Association for Academic Surgery
GIPR expression in gastric and duodenal neuroendocrine tumors

https://doi.org/10.1016/j.jss.2014.01.044Get rights and content

Abstract

Background

Compounds targeting somatostatin-receptor-type-2 (SSTR2) are useful for small bowel neuroendocrine tumor (SBNET) and pancreatic neuroendocrine tumor (PNET) imaging and treatment. We recently characterized expression of 13 cell surface receptor genes in SBNETs and PNETs, identifying three drug targets (GIPR, OXTR, and OPRK1). This study set out to characterize expression of this gene panel in the less common neuroendocrine tumors of the stomach and duodenum (gastric and duodenal neuroendocrine tumors [GDNETs]).

Methods

Primary tumors and adjacent normal tissue were collected at surgery, RNA was extracted, and expression of 13 target genes was determined by quantitative polymerase chain reaction. Expression was normalized to GAPDH and POLR2A internal control genes. Expression relative to normal tissue (ddCT) and absolute expression (dCT) were calculated. Wilcoxon tests compared median expression with false discovery rate correction for multiple comparisons.

Results

Gene expression was similar in two gastric and seven duodenal tumors, and these were analyzed together. Like SBNETs (n = 63) and PNETs (n = 51), GDNETs showed significant overexpression compared with normal tissue of BRS3, GIPR, GRM1, GPR113, OPRK1, and SSTR2 (P < 0.05 for all). Of these, SSTR2 had the highest absolute expression in GDNETs (median dCT 4.0). Absolute expression of BRS3, GRM1, GPR113, and OPRK1 was significantly lower than SSTR2 in GDNETs (P < 0.05 for all), whereas expression of GIPR was similar to SSTR2 (median 4.3, P = 0.4).

Conclusions

As in SBNETs and PNETs, GIPR shows absolute expression close to SSTR2 but has greater overexpression relative to normal tissue (21.1 versus 3.5-fold overexpression). We conclude that GIPR could provide an improved signal-to-noise ratio for imaging versus SSTR2 and represents a promising novel therapeutic target in GDNETs.

Introduction

Duodenal neuroendocrine tumors constitute one of the rarest types of neuroendocrine tumors (NETs), with an annual incidence of 0.19 per 100,000 in the United States, representing approximately 4% of all NETs [1], [2]. Gastric neuroendocrine tumors have an incidence of 0.30 per 100,000, and their numbers have increased dramatically over time because of the proliferation of upper endoscopy and possibly the use of acid suppressive medications [1], [3], [4]. Although most gastric and duodenal neuroendocrine tumors (GDNETs) are small, amenable to endoscopic resection and rarely metastatic, a subpopulation exists of generally larger and more aggressive tumors requiring a more extensive staging evaluation, operative treatment, and often postoperative medical therapy [3].

Staging, preoperative planning, and medical treatment of other types of NETs exploit high expression of the somatostatin receptor in these tumors [5], [6]. Molecules such as octreotide, which binds principally to the somatostatin-receptor-type-2 (SSTR2), can decrease symptoms from hormone overproduction syndromes and reduce tumor progression in midgut NET patients [7]. Linking somatostatin analogs to radioligands such as 111In or 68Ga allows them to accumulate selectively at the tumor, permitting radiographic visualization of tumor tissues [8], [9], [10]. In peptide receptor radionuclide therapy, somatostatin analogs linked to higher-energy isotopes (90Y or 177Lu) can actually kill tumor cells [11]. Yet even with the success of these tests and therapies in some patients, not all NETs express somatostatin receptors and not all patients respond to octreotide [9], [12], [13], [14].

To build on the success of somatostatin-based NET treatments and respond to the dilemma of patients in whom they fail, our group used G protein–coupled receptor and exon expression microarrays to find new therapeutic targets, determining expression of 384 genes in a small number of initial tumor samples [15] and validating expression of a panel of 13 genes in over 100 SBNETs and PNETs [15], [16], [17]. In these studies, OPRK1 and OXTR in SBNETs and GIPR in both SBNETs and PNETs emerged as potentially useful receptors. These three genes displayed absolute expression similar to SSTR2, while having significantly higher expression in tumors relative to normal tissues. This suggested that imaging strategies directed at these receptors might display improved signal-to-background characteristics, and treatments might have fewer effects on noncancerous tissue.

In GDNETs, the optimal use of somatostatin-directed therapies is unclear. Because of GDNETs' rarity, most studies of somatostatin-based therapies do not include them or analyze them along with other tumor types [4]. To inform the use of peptide receptor–directed imaging and therapy in GDNETs and assess potential new therapeutic targets, we set out to determine expression levels of this 13-gene panel in the rare and poorly studied population of GDNETs and compared these results with our previous findings in SBNETs and PNETs.

Section snippets

Patients and clinical data

Patients undergoing surgery for abdominal NETs at a single center since 2005 were enrolled under an Institutional Review Board–approved protocol with full informed consent. Clinical and pathologic information for these patients was reviewed and included in the Iowa Neuroendocrine Tumor Registry Database [18]. Tumor and adjacent normal tissues were collected at the time of surgery and maintained in RNALater solution (Life Technologies, Grand Island, NY). GDNETs included NETs of the stomach and

Patient and tumor characteristics

Patients with gastric (n = 2) and duodenal (n = 7) NETs were included, and results compared with patients with SBNETs (n = 63) and PNETs (n = 51). Gene expression in the gastric NETs was found to be similar to that in duodenal tumors, and these were analyzed together (GDNETs, n = 9). Median age at surgery for GDNET patients was 57.3 (range 52.6–70.9). Median progression-free survival and overall survival were not yet reached. Preoperative laboratory values were available for most patients. The

Discussion

GDNETs that require operative treatment are a rare subset of all GDNETs for which optimal management remains uncertain. The present study demonstrates that GDNETs express SSTR2 at levels similar to SBNETs and PNETs, supporting the potential efficacy of somatostatin receptor–directed diagnostics and therapies in these tumors. That GDNETs also highly express GIPR, but with greater overexpression compared with background tissues, suggests that as in SBNETs and PNETs, GIPR is a potential target for

Acknowledgment

The authors gratefully acknowledge their patients for their participation.

Supported by National Institutes of Health 5T32#CA148062-03 (S.K.S., J.E.M., J.C.C.).

None of the authors has any potential conflicts of interest to disclose.

Author Contributions: Concept and design: S.K.S., J.C.C., M.S.O., T.M.O., J.R.H.; Analysis and Interpretation: S.K.S.; Data Collection: S.K.S., J.E.M., J.C.C., D.W.; Writing: S.K.S., J.E.M.; Critical Revision: M.S.O., T.M.O., J.R.H.; Obtaining Funding: J.R.H.

References (37)

  • S.K. Sherman et al.

    Gene expression in neuroendocrine tumor liver metastases accurately distinguishes between pancreas and small bowel primary tumors

    J Am Coll Surg

    (2013)
  • J.C. Yao et al.

    One hundred years after “carcinoid”: epidemiology of and prognostic factors for neuroendocrine tumors in 35,825 cases in the United States

    J Clin Oncol

    (2008)
  • R.W. Randle et al.

    Clinical Outcomes for neuroendocrine tumors of the duodenum and ampulla of Vater: a population-based study

    J Gastrointest Surg

    (2013)
  • G. Delle Fave et al.

    ENETS Consensus Guidelines for the management of patients with gastroduodenal neoplasms

    Neuroendocrinology

    (2012)
  • A. Rinke et al.

    Placebo-controlled, double-blind, prospective, randomized study on the effect of octreotide LAR in the control of tumor growth in patients with metastatic neuroendocrine midgut tumors: a report from the PROMID Study Group

    J Clin Oncol

    (2009)
  • E.P. Krenning et al.

    Somatostatin receptor scintigraphy with [111In-DTPA-D-Phe1]- and [123I-Tyr3]-octreotide: the Rotterdam experience with more than 1000 patients

    Eur J Nucl Med

    (1993)
  • D.J. Kwekkeboom et al.

    Somatostatin-receptor-based imaging and therapy of gastroenteropancreatic neuroendocrine tumors

    Endocr Relat Cancer

    (2010)
  • F.S. Dahdaleh et al.

    The value of preoperative imaging in small bowel neuroendocrine tumors

    Ann Surg Oncol

    (2013)
  • Cited by (18)

    • Small Bowel Neuroendocrine Tumors

      2020, Current Problems in Surgery
      Citation Excerpt :

      Somatostatin analogues (SSAs) have been used for decades to control NET hormonal symptoms.108 Small bowel and other NETs express high levels of somatostatin receptors,109-112 and activation of these receptors by synthetic somatostatin peptide mimetics decreases signaling in cell proliferation pathways while reducing hormone secretion.113 First-line SSA treatment improves hormonal symptoms in 70% to 80% of patients.114

    View all citing articles on Scopus

    Presented at the 9th Annual Academic Surgical Congress; February 4, 2014; San Diego, California.

    View full text