Associate editor: G.J. Sanger
Potassium-competitive acid blockade: A new therapeutic strategy in acid-related diseases

https://doi.org/10.1016/j.pharmthera.2005.05.005Get rights and content

Abstract

Current therapies to treat gastroesophageal reflux disease (GERD), peptic ulcer disease (PUD), and other acid-related diseases either prevent stimulation of the parietal cell (H2 receptor antagonists, H2RAs) or inhibit gastric H+,K+-ATPase (e.g., proton pump inhibitors, PPIs). Of the 2 approaches, the inhibition of the final step in acid production by PPIs provides more effective relief of symptoms and healing. Despite the documented efficacy of the PPIs, therapeutic doses have a gradual onset of effect and do not provide complete symptom relief in all patients. There is scope for further improvements in acid suppressive therapy to maximize healing and offer more complete symptom relief. It is unlikely that cholecystokinin2 (CCK2, gastrin) receptor antagonists, a class in clinical trials, will be superior to H2RAs or PPIs. However, a new class of acid suppressant, the potassium-competitive acid blockers (P-CABs), is undergoing clinical trials in GERD and other acid-related diseases. These drugs block gastric H+,K+-ATPase by reversible and K+-competitive ionic binding. After oral doses, P-CABs rapidly achieve high plasma concentrations and have linear, dose-dependent pharmacokinetics. The pharmacodynamic properties reflect the pharmacokinetics of this group (i.e., the effect on acid secretion is correlated with plasma concentrations). These agents dose dependently inhibit gastric acid secretion with a fast onset of action and have similar effects after single and repeated doses (i.e., full effect from the first dose). Animal studies comparing P-CABs with PPIs suggest some important pharmacodynamic differences (e.g., faster and better control of 24-hr intragastric acidity). Studies in humans comparing PPIs with P-CABs will help to define the place of this new class in the management of acid-related diseases.

Introduction

By the 19th century, gastric acid was known to be important in many physiological processes, such as protein digestion and the absorption of calcium and iron. Alongside these functions, sterilization of food is now also recognized as a vital function of gastric acid. Over time, awareness of its central role in the etiology of peptic ulcer disease (PUD), and more recently, gastroesophageal reflux disease (GERD), has also grown. This knowledge, together with an increased understanding of the physiology of acid production, directed the search for therapies in these diseases through inhibition of gastric acid secretion. However, until just under 30 years ago, patients with acid-related diseases had to rely on antacids, atropine, and other interventions (e.g., behavioral) for the relief of symptoms. The treatment of PUD was challenging and, apart from surgery, employed largely ineffective approaches that included a bland diet, antacids, and anticholinergics. Although antacids, and other approaches such as dietary changes and sucralfate, provide a degree of relief from the symptoms of GERD, healing of erosions was very difficult to achieve except with high and frequent doses of antacids (Grove et al., 1985).

The emergence of H2 receptor antagonists (H2RAs) in the 1970s represented the first major advance in the treatment of GERD and PUD, as they provided better symptom control and allowed higher healing rates compared with antacids. However, these agents have a relatively short duration of action, their effect is diminished by meal-stimulated secretion, and tolerance to their antisecretory effect can develop. Other approaches used to treat GERD (e.g., prokinetics, sucralfate) have generally not met expectations.

The first proton pump inhibitor (PPI), omeprazole, was approved for use in 1988 (Lindberg et al., 1990) and was the forerunner of a more effective class of agents than H2RAs. PPIs provide superior symptom relief and achieve higher healing rates in GERD and PUD than do H2RAs (Chiba et al., 1997, Van Pinxteren et al., 2001, Salas et al., 2002). Despite the undoubted efficacy of PPIs, there are still areas in which they could be improved upon (e.g., faster and better symptom control and more rapid healing). The quest for better therapy has driven research into other acid-suppressive treatments. This review examines the development and properties of current and potential treatments to suppress acid production, with particular emphasis on the potassium-competitive acid blocker (P-CAB) class.

Section snippets

Physiology of acid secretion

Hydrochloric acid (HCl) is secreted into the lumen of the stomach by parietal cells in the glands of the oxyntic mucosa. Gastric H+,K+-ATPase is fundamental to this process of acid secretion. This enzyme, located in the apical membrane of the parietal cell, transports H+ into the parietal cell canaliculus in exchange for K+. The secretion of H+ is accompanied by the passage of Cl across the apical membrane into the canaliculus, which ensures that acid secretion is electroneutral. For each H+

Structure and properties of gastric H+,K+-ATPase

The gastric H+,K+-ATPase is an α/β heterodimer, with each subunit having distinct functions. The α subunit, which has 10 helical transmembrane segments (M1–M10), is responsible for the catalytic activity of the enzyme and contains sites for ATP binding, as well as the cation (K+ and H+) binding site. The latter binding site is located near the middle of the membrane domains in the α subunit (Munson et al., 2000). It appears to be formed by amino acid residues from M4, M5, and M6, with the ion

Targeting gastric acid secretion

The treatment of peptic ulcer disease (PUD) has been based on Karl Schwartz's dictum of ‘no acid, no ulcer’. Although recent advances in our understanding have highlighted the multifactorial pathogenesis of both PUD and GERD, gastric acid is still recognized as a central component in both diseases. There is a correlation between healing of GERD lesions and the proportion of time (over 24 hr) when intragastric pH is greater than 4 (Bell et al., 1992). Furthermore, the period during which

Development of the potassium-competitive acid blocker class

During acid secretion, the surface of gastric H+,K+-ATPase faces the extremely acidic parietal cell canaliculus with a high affinity for K+. Given the importance of the cation for enzyme function, agents that compete with the binding of K+ have the potential to block acid secretion. It is this principle that underlies the mode of action of P-CABs (previously known as acid pump antagonists or APAs). These agents can be classified as imidazopyridines (e.g., SCH28080, AZD0865, and BY841),

Summary

The evolution of our understanding of the biochemistry and physiology of gastric acid secretion has led to the development of therapies to inhibit gastric acid secretion. PPIs are currently recognized to be the most effective available agents for the treatment of acid-related diseases. They offer superior symptom control and healing rates compared with H2RAs in both PUD and GERD. However, PPIs exhibit a delayed onset of acute effect and achieve full effect only slowly and incrementally over

References (127)

  • L.A. Dunbar et al.

    Ion pumps in polarized cells: sorting and regulation of the Na+, K+- and H+, K+-ATPases

    J Biol Chem

    (2001)
  • M. Eltze et al.

    Pharmacological evidence for selective inhibition of gastric acid secretion by telenzepine, a new antimuscarinic drug

    Eur J Pharmacol

    (1985)
  • S. Fiorucci et al.

    Gastric acidity and gastroesophageal reflux patterns in patients with esophagitis

    Gastroenterology

    (1992)
  • J.G. Forte et al.

    The membrane-recruitment-and-recycling hypothesis of gastric HCl secretion

    Trends Cell Biol

    (1996)
  • H. Fujisaki et al.

    Inhibitions of acid secretion by E3810 and omeprazole, and their reversal by glutathione

    Biochem Pharmacol

    (1991)
  • C. Gatto et al.

    Stabilization of the H,K-ATPase M5M6 membrane hairpin by K+ ions. Mechanistic significance for p2-type ATPases

    J Biol Chem

    (1999)
  • K. Gedda et al.

    Turnover of the gastric H+,K(+)-adenosine triphosphatase alpha subunit and its effect on inhibition of rat gastric acid secretion

    Gastroenterology

    (1995)
  • F. Grahammer et al.

    The cardiac K+ channel KCNQ1 is essential for gastric acid secretion

    Gastroenterology

    (2001)
  • S. Gustavsson et al.

    Rapid healing of duodenal ulcers with omeprazole: double-blind dose-comparative trial

    Lancet

    (1983)
  • W.B. Im et al.

    Irreversible inactivation of rat gastric (H+-K+)-ATPase in vivo by omeprazole

    Biochem Biophys Res Commun

    (1985)
  • W.B. Im et al.

    Reversal of antisecretory activity of omeprazole by sulfhydryl compounds in isolated rabbit gastric glands

    Biochim Biophys Acta

    (1985)
  • D.J. Keeling et al.

    SCH 28080 is a lumenally acting, K+-site inhibitor of the gastric (H+ + K+)-ATPase

    Biochem Pharmacol

    (1988)
  • D.J. Keeling et al.

    The binding of a K+ competitive ligand, 2-methyl,8-(phenylmethoxy)imidazo(1,2-a)pyridine 3-acetonitrile, to the gastric (H+ + K+)-ATPase

    J Biol Chem

    (1989)
  • D.J. Keeling et al.

    SK&F 96067 is a reversible, lumenally acting inhibitor of the gastric (H+ + K+)-ATPase

    Biochem Pharmacol

    (1991)
  • J.B. Koenderink et al.

    A conformation-specific interhelical salt bridge in the K+ binding site of gastric H,K-ATPase

    J Biol Chem

    (2004)
  • L. Lachman et al.

    Twenty-four-hour intragastric pH: tolerance within 5 days of continuous ranitidine administration

    Am J Gastroenterol

    (2000)
  • N. Lambrecht et al.

    Comparison of covalent with reversible inhibitor binding sites of the gastric H, K-ATPase by site-directed mutagenesis

    J Biol Chem

    (2000)
  • M. Maeda et al.

    Human gastric (H+ + K+)-ATPase gene. Similarity to (Na+ + K+)-ATPase genes in exon/intron organization but difference in control region

    J Biol Chem

    (1990)
  • F. Makovec et al.

    Characterization of antisecretory and antiulcer activity of CR 2945, a new potent and selective gastrin/CCK(B) receptor antagonist

    Eur J Pharmacol

    (1999)
  • J.P. Mattsson et al.

    Characterization of proton transport in bone-derived membrane vesicles

    Biochim Biophys Acta

    (1993)
  • J. Mendlein et al.

    Interaction of a K(+)-competitive inhibitor, a substituted imidazo[1,2a] pyridine, with the phospho- and dephosphoenzyme forms of H+,K(+)-ATPase

    J Biol Chem

    (1990)
  • N. Miura et al.

    Pharmacological profiles of Z-360, a novel CCK/gastrin (CCK2) receptor antagonist with excellent oral potency

    Gastroenterology

    (2001)
  • H. Morita et al.

    Effects of Z-360, a novel CCKB/gastrin (CCK2) receptor antagonist, on meal-induced acid secretion and experimental ulcer models in dogs and rats

    Gastroenterology

    (2001)
  • K.B. Munson et al.

    Identification of an extracytoplasmic region of H+,K(+)-ATPase labeled by a K(+)-competitive photoaffinity inhibitor

    J Biol Chem

    (1991)
  • M.E. Parsons et al.

    Properties of the reversible, K(+)-competitive inhibitor of the gastric (H+/K+)-ATPase, SK&F 97574: II. Pharmacological properties

    Biochem Pharmacol

    (1995)
  • A.J. Pope et al.

    Reversible inhibitors of the gastric H+/K(+)-transporting ATPase: a new class of anti-secretory agent

    Trends Pharmacol Sci

    (1993)
  • A.J. Pope et al.

    Properties of the reversible K(+)-competitive inhibitor of the gastric (H+/K+)-ATPase, SK&F 97574: I. In vitro activity

    Biochem Pharmacol

    (1995)
  • C. Prinz et al.

    Histamine secretion from rat enterochromaffinlike cells

    Gastroenterology

    (1993)
  • E.C. Rabon et al.

    Rubidium occlusion within tryptic peptides of the H,K-ATPase

    J Biol Chem

    (1993)
  • K. Andersson et al.

    Preclinical profile of AZD0865, a novel, selective, potassium-competitive acid blocker

    Gastroenterology

    (2004)
  • Andersson, K., Hallberg, H., & Holstein, B. (in press). AZD0865, a potassium-competitive acid blocker (P-CAB), does not...
  • W. Beil et al.

    Mechanism of gastric antisecretory effect of SCH 28080

    Br J Pharmacol

    (1986)
  • V. Beisvag et al.

    Identification and regulation of the gastric H+/K+-ATPase in the rat heart

    Acta Physiol Scand

    (2003)
  • N.J. Bell et al.

    Appropriate acid suppression for the management of gastro-oesophageal reflux disease

    Digestion

    (1992)
  • J. Beltinger et al.

    Effects of spiroglumide, a gastrin receptor antagonist, on acid secretion in humans

    Eur J Clin Invest

    (1999)
  • M. Besancon et al.

    Membrane topology and omeprazole labeling of the gastric H+, K(+)-adenosinetriphosphatase

    Biochemistry

    (1993)
  • J. Black et al.

    Gastrin agonists and antagonists

    Pharmacol Toxicol

    (2002)
  • C. Briving et al.

    Mechanism of action of AZD0865, an H+,K+-ATPase selective, potassium-competitive acid blocker

    Gastroenterology

    (2004)
  • C. Cederberg et al.

    Comparison of once-daily intravenous and oral omeprazole on pentagastrin-stimulated acid secretion in duodenal ulcer patients

    Digestion

    (1992)
  • J.A. Crawley et al.

    How satisfied are chronic heartburn suffers with their prescription medications? Results of the Patient Unmet Needs Survey

    J Clin Outcomes Manag

    (2000)
  • Cited by (154)

    • In Esophageal Squamous Cells From Eosinophilic Esophagitis Patients, Th2 Cytokines Increase Eotaxin-3 Secretion Through Effects on Intracellular Calcium and a Non-Gastric Proton Pump

      2021, Gastroenterology
      Citation Excerpt :

      Like PPIs, P-CABs also inhibit gastric and nongastric H+,K+ATPases, but through a different biochemical mechanism than PPIs.27,28 P-CABs disable H+,K+ATPase by binding ionically, rather than covalently, to the enzyme’s high-affinity K+ site, thereby blocking the K+ binding required to pump hydrogen ions (protons) out of the cell in exchange for K+ ions.46 Although commercial development of SCH 28080 was halted due to its hepatotoxicity, the P-CABs revaprazan and vonoprazan are used clinically in Asian countries,47 and vonoprazan has been shown to improve symptoms and esophageal eosinophilia in Japanese patients with EoE who were refractory to PPIs.48

    View all citing articles on Scopus
    View full text