Gastroenterology

Gastroenterology

Volume 135, Issue 5, November 2008, Pages 1469-1492
Gastroenterology

Reviews in Basic and Clinical Gastroenterology
Gastrointestinal Neuroendocrine Tumors: Pancreatic Endocrine Tumors

https://doi.org/10.1053/j.gastro.2008.05.047Get rights and content

Pancreatic endocrine tumors (PETs) have long fascinated clinicians and investigators despite their relative rarity. Their clinical presentation varies depending on whether the tumor is functional or not, and also according to the specific hormonal syndrome produced. Tumors may be sporadic or inherited, but little is known about their molecular pathology, especially the sporadic forms. Chromogranin A appears to be the most useful serum marker for diagnosis, staging, and monitoring. Initially, therapy should be directed at the hormonal syndrome because this has the major initial impact on the patient's health. Most PETs are relatively indolent but ultimately malignant, except for insulinomas, which predominantly are benign. Surgery is the only modality that offers the possibility of cure, although it generally is noncurative in patients with Zollinger–Ellison syndrome or nonfunctional PETs with multiple endocrine neoplasia-type 1. Preoperative staging of disease extent is necessary to determine the likelihood of complete resection although debulking surgery often is believed to be useful in patients with unresectable tumors. Once metastatic, biotherapy is usually the first modality used because it generally is well tolerated. Systemic or regional therapies generally are reserved until symptoms occur or tumor growth is rapid. Recently, a number of newer agents, as well as receptor-directed radiotherapy, are being evaluated for patients with advanced disease. This review addresses a number of recent advances regarding the molecular pathology, diagnosis, localization, and management of PETs including discussion of peptide-receptor radionuclide therapy and other novel antitumor approaches. We conclude with a discussion of future directions and unsettled problems in the field.

Section snippets

Epidemiology

PETs occur in 0.5%–1.5% of autopsies but are functional or symptomatic in less than 1 in 1000, resulting in a clinical detection rate of 1:100,000 population, which comprises 1%–2% of pancreatic neoplasms.8 In older studies, nonfunctional PETs (NF-PETs), insulinomas, and gastrinomas had equal frequency;9 however, in recent studies NF-PETs were twice as frequent.10, 11 The relative frequency of PETs varies in surgical or medical series, but most studies suggest a relative order of: NF-PET >

Classification/Pathology

PETs are divided clinically into 2 groups: functional and NF-PETs. Functional PETs secrete biologically active peptides causing 1 of 9 well-established syndromes (Table 1). NF-PETs are not associated with a specific hormonal syndrome either because no peptide is secreted or the substance secreted does not cause specific symptoms. Most (>70%) NF-PETs are not truly nonfunctional because they secrete substances such as pancreatic polypeptide, other peptides (neurotensin, ghrelin, and so forth),

Molecular Pathogenesis

Little is known about the molecular pathogenesis of PETs.1, 2, 8 This has occurred in part because alterations in common oncogenes (fos, jun, myc, k-ras, and so forth) or common tumor-suppressor genes (p53, retinoblastoma, and so forth) are not generally implicated in their pathogenesis.1, 2, 8 Some of the most important insights have come from studies of inherited PET syndromes.1, 2, 8, 20 Altered genes causing these syndromes are important in some cases of sporadic PETs (ie, nonfamilial

Tumor Biology, Prognosis, and Tumor Markers

PETs differ in their malignant potential and location (Table 1). Some PETs (insulinomas, glucagonomas, and VIPomas in adults) are found almost entirely within the pancreas, whereas others, although still referred to as PETs, actually occur in extrapancreatic locations (duodenal gastrinomas [60%–80%],30, 31, 32 small intestinal somatostatinomas [40%–50%], and growth hormone-releasing factor secreting tumor (GRFomas) primarily in the lung [>70%]) (Table 1). Insulinomas are malignant in 5%–15%,

Gastrinoma: Clinical Features/Diagnosis

Gastrinomas secrete gastrin, which causes hyperchlorhydria, thereby producing the Zollinger–Ellison syndrome (ZES).31, 45, 59, 60 With a long mean delay (6.1 y) in presentation/diagnosis,45, 61, 62 patients generally present with acid-peptic conditions including complicated and uncomplicated ulcers and/or gastroesophageal reflux disease (Table 2,top). Occasionally other manifestations such as diarrhea, malabsorption, or, in MEN1/ZES patients, various other endocrine features predominate (Table 2

Tumor Localization/Staging

Imaging studies are essential for the management of patients with PETs. They are needed to localize the primary tumor as well as for staging to guide management, including surgical plans (curative resection, debulking, or medical management only), to monitor tumor growth, and for follow-up evaluation after therapy.6, 9, 103, 104, 105, 106, 107, 108

Gastrinoma: Medical Management

In ZES acid hypersecretion is the most important clinical effect.45, 62, 167, 168 Because of their potency and long duration of action, PPIs are the agents of choice for management.31, 45, 53, 167, 169, 170 Histamine H2-receptor antagonists or SS analogues are effective, but the former drug class is limited by the need for frequent, high-dose administration,167, 169 whereas the latter class is limited by the need for parenteral therapy.

Once- or twice-daily oral PPIs (ie, omeprazole [40 mg],

Surgical Therapy for Cure

Surgery is the only treatment modality with the potential to cure patients with PETs. However, surgery is only likely to be effective in patients without diffuse metastatic disease who are able to tolerate the intervention and, in the case of ZES specifically, only in those with sporadic disease.13, 113, 197, 198, 199 Negative preoperative localization should not be considered a contraindication to surgery in patients with proven functional PETs because an experienced PET surgeon will very

General Treatment of Metastatic Disease

In recent studies the long-term outcome in patients with PETs is increasingly dependent on tumor growth. However, even with widespread liver metastases many patients remain relatively well with slow progression, especially early on in the disease process, such that many authorities advocate delaying the introduction of disease-modifying agents until there is clear development of enlarging tumor burden, or symptoms develop. Furthermore, standard antitumor therapies are not curative and

Liver-Directed Therapy (Embolization, Chemoembolization)

Most malignant PETs metastasize to the liver where they derive their blood supply from hepatic artery branches (75%–80%), in contrast to native liver tissue, which derives the majority of its blood supply from the portal vein.9, 248, 249 Recent studies have shown that liver metastases show rapid growth in less than 50% of patients and up to 30% show no growth on follow-up evaluation.33, 37, 250 Consequently, the usual approach to palliative therapy for liver metastases is to delay therapy until

Future Directions and Unsettled Problems

Even though there have been many advances in recent years in the diagnosis/management of PETs, it is not clear that survival in patients with advanced disease has improved. In fact, in a recent review294 of survival for all GI NETs (both carcinoids and PETs), no change in survival was reported over a 30-year period. Numerous factors contribute to this including their continued delay in diagnosis (mean, 4–6 y); the lack of general availability to most patients of the expertise and experience

References (308)

  • R.V. Benya et al.

    Zollinger-Ellison syndrome can be the initial endocrine manifestation in patients with multiple endocrine neoplasia-type 1

    Am J Med

    (1994)
  • J.E. McGuigan et al.

    Secretin injection test in the diagnosis of gastrinoma

    Gastroenterology

    (1980)
  • C.S. Grant

    Insulinoma

    Best Pract Res Clin Gastroenterol

    (2005)
  • C.C. Case et al.

    Reproduction of features of the glucagonoma syndrome with continuous intravenous glucagon infusion as therapy for tumor-induced hypoglycemia

    Endocr Pract

    (2003)
  • E.A. Mullans et al.

    Iatrogenic necrolytic migratory erythema: a case report and review of nonglucagonoma-associated necrolytic migratory erythema

    J Am Acad Dermatol

    (1998)
  • A.A. Ghaferi et al.

    Pancreatic VIPomas: subject review and one institutional experience

    J Gastrointest Surg

    (2008)
  • B. Moayedoddin et al.

    Spectrum of malignant somatostatin-producing neuroendocrine tumors

    Endocr Pract

    (2006)
  • L. Gullo et al.

    Nonfunctioning pancreatic endocrine tumors: a multicenter clinical study

    Am J Gastroenterol

    (2003)
  • R. Arnold

    Endocrine tumors of the gastrointestinal tract: part 11

    Best Practice Res Clin Gastroent

    (2005)
  • R. Arnold

    Endocrine tumors of the gastrointestinal tract: part 1

    Best Practice Res Clin Gastroent

    (2005)
  • K. Oberg et al.

    Neuroendocrine tumors

    Best Pract Res Clin Endocrinol Metab

    (2007)
  • W.W. de Herder et al.

    ENETS consensus guidelines for the management of patients with digestive neuroendocrine tumors part 1-stomach, duodenum and pancreas

    Neuroendocrinology

    (2006)
  • U. Plockinger et al.

    Guidelines for the diagnosis and treatment of neuroendocrine gastrointestinal tumoursA consensus statement on behalf of the European Neuroendocrine Tumour Society (ENETS)

    Neuroendocrinology

    (2004)
  • R.T. Jensen

    Endocrine neoplasms of the pancreas

  • T. Ito et al.

    Preliminary results of a Japanese nationwide survey of neuroendocrine gastrointestinal tumors

    J Gastroenterol

    (2007)
  • F. Panzuto et al.

    Prognostic factors and survival in endocrine tumor patients: comparison between gastrointestinal and pancreatic localization

    Endocr Relat Cancer

    (2005)
  • R.T. Jensen et al.

    Inherited pancreatic endocrine tumor syndromes: advances in molecular pathogenesis, diagnosis, management and controversies

    Cancer

    (2008)
  • E. Brignardello et al.

    Ectopic secretion of LH by an endocrine pancreatic tumor

    J Endocrinol Invest

    (2004)
  • I. Samyn et al.

    Paraneoplastic syndromes in cancer: case 1Polycythemia as a result of ectopic erythropoietin production in metastatic pancreatic carcinoid tumor

    J Clin Oncol

    (2004)
  • K. Kawano et al.

    Peptide YY producing strumal carcinoid of the ovary as the cause of severe constipation with contralateral epithelial ovarian cancer

    J Obstet Gynaecol Res

    (2007)
  • G. Rindi et al.

    TNM staging of foregut (neuro)endocrine tumors: a consensus proposal including a grading system

    Virchows Arch

    (2006)
  • S.C. Chandrasekharappa et al.

    Positional cloning of the gene for multiple endocrine neoplasia—-type 1

    Science

    (1997)
  • V. Busygina et al.

    Multiple endocrine neoplasia type 1 (MEN1) as a cancer predisposition syndrome: clues into the mechanisms of MEN1-related carcinogenesis

    Yale J Biol Med

    (2006)
  • S.U. Goebel et al.

    Genotype/phenotype correlations of MEN1 gene mutations in sporadic gastrinoma

    J Clin Endocrinol Metab

    (2000)
  • L.V. Debelenko et al.

    Allelic deletions on chromosome 11q13 in multiple endocrine neoplasia type-I-associated sporadic gastrinomas and pancreatic endocrine tumors

    Cancer Res

    (1997)
  • A. Perren et al.

    Molecular genetics of gastroenteropancreatic endocrine tumors

    Ann N Y Acad Sci

    (2004)
  • G. Capurso et al.

    Gene expression profiles of progressive pancreatic endocrine tumours and their liver metastases reveal potential novel markers and therapeutic targets

    Endocr Relat Cancer

    (2006)
  • W.G. Dilley et al.

    Global gene expression in neuroendocrine tumors from patients with the MEN1 syndrome

    Mol Cancer

    (2005)
  • D.E. Hansel et al.

    Met proto-oncogene and insulin-like growth factor binding protein 3 overexpression correlates with metastatic ability in well-differentiated pancreatic endocrine neoplasms

    Clin Cancer Res

    (2004)
  • A. Maitra et al.

    Global expression analysis of well-differentiated pancreatic endocrine neoplasms using oligonucleotide microarrays

    Clin Cancer Res

    (2003)
  • R.T. Jensen et al.

    Gastrinoma (duodenal and pancreatic)

    Neuroendocrinology

    (2006)
  • A.K. Thom et al.

    Location, incidence and malignant potential of duodenal gastrinomas

    Surgery

    (1991)
  • R.T. Jensen

    Natural history of digestive endocrine tumors

  • F. Gibril et al.

    Bone metastases in patients with gastrinomas: a prospective study of bone scanning, somatostatin receptor scanning, and MRI in their detection, their frequency, location and effect of their detection on management

    J Clin Oncol

    (1998)
  • F. Yu et al.

    Prospective study of the clinical course, prognostic factors and survival in patients with longstanding Zollinger-Ellison syndrome

    J Clin Oncol

    (1999)
  • V.E. Sutliff et al.

    Growth of newly diagnosed, untreated metastatic gastrinomas and predictors of growth patterns

    J Clin Oncol

    (1997)
  • I. Madeira et al.

    Prognostic factors in patients with endocrine tumours of the duodenopancreatic area

    Gut

    (1998)
  • J. Soga et al.

    Vipoma/diarrheogenic syndrome: a statistical evaluation of 241 reported cases

    J Exp Clin Cancer Res

    (1998)
  • G. Nesi et al.

    Somatostatinoma: clinico-pathological features of three cases and literature reviewed

    J Gastroenterol Hepatol

    (2008)
  • Q.D. Chu et al.

    Predictive factors associated with long-term survival in patients with neuroendocrine tumors of the pancreas

    Ann Surg Oncol

    (2002)
  • Cited by (627)

    • Insulinoma in a patient with Congenital adrenal hyperplasia

      2023, Journal of Clinical and Translational Endocrinology: Case Reports
    • Pancreatic Cystic Lesions: Imaging Techniques and Diagnostic Features

      2023, Gastrointestinal Endoscopy Clinics of North America
    View all citing articles on Scopus

    The authors disclose the following: this work is partially supported by the Intramural Program of the NIDDK, National Institutes of Health.

    View full text