities for pancreatic vipomas: a systematic review. J buon. 2019;24(2):415–23. [PubMed: 31127985]
15.
Angelousi A, Koffas A, Grozinsky-Glasberg S, Gertner J, Kassi E, Alexandraki K, et al. Diagnostic and Management Challenges in Vasoactive Intestinal Peptide Secreting Tumors: A Series of 15 Patients. Pancreas. 2019;48(7):934–42. [PubMed: 31268974]
16.
Brugel M, Walter T, Goichot B, Smith D, Lepage C, Do Cao C, et al. Efficacy of treatments for VIPoma: A GTE multicentric series. Pancreatology. 2021;21(8):1531–9. [PubMed: 34404601]
17.
Mete O, Basturk O, Brosens LA, de Herder W, Hong SM, La Rosa S. VIPoma. In: Klimstra DS, Osamura RY, editors. WHO Classification of Tumours Endocrine and neuroendocrine tumours. Lyon: International Agency for Research on Cancer; 2023.
18.
Siddappa PK, Vege SS. Vasoactive Intestinal Peptide-Secreting Tumors: A Review. Pancreas. 2019;48(9):1119–25. [PubMed: 31609932]
19.
Soga J, Yakuwa Y. Vipoma/diarrheogenic syndrome: a statistical evaluation of 241 reported cases. J Exp Clin Cancer Res. 1998;17(4):389–400. [PubMed: 10089056]
20.
Kamp K, Feelders RA, van Adrichem RC, de Rijke YB, van Nederveen FH, Kwekkeboom DJ, et al. Parathyroid hormone-related peptide (PTHrP) secretion by gastroenteropancreatic neuroendocrine tumors (GEP-NETs): clinical features, diagnosis, management, and follow-up. J Clin Endocrinol Metab. 2014;99(9):3060–9. [PubMed: 24905065]
21.
Kaltsas G, Androulakis II, de Herder WW, Grossman AB. Paraneoplastic syndromes secondary to neuroendocrine tumours. Endocr Relat Cancer. 2010;17(3):R173–93. [PubMed: 20530594]
22.
Halfdanarson TR, Strosberg JR, Tang L, Bellizzi AM, Bergsland EK. The North American Neuroendocrine Tumor Society Consensus Guidelines for Surveillance and Medical Management of Pancreatic Neuroendocrine Tumors. Pancreas. 2020;49(7):863–81. OʼDorisio TM, et al. [PubMed: 32675783]
23.
Lévy-Bohbot N, Merle C, Goudet P, Delemer B, Calender A, Jolly D, et al. Prevalence, characteristics and prognosis of MEN 1-associated glucagonomas, VIPomas, and somatostatinomas: study from the GTE (Groupe des Tumeurs Endocrines) registry. Gastroenterol Clin Biol. 2004;28(11):1075–81. [PubMed: 15657529]
24.
Niederle B, Selberherr A, Bartsch D, Brandi ML, Doherty GM, Falconi M, et al. Multiple Endocrine Neoplasia Type 1 (MEN1) and the Pancreas - Diagnosis and Treatment of Functioning and Non-Functioning Pancreatic and Duodenal Neuroendocrine Neoplasia within the MEN1 Syndrome - An International Consensus Statement. Neuroendocrinology. 2020 [PubMed: 32971521]
25.
Smith SL, Branton SA, Avino AJ, Martin JK, Klingler PJ, Thompson GB, et al. Vasoactive intestinal polypeptide secreting islet cell tumors: a 15-year experience and review of the literature. Surgery. 1998;124(6):1050–5. [PubMed: 9854582]
26.
Ghaferi AA, Chojnacki KA, Long WD, Cameron JL, Yeo CJ. Pancreatic VIPomas: subject review and one institutional experience. J Gastrointest Surg. 2008;12(2):382–93. [PubMed: 17510774]
27.
Leibowitz-Amit R, Mete O, Asa SL, Ezzat S, Joshua AM. Malignant pheochromocytoma secreting vasoactive intestinal peptide and response to sunitinib: a case report and literature review. Endocr Pract. 2014;20(8):e145–50. [PubMed: 24936559]
28.
Murahashi N, Abe T, Matsumoto R, Oosawa T, Yoshinaga K, Shiga T, et al. Hinyokika Kiyo. 2019;65(7):277–82. [A Case of WDHA Water Diarrhea Hypokalemia Achlorhydria Syndrome that Developed after Multimodal Therapy for Retroperitoneal Paraganglioma] [PubMed: 31501391]
29.
Onozawa M, Fukuhara T, Minoguchi M, Takahata M, Yamamoto Y, Miyake T, et al. Hypokalemic rhabdomyolysis due to WDHA syndrome caused by VIP-producing composite pheochromocytoma: a case in neurofibromatosis type 1. Jpn J Clin Oncol. 2005;35(9):559–63. [PubMed: 16027147]
30.
Ikuta S, Yasui C, Kawanaka M, Aihara T, Yoshie H, Yanagi H, et al. Watery diarrhea, hypokalemia and achlorhydria syndrome due to an adrenal pheochromocytoma. World J Gastroenterol. 2007;13(34):4649–52. [PMC free article: PMC4611845] [PubMed: 17729424]
31.
Hanna FW, Ardill JE, Johnston CF, Cunningham RT, Curry WJ, Russell CF, et al. Regulatory peptides and other neuroendocrine markers in medullary carcinoma of the thyroid. J Endocrinol. 1997;152(2):275–81. [PubMed: 9071985]
32.
Said SI. Evidence for secretion of vasoactive intestinal peptide by tumours of pancreas, adrenal medulla, thyroid and lung: support for the unifying APUD concept. Clin Endocrinol (Oxf). 1976;5 Suppl:201s–4s. [PubMed: 29733]
33.
Said SI, Faloona GR. Elevated plasma and tissue levels of vasoactive intestinal polypeptide in the watery-diarrhea syndrome due to pancreatic, bronchogenic and other tumors. N Engl J Med. 1975;293(4):155–60. [PubMed: 166313]
34.
Bloom SR. Vasoactive intestinal peptide, the major mediator of the WDHA (pancreatic cholera) syndrome: value of measurement in diagnosis and treatment. Am J Dig Dis. 1978;23(4):373–6. [PubMed: 208414]
35.
Domschke S, Domschke W, Bloom SR, Mitznegg P, Mitchell SJ, Lux G, et al. Vasoactive intestinal peptide in man: pharmacokinetics, metabolic and circulatory effects. Gut. 1978;19(11):1049–53. [PMC free article: PMC1412244] [PubMed: 730072]
36.
Modlin IM, Bloom SR, Mitchell S. Plasma vasoactive intestinal polypeptide (VIP) levels and intestinal ischaemia. Experientia. 1978;34(4):535–6. [PubMed: 639959]
37.
Doherty CC, Buchanan KD, Ardill J, McGeown MG. Elevations of gastrointestinal hormones in chronic renal failure. Proc Eur Dial Transplant Assoc. 1978;15:456–65. [PubMed: 740678]
38.
Clark AJ, Adrian TE, McMichael HB, Bloom SR. Vasoactive intestinal peptide in shock and heart failure. Lancet. 1983;1(8323):539. [PubMed: 6131249]
39.
Nikou GC, Toubanakis C, Nikolaou P, Giannatou E, Safioleas M, Mallas E, et al. VIPomas: an update in diagnosis and management in a series of 11 patients. Hepatogastroenterology. 2005;52(64):1259–65. [PubMed: 16001675]
40.
de Herder WW, Zandee WT, Hofland J. Insulinoma. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, et al., editors. Endotext. South Dartmouth (MA): MDText.com, Inc.; 2000.
41.
de Mestier L, Hentic O, Cros J, Walter T, Roquin G, Brixi H, et al. Metachronous hormonal syndromes in patients with pancreatic neuroendocrine tumors: a case-series study. Ann Intern Med. 2015;162(10):682–9. [PubMed: 25984844]
42.
Crona J, Norlén O, Antonodimitrakis P, Welin S, Stålberg P, Eriksson B. Multiple and Secondary Hormone Secretion in Patients With Metastatic Pancreatic Neuroendocrine Tumours. J Clin Endocrinol Metab. 2016;101(2):445–52. [PubMed: 26672633]
43.
Marques B, Monteiro AR, Martins RG, Couto J, Rodrigues F, Ribeiro J. Metastatic VIPoma, Cosecreting Insulin, With Complete Response to Lanreotide, Capecitabine, and Temozolomide. Pancreas. 2020;49(3):e19–e20. [PubMed: 32168252]
44.
Ito T, Jensen RT. Perspectives on the current pharmacotherapeutic strategies for management of functional neuroendocrine tumor syndromes. Expert Opin Pharmacother. 2021;22(6):685–93. [PMC free article: PMC8742468] [PubMed: 33131345]
45.
O'Dorisio TM, Mekhjian HS, Gaginella TS. Medical therapy of VIPomas. Endocrinol Metab Clin North Am. 1989;18(2):545–56. [PubMed: 2545444]
46.
Kaltsas G, Caplin M, Davies P, Ferone D, Garcia-Carbonero R, Grozinsky-Glasberg S, et al. ENETS Consensus Guidelines for the Standards of Care in Neuroendocrine Tumors: Pre- and Perioperative Therapy in Patients with Neuroendocrine Tumors. Neuroendocrinology. 2017;105(3):245–54. [PMC free article: PMC5637287] [PubMed: 28253514]
47.
Frilling A, Modlin IM, Kidd M, Russell C, Breitenstein S, Salem R, et al. Recommendations for management of patients with neuroendocrine liver metastases. Lancet Oncol. 2014;15(1):e8–21. [PubMed: 24384494]
48.
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;42(4):557–77. [PMC free article: PMC4304762] [PubMed: 23591432]
49.
Pavel M, O'Toole D, Costa F, Capdevila J, Gross D, Kianmanesh R, et al. ENETS Consensus Guidelines Update for the Management of Distant Metastatic Disease of Intestinal, Pancreatic, Bronchial Neuroendocrine Neoplasms (NEN) and NEN of Unknown Primary Site. Neuroendocrinology. 2016;103(2):172–85. [PubMed: 26731013]
50.
Pavel M, Öberg K, Falconi M, Krenning EP, Sundin A, Perren A, et al. Gastroenteropancreatic neuroendocrine neoplasms: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2020;31(7):844–60. [PubMed: 32272208]
51.
Caplin ME, Pavel M, Ćwikła JB, Phan AT, Raderer M, Sedláčková E, et al. Lanreotide in metastatic enteropancreatic neuroendocrine tumors. N Engl J Med. 2014;371(3):224–33. [PubMed: 25014687]
52.
Dromain C, Loaiza-Bonilla A, Mirakhur B, Beveridge TJR, Fojo AT. Novel Tumor Growth Rate Analysis in the Randomized CLARINET Study Establishes the Efficacy of Lanreotide Depot/Autogel 120 mg with Prolonged Administration in Indolent Neuroendocrine Tumors. Oncologist. 2021;26(4):e632–e8. [PMC free article: PMC8018300] [PubMed: 33393112]
53.
O'Dorisio TM, Anthony LB. A. 25-Year Experience of Gastroenteropancreatic Neuroendocrine Tumors and Somatostatin (Congeners) Analogs: From Symptom Control to Antineoplastic Therapy. Front Horm Res. 2015;44:177–92. [PubMed: 26303712]
54.
Chen C, Zheng Z, Li B, Zhou L, Pang J, Wu W, et al. Pancreatic VIPomas from China: Case reports and literature review. Pancreatology. 2019;19(1):44–9. [PubMed: 30391116]
55.
Yao JC, Pavel M, Lombard-Bohas C, Van Cutsem E, Voi M, Brandt U, et al. Everolimus for the Treatment of Advanced Pancreatic Neuroendocrine Tumors: Overall Survival and Circulating Biomarkers From the Randomized, Phase III RADIANT-3 Study. J Clin Oncol. 2016;34(32):3906–13. [PMC free article: PMC5791842] [PubMed: 27621394]
56.
Yao JC, Shah MH, Ito T, Bohas CL, Wolin EM, Van Cutsem E, et al. Everolimus for advanced pancreatic neuroendocrine tumors. N Engl J Med. 2011;364(6):514–23. [PMC free article: PMC4208619] [PubMed: 21306238]
57.
Raymond E, Dahan L, Raoul JL, Bang YJ, Borbath I, Lombard-Bohas C, et al. Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. N Engl J Med. 2011;364(6):501–13. [PubMed: 21306237]
58.
Fazio N, Kulke M, Rosbrook B, Fernandez K, Raymond E. Updated Efficacy and Safety Outcomes for Patients with Well-Differentiated Pancreatic Neuroendocrine Tumors Treated with Sunitinib. Target Oncol. 2021;16(1):27–35. [PMC free article: PMC7810649] [PubMed: 33411058]
59.
Bourcier ME, Vinik AI. Sunitinib for the treatment of metastatic paraganglioma and vasoactive intestinal polypeptide-producing tumor (VIPoma). Pancreas. 2013;42(2):348–52. [PubMed: 23407483]
60.
de Mestier L, Walter T, Brixi H, Lombard-Bohas C, Cadiot G. Sunitinib achieved fast and sustained control of VIPoma symptoms. Eur J Endocrinol. 2015;172(1):K1–3. [PubMed: 25305306]
61.
Anderson JV, Bloom SR. Treatment of malignant endocrine pancreatic tumours with human leucocyte interferon. Lancet. 1987;1(8524):97. [PubMed: 2879191]
62.
Oberg K, Alm G, Lindström H, Lundqvist G. Successful treatment of therapy-resistant pancreatic cholera with human leucocyte interferon. Lancet. 1985;1(8431):725–7. [PubMed: 2857998]
63.
Barraclough MA, Bloom SR. Vipoma of the pancreas: observations on the diarhrhea and circulatory disturbances. Arch Intern Med. 1979;139(4):467–71. [PubMed: 435002]
64.
Brabander T, van der Zwan WA, Teunissen JJM, Kam BLR, Feelders RA, de Herder WW, et al. Long-Term Efficacy, Survival, and Safety of [(177)Lu-DOTA(0),Tyr(3)]octreotate in Patients with Gastroenteropancreatic and Bronchial Neuroendocrine Tumors. Clin Cancer Res. 2017;23(16):4617–24. [PubMed: 28428192]
65.
Zandee WT, Brabander T, Blazevic A, Kam BLR, Teunissen JJM, Feelders RA, et al. Symptomatic and Radiological Response to 177Lu-DOTATATE for the Treatment of Functioning Pancreatic Neuroendocrine Tumors. J Clin Endocrinol Metab. 2019;104(4):1336–44. [PubMed: 30566620]
66.
Audil HY, Eiring RA, Kendi AT, Halfdanarson TR. A Case of Metastatic VIPoma With Complete Response to Peptide Radionuclide Receptor Therapy. Pancreas. 2021;50(4):e45–e6. [PubMed: 33939683]
67.
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;8(5):383–7. [PMC free article: PMC4981295] [PubMed: 27583030]
68.
Hengst K, Nashan B, Avenhaus W, Ullerich H, Schlitt HJ, Flemming P, et al. Metastatic pancreatic VIPoma: deteriorating clinical course and successful treatment by liver transplantation. Z Gastroenterol. 1998;36(3):239–45. [PubMed: 9577908]
69.
Bramley PN, Lodge JP, Losowsky MS, Giles GR. Treatment of metastatic Vipoma by liver transplantation. Clin Transplant. 1990;4(5 part 1):276–8. [PubMed: 10150088]
70.
Johnston PC, Ardill JE, Johnston BT, Mc Cance DR. Vasoactive intestinal polypeptide secreting pancreatic tumour with hepatic metastases: long term survival after orthotopic liver transplantation. Ir J Med Sci. 2010;179(3):439–41. [PubMed: 18825477]
Copyright © 2000-2024, MDText.com, Inc.

This electronic version has been made freely available under a Creative Commons (CC-BY-NC-ND) license. A copy of the license can be viewed at http://creativecommons.org/licenses/by-nc-nd/2.0/.

Bookshelf ID: NBK278960PMID: 25905195

Views

  • PubReader
  • Print View
  • Cite this Page

LINKS TO WWW.ENDOTEXT.ORG

Related information

  • PMC
    PubMed Central citations
  • PubMed
    Links to PubMed

Similar articles in PubMed

See reviews...See all...

Recent Activity

Your browsing activity is empty.

Activity recording is turned off.

Turn recording back on

See more...