Elsevier

Surgical Oncology

Volume 21, Issue 4, December 2012, Pages 281-292
Surgical Oncology

Review
Cholecystokinin and gastrin receptors targeting in gastrointestinal cancer

https://doi.org/10.1016/j.suronc.2012.06.004Get rights and content

Abstract

Cholecystokinin and Gastrin are amongst the first gastrointestinal hormone discovered. In addition to classical actions (contraction of gallbladder, growth and secretion in the stomach and pancreas), these also act as growth stimulants for gastrointestinal malignancies and cell lines. Growth of these tumours is inhibited by antagonists of the cholecystokinin and gastrin receptors. These receptors provides most promising approach in clinical oncology and several specific radiolabelled ligands have been synthesized for specific tumour targeting and therapy of tumours overexpressing these receptors. Therefore, definition of the molecular structure of the receptor involved in the autocrine/paracrine loop may contribute to novel therapies for gastrointestinal cancer. Hence, this review tries to focus on the role and distribution of these hormones and their receptors in gastrointestinal cancer with a brief talk about the clinical trial using available agonist and antagonist in gastrointestinal cancers.

Introduction

Gastrointestinal/gut hormones are chemical messengers that exist in multiple molecular forms and bind to multiple cell-surface receptors which are coupled to one of the several possible signal transduction systems. Thus activation of these receptors by their cognitive ligand leads diverse biologic responses and regulates a broad range of physiologic functions. Although these are primarily expressed within the tissues of gut, these peptide hormones are widely distributed throughout the body and act on multiple target tissues [1]. These also regulate gastrointestinal homeostasis by affecting cell proliferation, differentiation, apoptosis and gene expression. The aberrant control of these biological processes is thought to play an important role in the establishment of gastrointestinal neoplasia [2].

Since gut peptides receptors are (over)expressed on cancer cells, these represent the basis for receptor-targeted tumour imaging in nuclear medicine with radiolabeled peptides as well as growth factor and growth factor receptor alterations may provide new potential targets for the cancer therapy [3], [4]. For e.g., cholectstokinin type-B receptor/gastrin receptor (CCK2R/GR) ligands allow for a sensitive and reliable staging of patients with metastatic medullary thyroid cancer (MTC) and thus adding in diagnosis and therapy of these tumours expressing CCK2R/GR [5]. As a consequence several radiolabeled cholecystokinin (CCK) or gastrin (Gs) analogues/derivatives have been formulated and use of these analogues for diagnostic imaging and possible therapy of cancers that overexpress these receptors in patients using in vivo CCK receptor scintigraphy are being explored proving the feasibility of targeting CCK receptors in human tumours [6], [7], [8], [9]. Thus knowing the exact contribution and type of these receptor in carcinogenesis will allow the development of noncytotoxic modalities therapy with hormones, antihormones or hormone ablation [10], [11].

Section snippets

CCK, Gs hormone and its receptor

CCK and Gs both belong to one classical family of gastrointestinal peptides that regulate a variety of functions in the gastrointestinal tract and central nervous system. Both the peptides share a common C-terminal pentapeptide sequence (Gly-Trp-Met-Asp-PheNH2) (Fig. 1) but have different biological roles. Gs is produced by G cell; endocrine cells located in the gastric antrum. It is the major stimulant of gastric acid secretion and has a growth stimulatory effect on the stomach, exocrine

CCK Gs, its receptor and cancer

Gs or CCK showed growth stimulatory effect on cancer cell lines originating from the biliary tract [19]. CCK enhanced carcinogens mediated induction of acinar tumours in the pancreas. Gs is a central growth factor encouraging malignancies of the gastrointestinal tract, MTC, small cell lung cancer as well as tumours of the central and peripheral nervous systems [20], [21], [22]. It stimulates the survival or proliferation of normal cells along with gastric, colorectal or pancreatic cancer cells

CCK, Gs and its receptor in gallbladder cancer (GBC)

CCK is the chief humoural modulator of gallbladder that interacts with CCK1R and elicits the contraction of gallbladder by activating post-membrane signalling passage in smooth muscle [64]. In humans, the release of CCK is correlated intimately with contraction of the gallbladder [65] and CCK1R were demonstrated on smooth muscle cells of gallbladder [66]. Abnormality in the CCK-gallbladder relationship in comparison with normal subjects [67], [68] and reduced or delayed postprandial gallbladder

CCK, Gs and its receptor in stomach carcinoma (Sc)

Knockouts studies for Gs gene or CCK2R/GR gene has confirmed that Gs is well known growth factor for enterochromaffin-like cells (ECL) in the stomach determining the fate of ECL cells [36], [81], [82], [83]. Long-standing condition of hypergastrenemia were associated with ECL cell hyperplasia and ultimately to malignant carcinoids transformation [84], [85], [86], [87]. It is also regarded as an important biomarker for gastric inflammation [88]. Gs and CCK2R/GR was also found to play an

CCK, Gs and its receptor in pancreatic carcinoma

CCK and Gs was found to increase pancreatic DNA, RNA and protein content [13], [14], [106], [107], [108] resulting in increased pancreatic size and weight [109]. Experimental hypercholecystokininemia stimulate pancreatic growth and induced pancreatic hypertrophy, hyperplasia, and dysplasia thus enhancing normal, preneoplastic and malignant pancreatic growth [110] and this effect was mediated by CCK1R [111].

CCK is the most potent peptide influencing growth of human pancreatic cancers [112], [113]

CCK, Gs and its receptor in esophageal carcinoma

Endogenous hypergastrenemia was found to promote chemically induced esophageal carcinogenesis [144]. CCK2R/GR has been also identified in both lower and mid esophageal mucosa [145] and in human esophageal adenocarcinoma (OAC). Gs binding to these receptors increases proliferation of cells in a dose-dependent way and this effect are abolished by CCK2R/GR specific antagonist [146]. However, there was no effect on those lacking CCK2R/GR. Thus CCK2R/GR seems to be involved in the development of OAC

CCK, Gs and its receptor in colon carcinoma

The role of CCK, Gs and their receptors in the pathogenesis of colon cancer has been discussed for many years but it still remains a disputable issue. Both in vivo and in vitro studies have asserted Gs as a growth stimulus for normal colonic mucosa as well as colon carcinoma in a receptor-mediated fashion. The first evidence for autocrine growth stimulatory role of Gs/CCK-like peptide in cultured colon tumour cells was provided by Hoosein et al., (1990) [58], though some studies have reported

Peptide-receptor targeting in cancer

In law, cell surface receptors overexpressed on tumour cells are of raising clinical importance, as it furnishes targets for anticancer drugs linked to receptor ligands [177]. Moreover, during the past decade, the specific receptor binding property of the ligand/peptide have been tapped by labelling the ligand with a radionuclide and using these radiolabeled receptor-binding ligand as a vehicle to guide the radioactivity to the tissues expressing a particular receptor, in vivo. These

Targeting Cck1r and Cck2r in gastrointestinal cancer

Cholecystokinin receptor is a novel and promising candidate in the field of receptor-targeted tumour imaging and a means of delivering CCK-receptor-mediated cytotoxic agents for anticancer drugs therapy [6]. Over the past few years the increased concern: (1) to potential therapeutic denotations of the tumour growth effect of gastrin and CCK, (2) possibility of using the CCK2R and CCK1R over/expression for tumour imaging or therapy has pipelined several body for the search of CCK/Gs receptor

Clinical trials

Compounds directed at the CCK2R/GR have shown promising results in clinical trials in humans. Initial randomized, controlled study using proglumide, the GR/CCK2R antagonist, as therapy in patients with gastric carcinoma failed to show an overall effect on survival. Since porglumide has relatively low affinity with the CCK2R/GR and also has partial agonist activity, further clinical trials of more specific and potent GR antagonists is required which may have a greater effect on survival to

Conclusion

CCK1R expression was found in bulk of GBC, esophageal and pancreatic cancer while CCK2R/GR was found in majority of SC, pancreatic and colon carcinoma. These affirms that Gs and CCK wield a trophic effect on normal gut mucosa and can also act in autocrine, endocrine or paracrine mode to regulate growth of some cancers of the GI tract and pancreas through cell specific receptor [207]. The availability of an adequately specific, sensitive and high affinity radioligand fulfils the successful

Funding

This study was supported by the grant from Indian Council of Medical Research (ICMR Project Ref No. 3/2/2/187/2009/NCD-III).

Conflict of interest statement

The authors declare that there are no conflict of interests.

Authorship statement

Guarantor of the integrity of the study: Hari S. Shukla.

Study concepts: Hari S. Shukla.

Study design: Rajani Rai.

Definition of intellectual content: Hari S. Shukla, Mallika Tewari, Rajani Rai.

Literature research: Rajani Rai, Vishal Chandra, Mallika Tewari, Mohan Kumar.

Data acquisition: Rajani Rai, Vishal Chandra.

Data analysis: Rajani Rai, Mallika Tewari.

Manuscript preparation: Mallika Tewari, Rajani Rai, Vishal Chandra.

Manuscript editing: Rajani Rai, Mallika Tewari, Hari S. Shukla.

Manuscript

References (208)

  • A.M. Grabowska et al.

    Role of gastrin peptides in carcinogenesis

    Cancer Lett

    (2007)
  • R. Moonka et al.

    Cholecystokinin-A receptor messenger RNA expression in human pancreatic cancer

    J Gastrointest Surg

    (1999)
  • Z.J. Cheng et al.

    Heterodimerization of type A and B cholecystokinin receptors enhance signaling and promote cell growth

    J Biol Chem

    (2003)
  • M.R. Hellmich et al.

    Human colorectal cancers express a constitutively active cholecystokinin-B/gastrin receptor that stimulates cell growth

    J Biol Chem

    (2000)
  • Z.J. Cheng et al.

    Analysis of the cellular and molecular mechanisms of trophic action of a misspliced form of the type B cholecystokinin receptor present in colon and pancreatic cancer

    Cancer Lett

    (2005)
  • C. Sanchez et al.

    Characterization of a novel five-transmembrane domain cholecystokinin-2receptor splice variant identified in human tumors

    Mol Cell Endocrinol

    (2012)
  • N. Iwata et al.

    Autocrine loop through cholecystokinin-B/gastrin receptors involved in growth of human leukemia cells

    Blood

    (1996)
  • N. Remy-Heintz et al.

    Evidence for autocrine growth stimulation by a gastrin/CCK-like peptide of the gastric cancer HGT-1 cell line

    Mol Cell Endocrinol

    (1993)
  • N.M. Hoosein et al.

    Evidence for autocrine growth stimulation of cultured colon tumor cells by a gastrin/cholecystokinin-like peptide

    Exp Cell Res

    (1990)
  • J.C. Reubi et al.

    Distribution of CCK1 and CCK2 receptors in normal and diseased human pancreatic tissue

    Gastroenterology

    (2003)
  • P. Yu et al.

    Cholecystokinin-coupled intracellular signaling in human gallbladder muscle

    Gastroenterology

    (1994)
  • S. Tierney et al.

    Physiology and pathophysiology of gallbladder motility

    Surg Clin North Am

    (1993)
  • J. Lamote et al.

    Effect of cholecystokinin-octapeptide, caerulein, and pentagastrin on epithelial cell proliferation in the murine gallbladder

    Gastroenterology

    (1982)
  • R. Rai et al.

    Expression profile of cholecystokinin type-A receptor in gallbladder cancer and gallstone disease

    Hepatobiliary Pancreat Dis Int

    (2011)
  • N. Langhans et al.

    Abnormal gastric histology and decreased acid production in cholecystokinin-B/gastrin receptor-deficient mice

    Gastroenterology

    (1997)
  • D. Chen et al.

    Differentiation of gastric ECL cells is altered in CCK(2) receptor-deficient mice

    Gastroenterology

    (2002)
  • T.J. Koh et al.

    Gastrin deficiency results in altered gastric differentiation and decreased colonic proliferation in mice

    Gastroenterology

    (1997)
  • I.M. Modlin et al.

    The gastric enterochromaffin-like cell: an enigmatic cellular link

    Gastroenterology

    (1996)
  • P.L. Peghini et al.

    Effect of chronic hypergastrinemia on human enterochromaffin-like cells: insights from patients with sporadic gastrinomas

    Gastroenterology

    (2002)
  • N. Okada et al.

    Evaluation of cholecystokinin, gastrin, CCK-A receptor, and CCK-B/gastrin receptor gene expressions in gastric cancer

    Cancer Lett

    (1996)
  • R.J. Bold et al.

    Biomolecular advances in gastrointestinal hormones

    Arch Surg

    (1993)
  • K. Hur et al.

    Expression of gastrin and its receptor in human gastric cancer tissues

    J Cancer Res Clin Oncol

    (2006)
  • M. Béhé et al.

    Cholecystokinin-B (CCK-B)/gastrin receptor targeting peptides for staging and therapy of medullary thyroid cancer and other CCK-B receptor expressing malignancies

    Biopolymers

    (2002)
  • J.C. Reubi

    Targeting CCK receptors in human cancers

    Curr Top Med Chem

    (2007)
  • J.K. Sosabowski et al.

    Targeting of CCK-2 receptor-expressing tumors using a radiolabeled divalent gastrin peptide

    J Nucl Med

    (2009)
  • J.C. Reubi et al.

    Peptide-based probes for cancer imaging

    J Nucl Med

    (2008)
  • R.J. Bold et al.

    Progress toward hormonal therapy of gastrointestinal cancer

    Ann Surg

    (1996)
  • E.E. Cawston et al.

    Therapeutic potential for novel drugs targeting the type 1 cholecystokinin receptor

    Br J Pharmacol

    (2010)
  • T.E. Solomon et al.

    Interaction of caerulein and secretin on pancreatic size and composition in rat

    Am J Physiol

    (1978)
  • D.L. Mainz et al.

    Hormonal control of pancreatic growth

    J Clin Invest

    (1973)
  • S.A. Wank et al.

    Cholecystokinin receptor family. Molecular cloning, structure, and functional expression in rat, guinea pig, and human

    Ann N Y Acad Sci

    (1994)
  • S.A. Wank

    Cholecystokinin receptors

    Am J Physiol

    (1995)
  • L.J. Miller et al.

    Biochemical and cell biological mechanisms of cholecystokinin receptor regulation

    Curr Top Med Chem

    (2007)
  • J.R. Upp et al.

    Clinical significance of gastrin receptors in human colon cancers

    Cancer Res

    (1989)
  • J. Ishizuka et al.

    The effect of gastrin on growth of human stomach cancer cells

    Ann Surg

    (1992)
  • S. Watson et al.

    Gastrin: growth enhancing effects on human gastric and colonic tumour cells

    Br J Cancer

    (1989(b))
  • S.A. Watson et al.

    Growth-promoting action of gastrin on human colonic and gastric tumour cells cultured in vitro

    Br J Surg

    (1988)
  • N. Kobayashi et al.

    Z-360, a novel cholecystokinin-2/gastrin receptor antagonist, inhibits gemcitabine-induced expression of the vascular endothelial growth factor gene in human pancreatic cancer cells

    Biol Pharm Bull

    (2010)
  • M.N. Lango et al.

    Gastrin-releasing peptide receptor-mediated autocrine growth in squamous cell carcinoma of the head and neck

    J Natl Cancer Inst

    (2002)
  • D.F. McWilliams et al.

    Coexpression of gastrin and gastrin receptors (CCK-B and delta CCK-B) in gastrointestinal tumour cell lines

    Gut

    (1998)
  • Cited by (15)

    • Changes in plasma levels of cholecystokinin, neurotensin, VIP and PYY in gastric and colorectal cancer – Preliminary results

      2019, Peptides
      Citation Excerpt :

      Impaired release of CCK or mutations of the CCK receptors may contribute to pancreatic pathologies (e.g. acute/chronic pancreatitis), impairment of gastrointestinal motility (e.g. irritable bowel syndrome) and hypergastrinemia-related disorders (e.g. hyperplasia, carcinoid) [8]. CCK promotes development of gastrointestinal malignancies, acting via CCK1 and CCK2 receptors expressed by the tumor cells and via an autocrine/paracrine loop [4,7–10]. CCK receptors belong to the family with seven transmembrane segments.

    • Immunogenicity and safety of a novel tetanus toxoid-conjugated anti-gastrin vaccine in BALB/c mice

      2018, Vaccine
      Citation Excerpt :

      The signal transduction pathways posited for the promotion of tumours by gastrin-17 and progastrin are both numerous and complex [14–16], but targeting them has become an investigative strategy for the treatment of related GI tumours by anti-gastrin therapies, which specifically include endocrine-secretion inhibitors, receptor antagonism, and anti-gastrin antibody methods. Endocrine-secretion inhibition (e.g., by somatostatin [17] and antisense oligonucleotides [18]) and receptor antagonism (e.g., by cholecystokinin receptor antagonists [19–21]) are methods that have often had low anti-gastrin specificity, weak anti-tumour effects, and necessarily large dosages, with a significant number of toxic side effects. Anti-gastrin antibody methods include both passive and active immunizations.

    • Gastrin and Gastric Cancer

      2017, Cellular and Molecular Gastroenterology and Hepatology
      Citation Excerpt :

      Because gastrin has been shown to stimulate growth of gastric cancer and other gastrointestinal malignancies, researchers have been studying means to block the ligand:receptor interaction in an attempt to slow or arrest tumor growth. Numerous investigations have been conducted in cell culture and animal models of gastric cancer using small-molecule CCK-B receptor antagonists,91,92 and their use in human trials has been reviewed.76,93,94 Clinical trials in human subjects with agents directed to the gastrin:CCK-B–receptor pathway are rare and most studies have investigated agents in other gastrointestinal cancers rather than in gastric cancer, such as gastrazole in pancreatic cancer95 or netazepide96 in gastric neuroendocrine tumors.

    View all citing articles on Scopus
    View full text