You are on page 1of 37

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/7082742

Acid Suppression Therapy: Where Do We Go from Here?

Article in Digestive Diseases · February 2006


DOI: 10.1159/000091298 · Source: PubMed

CITATIONS READS
73 1,055

3 authors, including:

Carmelo Scarpignato Francesco Di Mario


Università di Parma Iuliu Haţieganu University of Medicine and Pharmacy
464 PUBLICATIONS 7,108 CITATIONS 783 PUBLICATIONS 10,311 CITATIONS

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

SEROLOGY AS A FIRST-LINE DIAGNOSTIC TOOL FOR HELICOBACTER PYLORI INFECTION AND ASSOCIATED ACUTE INFLAMMATION OF THE STOMACH: POSSIBILITY OR
REALITY? A RETROSPECTIVE STUDY ON 524 PATIENTS. View project

Study of environnemental radioprotection using RESRAD-Biota code in Center, Cameroon View project

All content following this page was uploaded by Carmelo Scarpignato on 04 January 2015.

The user has requested enhancement of the downloaded file.


Review Article

Dig Dis 2006;24:11-46


DOI: 10.1159/000091298

Acid Suppression Therapy:


Where Do We Go from Here?
Carmelo Scarpignato a Iva Pelosini a Francesco Di Mario b
a
Laboratory of Clinical Pharmacology, Department of Anatomy, Pharmacology and Forensic Sciences, and
b
Department of Clinical Sciences, Section of Gastroenterology, School of Medicine and Dentistry,
University of Parma, Parma, Italy

Key Words of them (namely potassium-competitive acid blockers


Gastric acid secretion  Acid inhibition  Antisecretory (P-CABs) and CCK2-receptor antagonists) have already
drugs  H2-receptor antagonists  Ebrotidine  reached clinical testing while some others (like the anti-
Lafutidine  Proton pump inhibitors  Ilaprazole- gastrin vaccine, H3-receptor ligands or gastrin-releasing
Tenatoprazole  Potassium-competitive acid blockers  peptide receptor antagonists) are still in preclinical de-
CCK2-receptor antagonists  Antigastrin vaccine velopment and need the proof of concept in human be-
ings. Of the current approaches to reduce acid secretion,
P-CABs and CCK2-receptor antagonists hold the greatest
Abstract promise, with several compounds already in clinical tri-
The dramatic success of pharmacological acid suppres- als. Although the quick onset of action of P-CABs (i.e. a
sion in healing peptic ulcers and managing patients with full effect from the first dose) is appealing, the results of
gastroesophageal reflux disease (GERD) has been re- phase II studies with one such agent (namely AZD0865)
flected in the virtual abolition of elective surgery for ulcer did not show any advantages over esomeprazole. Thanks
disease, a reduction in nonsteroidal anti-inflammatory to their limited efficacy and the development of tolerance
drug (NSAID)-associated gastropathy and the decision it is unlikely that CCK2 antagonists will be used alone as
by most patients with reflux symptoms to continue med- antisecretory compounds but, rather, their combination
ical therapy rather than undergo surgical intervention. with PPIs will be attempted with the aim of reducing the
However, a number of challenges remain in the manage- long-term consequences of hypergastrinemia. While H2-
ment of acid-related disorders. These include manage- receptor antagonists (especially soluble or over-the-
ment of patients with gastroesophageal symptoms who counter formulations) will become the ‘antacids of the
do not respond adequately to proton pump inhibitor third millennium’ and will be particularly useful for on-
(PPI) therapy, treatment of patients with nonvariceal up- demand symptom relief, clinicians will continue to rely
per gastrointestinal bleeding, prevention of stress-relat- on PPIs to control acid secretion in GERD and other acid-
ed mucosal bleeding, optimal treatment and prevention related diseases. In this connection, several new PPI for-
of NSAID-related gastrointestinal injury, and optimal mulations have been developed and two novel drugs
combination of antisecretory and antibiotic therapy for (namely ilaprazole and tenatoprazole) are being studied
the eradication of Helicobacter pylori infection. A num- in humans. The recently introduced immediate-release
ber of new drugs are currently being investigated to pro- (IR) omeprazole formulation (currently available only in
vide a significant advance on current treatments. Some the USA) quickly increases intragastric pH and, given at

© 2006 S. Karger AG, Basel Prof. Carmelo Scarpignato, MD, DSc, PharmD, FRCP, FCP, FACG
0257–2753/06/0242–0011$23.50/0 Laboratory of Clinical Pharmacology
Fax +41 61 306 12 34 School of Medicine and Dentistry, University of Parma
E-Mail karger@karger.ch Accessible online at: Via Volturno, 39, IT–43100 Parma (Italy)
www.karger.com www.karger.com/ddi Tel. +39 0521 903 863, Fax +1 603 843 5621, E-Mail scarpi@tin.it

DDI923.indd 11 02.05.2006 16:23:33


bedtime, seems to achieve a better control of nocturnal
acidity. IR formulations of other PPIs (including the in-
vestigational ones) will probably be available in the fu-
ture and will enlarge our therapeutic armamentarium.
Amongst the novel PPIs, tenatoprazole appears to be a
true advance in the acid suppression therapy. Its long
half-life (the longest among the available compounds)
and longer duration of antisecretory action, with no dif-
ference between day and night, will allow the drug to go
beyond the intrinsic limitations of currently available
PPIs. Thanks to its favorable pharmacokinetics, the so-
dium salt of S-tenatoprazole is being developed and the
preliminary results indicate that this drug has the poten-
tial to address unmet clinical needs. Although some de-
cades have elapsed since the introduction of effective
and safe antisecretory drugs in clinical practice and their Fig. 1. Chemical structure of currently used PPIs.
use has stood the test of time, the ongoing research will
further provide the clinician with more effective means
of controlling acid secretion.
Copyright © 2006 S. Karger AG, Basel proton pump (H+,K+-ATPase) to secrete hydrogen ions
into the gastric lumen in exchange for potassium ions [1].
The recognition that H+,K+-ATPase was the final step of
Introduction acid secretion culminated in the development of a class
of drugs, the proton pump inhibitors (PPIs), which are
Gastric acid secretion is under nervous and hormonal targeted at inhibiting this enzyme [2]. They represent one
influence [1]. This physiologic process is controlled by a of the most commonly prescribed classes of drugs in both
number of redundant second messenger pathways acti- gastroenterological and primary care settings and are con-
vated as a result of the binding of gastrin, acetylcholine, sidered a major advance in the treatment of acid-related
histamine, and prostaglandins to the specific receptors on diseases.
the basolateral surface of parietal cells. The stimulatory After sufficient acid secretion has occurred, a feedback
effect of acetylcholine and gastrin is mediated by an in- system terminates gastric acid secretion. A decrease of
crease in cytosolic calcium, whereas that of histamine is intragastric pH stimulates somatostatin (SST) release
mediated by activation of adenylate cyclase and genera- from antral D cells. This peptide not only inhibits acid
tion of cyclic AMP (cAMP). Strong potentiation between secretion but also blunts gastrin release and its stimula-
histamine and either gastrin or acetylcholine reflects post- tory effect on CCK2 receptors located on parietal and
receptor interaction between the distinct pathways as well ECL cells [1, 4]. Furthermore, acidification of the duode-
as the ability of acetylcholine and gastrin to release hista- num triggers secretin release, which also inhibits gastric
mine from mucosal enterochromaffin-like (ECL) cells [1]. acid secretion [5]. The inhibitory effects of SST and en-
Indeed, gastrin, the major circulating stimulus of acid se- dogenous prostaglandins (EPs) on acid secretion are me-
cretion, probably does not stimulate the parietal cells di- diated by receptors (SST2 and EP3, respectively) coupled
rectly but acts to mobilize histamine from the ECL cells by guanine nucleotide-binding proteins to inhibition of
in the oxyntic mucosa [2]. The gastrin-ECL cell pathway adenylate cyclase activity. All the pathways (both stimu-
has been investigated extensively in situ (gastric submu- latory and inhibitory) converge on and modulate the ac-
cosal microdialysis), in vitro (isolated ECL cells) and in tivity of the luminal enzyme (i.e. H+,K+-ATPase) [1, 2].
vivo (intact animals). Gastrin acts on CCK2 receptors to In 1973, pyridylmethyl benzimidazole sulfides were
control the synthesis of ECL cell histamine, accelerating originally discovered to be active PPIs: they were then
the expression of the histamine-forming enzyme, histi- modified to the corresponding sulfoxide timoprazole, pi-
dine decarboxylase (HDC), at both the transcription and coprazole and omeprazole in 1979. Omeprazole, the first
the translation/posttranslation levels [3]. The ultimate clinically available PPI, has been used in some countries
factor in acid secretion, however, is the stimulation of the for more than two decades [6]. More recently, other mem-

12 Dig Dis 2006;24:11–46 Scarpignato /Pelosini /Di Mario


Chiral
R3 R3
R4 R2 sulfoxide R4 R2

N N+

S + SOH
HN O H N NH
N

Canalicular
Pro-drug Sulfenic acid Diffusion
lumen
Parietal
R1 R1 Concentration
−H2O cell
Cytosol Conversion
R3 R3 Inhibition
R4 R2 R4 R2
Secretory
Achiral, membrane
N+ N+ optically
S S E S inactive
N NH E SH N N

R1 R1
Inactivated enzyme, Cyclic sulfenamide
fairly stable

Fig. 2. Chemical activation of PPIs within the parietal cell (modified from Kromer [9]).

bers of the PPI family including lansoprazole, pantopra- species interacts with the external surface of the H+,K+-
zole rabeprazole and esomeprazole have been developed ATPase that faces the lumen of the secretory space of the
and launched. parietal cell, resulting in disulfide bond formation with
one or more key cysteines located within the -subunit of
the enzyme; this is the residue that is intimately involved
Inhibition of Acid Secretion: Where Are We? in hydrogen ion transport. This covalent inhibition of the
enzyme by the thiophilic sulfenamide results in a specific
Chemistry and Pharmacology of PPIs and long-lasting impairment of gastric acid secretion. The
Chemically, all the available PPIs [7, 8] consist of a selectivity of PPIs for the parietal cell proton pump stems
benzimidazole ring and a pyridine ring, but vary in the from the fact that H+,K+-ATPase is the only pump in the
specific side ring substitution (fig. 1). They are all weak body that generates a sufficiently steep proton gradient of
protonatable pyridines, with a pKa of about 4.0 for more than 1:1,000,000, corresponding to pH 0.8–1.0 in-
omeprazole and lansoprazole, about 3.9 for pantoprazole, side the canaliculus of a secreting parietal cell [10].
and about 5.0 for rabeprazole. As a result, they accumu- PPIs are the most potent inhibitors of gastric acid se-
late specifically and selectively in the secretory canalicu- cretion available [8, 10]. They are most effective when
lus, the highly acidic space of the parietal cell [8, 9]. With- the parietal cell is stimulated to secrete acid postprandi-
in that space, PPIs undergo an acid-catalyzed conversion ally, a relationship that has important clinical implica-
to a reactive species, the thiophilic sulfenamides, which tions for timing of administration. Because the amount
are permanent cations (fig. 2). The rate of conversion var- of H+,K+-ATPase present in the parietal cell is greatest
ies among the compounds and is inversely proportional after a prolonged fast, PPIs should be administered before
to their pKa: rabeprazole (4.71) 1 omeprazole (4.09) 1 the first meal of the day. In most individuals, once-daily
lansoprazole (3.92) 1 pantoprazole (3.89). The reactive dosing is sufficient to produce the desired level of acid

Update on Acid Suppression Therapy Dig Dis 2006;24:11–46 13

DDI923.indd 13 02.05.2
inhibition, and a second dose, which is occasionally nec- Raising intragastric pH with PPIs improves antimi-
essary, should be administered before the evening meal crobial efficacy of chemotherapeutic agents towards H.
[11]. pylori through several mechanisms [15], thus increasing
During meals, neither all parietal cells nor all proton their bactericidal effectiveness. As a consequence, the
pumps are active. Since PPIs inhibit only activated en- combination of a PPI and two antimicrobials (mainly
zyme present in the canalicular membrane, the reduction clarithromycin and amoxicillin or metronidazole) is now
of gastric acid secretion after an initial dose will probably a well-established first-line regimen [16, 17], whenever
be suboptimal. As inactive enzyme is recruited into the eradication of the microorganism is indicated [18, 19].
secretory canaliculus, acid secretion will resume, albeit at Since Warren and Marshall first described the infectious
a reduced level. After the second dose is given on the next etiology of PU disease in 1984 [20, 21], a great deal of
day, more H+,K+-ATPase will have been recruited and evidence has accumulated to suggest that H. pylori erad-
subsequently inhibited, and after the third dose, addi- ication therapy cures PU disease [22–24] and can be ben-
tional recruitment and further acid inhibition will prob- eficial also to other H. pylori-related diseases [25]. A re-
ably occur [8, 11]. cent meta-analysis [26] clearly showed that prolonging
Once-daily PPI dosing inhibits maximal acid output therapy with PPIs after a PPI-based triple therapy for 7
by about 66% after 5 days. Thus, the occasional use of a days is not necessary to induce ulcer healing.
PPI taken on an ‘as needed’ basis would not be expected Both oral and intravenous PPIs have allowed for the
to reliably provide adequate acid inhibition and would first time a complete control over the marked acid hyper-
be unlikely to produce a consistent or satisfactory clinical secretion often seen in patients with ZES [27, 28] and are
response (in contrast to the H2-antagonists, which have a indeed the drugs of choice in the long-term management
more rapid onset of action) [8, 11]. Because of the delay of ZES patients. PPIs are effective even in patients un-
in optimal acid inhibition, the initial use of twice-daily responsive to H2-RAs [29] and do not show tachyphy-
dosing (for the first 2–3 days) may be helpful in achieving laxis.
more rapid inhibition of gastric acid secretion. In addi- In patients with reflux esophagitis the degree of muco-
tion to their delay in onset, and because PPI-derived sulf- sal healing is directly related to the proportion of time
enamides bind covalently to H+,K+-ATPase, the restora- during the 24-hour period for which the intragastric pH
tion of acid secretion will likewise be delayed, depending is maintained above 4 [30, 31]. As a consequence, numer-
upon enzyme turnover and the biological reversibility of ous studies have documented the marked efficacy of PPIs
the disulfide bond. Maximal acid secretory capacity may in controlling symptoms of GERD and healing esophagi-
not be restored for 24–48 h after discontinuing PPIs [8, tis and a large meta-analysis [32] clearly showed a distinct
11]. advantage – either in terms of healing and symptom re-
lief – of PPIs over H2-RAs, a finding confirmed in a re-
Clinical Applications of PPIs cent Cochrane Review [33].
PPIs are effective for treatment of all acid-related dis- Gastroduodenal mucosa possesses an array of defen-
orders, i.e. peptic ulcer (PU) and eradication of Helico- sive mechanisms and NSAIDs have a deleterious effect
bacter pylori, Zollinger-Ellison syndrome (ZES), gastro- on most of them. This results in a mucosa less able to cope
esophageal reflux disease (GERD) and its complications, with even a reduced acid load. The presence of acid ap-
nonsteroidal anti-inflammatory drugs (NSAID)-associ- pears to be a conditio sine qua non for NSAID injury,
ated gastroduodenal ulcers as well as PU bleeding. which is in fact pH-dependent. There is therefore a strong
In a landmark study, Burget et al. [12] found a highly rationale for PPI use in both treatment and prevention of
significant correlation between healing and the degree of NSAID-associated gastroduodenal ulcers [34]. Unlike
acid suppression, the duration of acid suppression, and H2-blockers, PPIs protect from NSAID injury not only
the length of therapy. The shape of the contour expression the duodenum, but also the stomach, where the majority
of these relationships showed that healing rate increases of mucosal lesions are usually located [35–38].
as the duration of suppression increases and as gastric pH The goal of medical therapy for bleeding ulcers has
increases. Being more potent and longer lasting than H2- been traditionally to sustain intragastric pH 16, in order
RAs, PPIs are therefore more effective in healing both to promote platelet aggregation, clot formation and stabil-
gastric and duodenal ulcers. And indeed, several meta- ity [39, 40]. H2-RAs show little benefit in patients with
analyses [13, 14] did show that these drugs are able to heal PU bleeding [41–43], which reflects their inadequate pH
gastroduodenal ulcer more rapidly than H2-RAs. control and the rapid onset of tolerance (see below). Sev-

14 Dig Dis 2006;24:11–46 Scarpignato /Pelosini /Di Mario

DDI923.indd 14 02.05.2006 16:24:52


eral meta-analyses [41, 42, 44–46] have shown that treat- Despite all the above limitations, there is still a place
ment with a PPI reduces the risk of re-bleeding and the for H2-RAs in the era of PPIs [55]. Besides the treatment
requirement for surgery after ulcer bleeding but has no of functional dyspepsia, where they are superior to pla-
benefit on overall mortality, an effect seen only with in- cebo [59], H2-RAs are useful for heartburn relief in pa-
travenous administration of high doses to high-risk pa- tients with mild forms of GERD [60], being better and
tients [47, 48]. One of these systematic reviews [48] actu- faster than antacids [61]. OTC (low-dose) formulations
ally suggested a significant benefit only for patients having are not only able to decrease gastric and esophageal acid-
endoscopic high-risk stigmata for re-bleeding. ity but actually potentiate the antacid effect in relieving
PPI therapy for ulcer bleeding has been more effica- meal-induced heartburn [62].
cious in Asia than elsewhere [49]. This may be because of The appreciation that even twice-daily PPIs may not
an enhanced pharmacodynamic effect of PPIs in Asian adequately control intragastric acidity during the night
patients [50, 51]. Combination of endotherapy with acid and that in a significant proportion of both healthy sub-
suppression is superior to monotherapy in reducing bleed- jects and GERD patients a ‘nocturnal acid breakthrough’
ing and surgery and superior to endotherapy alone in (NAB) does occur [63] has suggested the use of a bedtime
minimizing re-bleeding but not surgery [52]. dose of H2-RAs to improve acid control [64]. However,
Treatment of H. pylori infection is more effective than long-term use of these agents led to development of toler-
antisecretory noneradicating therapy (with or without ance so that their effect on NAB lessened with prolonged
long-term maintenance antisecretory therapy) in prevent- therapy [65]. In addition, it must be emphasized that no
ing recurrent bleeding from PU [53]. Recurrence of infec- study in patients with GERD has yet demonstrated that
tion seems to be responsible for recurrence of bleeding. addition of H2-RAs to twice-daily PPI therapy provides
Consequently, all patients with PU bleeding should be any further benefit above that derived from PPIs alone
tested for H. pylori infection, and eradication therapy be [66, 67]. However, a paper presented at a recent DDW in
prescribed to H. pylori-positive patients. Orlando [68] showed that, besides reducing NAB [64],
ranitidine, given at bedtime even at low doses, signifi-
Why Are PPIs Better Antisecretory Agents than cantly improves nocturnal heartburn in GERD patients
H2-RAs? on PPI therapy, already on the first day of therapy. Al-
H2-RAs have a relatively short duration of action and, though the benefit lasted only a few days, these data sug-
depending on the individual agent and whether the pa- gest that short-term or intermittent therapy with H2-RAs
tient is in a fed or fasting state, suppress acid for approx- might have a significant impact also on GERD-related
imately 4–8 h [54]. Consequently, multiple daily doses of symptoms.
these agents are likely to be required. Furthermore, H2- Up to 79% of patients with GERD experience noctur-
RAs produce incomplete inhibition of postprandial gas- nal symptoms associated with their condition. Improve-
tric acid secretion. Overall, these agents inhibit acid se- ment in nocturnal symptoms associated with reduction
cretion by up to 70% over a 24-hour period [54, 55]. in gastric acidity by PPIs is well documented [69]. The
A further shortcoming is that tolerance to standard clinical importance of specifically reducing NAB, how-
H2-RAs generally develops within 2 weeks of repeated ever, is controversial. In patients with GERD, recovery
administration, resulting in a decline in acid suppression of nocturnal gastric acid secretion is of little importance,
[56]. This can be explained by a gastrin-induced increase if it is not accompanied by exposure of the esophagus to
in ECL-derived histamine concentrations at the H2-re- acid. Nevertheless, patients with severe forms of chronic
ceptor on the parietal cell [57] and up-regulation of both GERD, especially those with Barrett’s esophagus, are
gastrin and H2-receptors [58]. In contrast, PPIs control more likely to have acid reflux during NAB [70]. A recent
both basal and food-stimulated acid secretion and pro- report [71] has shown 170% of patients had improved
duce more complete and longer lasting acid suppression nighttime symptoms after addition of an H2-RA to a
than H2-blockers [8, 11]. Such acid inhibition virtually twice-daily PPI regimen, thus emphasizing the clinical
abolishes the damaging peptic activity of gastric juice. In impact of controlling NAB.
addition, tolerance to PPIs has not been observed, an ad- All the above findings suggest that a hierarchy of over-
vantage presumably attributable to the fact that they act night pH control does exist [72], beginning with a PPI
at the final step of acid production, thereby blocking the given in the morning, adding an H2-RA at bedtime, pro-
effects of any compensatory mechanisms promoting acid gressing to PPI therapy twice daily and then again adding
secretion [11, 57]. an H2-RA at bedtime (table 1).

Update on Acid Suppression Therapy Dig Dis 2006;24:11–46 15

DDI923.indd 15 02.05.2006 16:24:52


Table 1. Relative efficacy of antisecretory regimens (from Katz and tic disease, and on the effectiveness and duration of acid
Tutuian [72]) suppression. Refractoriness occurs less often with PPIs
than with H2-RAs as the former decrease acid more ef-
Efficacy Therapeutic regimens
fectively [78]. H2-RA refractory disease usually responds
Highest PPIs twice daily with H2-RAs at bedtime to treatment with PPIs. However, although rare, refrac-
PPIs twice daily toriness to PPIs does exist [79–81], amounting to 5–10%

d
Lowest
PPIs in the morning with H2-RAs at bedtime
PPIs once daily
High-dose H2-RAs
Over-the-counter H2-RAs
in GERD and 2–8% in PU [78, 81].
Potential reasons for this less than optimal response
can be found when carefully examining the intragastric
pH profiles of patients with PPI-resistant PU [82] or
PPIs should always be given before a meal. GERD [83, 84]. In both cases, tracings will frequently
The administration of current DR PPIs is recommended 0.5– show that PPIs fail to adequately control intragastric acid-
1.0 h prior to a meal in order to ensure that proton pumps are ac-
ity, especially during the night, although other pathophys-
tivated in the parietal cell at a time when drugs are available in
plasma. Since PPIs all have similar plasma half-lives of 1–2 h, any iological features should be taken into account. Increasing
proton pump synthesized after drug levels fall will not be blocked the PPI dose will often offset this ‘relative’ resistance. If
from secreting acid. This kind of administration therefore allows NAB occurs in GERD patients with predominantly night-
the maximal antisecretory effect of the PPI dose and also mini- time symptoms, administering an H2-RA at bedtime
mizes any possibility of interaction with food.
would be worthwhile [70, 71], although the benefit of this
combination may be short-lasting [65]. In any event, the
reported failure of PPIs to effectively control either intra-
gastric (and intraesophageal) pH and symptoms clearly
Are Currently Available PPIs All the Same? unmask some limitations of currently available agents.
Although there are differences among PPIs concerning It should be mentioned, however, that there is a small
their pharmacokinetics, pharmacodynamics, influence group of patients who are ‘true’ omeprazole-resistant (de-
by food and antacids as well as potential for drug interac- fined as gastric pH !4 for 50% of 24 h), for whom an ab-
tions [7, 73], it is not always evident whether these often normality of the proton pump, due to mutations of cys-
subtle differences are clinically relevant. Several compre- teine 813 and 822, has been suggested [85]. Whether re-
hensive analyses of the available clinical trials concluded sistance to other PPIs exists has not been currently
that – when used at equivalent doses in the treatment of studied. In addition, although successful treatment with
various acid-related disorders – all the available agents PPIs in H2-RA-resistant GERD patients is the rule, few
are similarly effective [7, 74, 75]. These conclusions con- patients who showed therapeutic response to high-dose
cerning ‘clinical’ efficacy of PPIs are backed by similar H2-RAs after failure of high-dose omeprazole to control
results obtained in comparative ‘pharmacological’ analy- gastric acidity and gastroesophageal reflux have been re-
sis [76]. ported [86].
Guidance from the National Institute for Clinical Ex-
cellence (NICE) does not differentiate between PPIs ex- Shortcomings and Limitations of Current PPIs
cept on the grounds of price and accepted indications Although effective and safe, currently available PPIs
[77]. The physician’s choice of one PPI over another must are still far from the ideal antisecretory compound. An
therefore rest with her/his interpretation of the clinical ideal drug should allow full acid control around the clock
importance of the generally small differences among and dose-dependent and predictable pharmacokinetic
PPIs, their approval for treatment of specific clinical in- and pharmacodynamic properties. These properties and
dications within the physician’s practice jurisdiction, and the consequent clinical effects should also be reproduc-
the strength of the evidence based on the quantity and ible in a wide range of patient populations [87]. Finally,
quality of the supporting clinical trials [74]. an ideal agent will display high oral bioavailability, a rap-
id onset of action and few, if any, clinically relevant in-
Is There Any Acid Peptic Disease That Is Refractory teractions with food or concomitantly administered drugs
to PPIs? [88].
Nonhealing and/or delayed healing during acid inhibi- The so-called first-generation PPIs (omeprazole, pan-
tion treatment depend on the extent to which acid and toprazole and lansoprazole) have notable limitations.
‘non-acid’ factors are causative in the particular acid pep- These drugs exhibit substantial interpatient variability in

16 Dig Dis 2006;24:11–46 Scarpignato /Pelosini /Di Mario

DDI923.indd 16 02.05.2006 16:24:52


pharmacokinetics or may have significant interactions PPI [93]. Although these drugs also target H+,K+-ATPase,
with other drugs. The time of dosing and ingestion of they act via a distinct mechanism: they bind the extracy-
meals may also influence the pharmacokinetics of these toplasmatic surface and act as reversible K+-competitive
agents as well as their ability to suppress gastric acid se- antagonists [93, 94].
cretion. First-generation PPIs also have a relatively slow
onset of pharmacological action and may require several From Omeprazole to Esomeprazole:
doses to achieve maximum acid suppression and symp- Why a Chiral Switch?
tom relief, possibly limiting their usefulness in on-de- Rationalization within the pharmaceutical industry to
mand GERD therapy. First-generation PPIs may also fail combat escalating costs has included the close examina-
to provide 24-hour suppression of gastric acid, and NAB tion of research portfolios. Gastroenterology has been one
can occur even with twice-daily dosing [88]. of the casualties of this exercise and few companies cur-
Taking all the above considerations into account, it is rently retain a specific gastrointestinal research program
clear that there is room for improvement and a new gen- [95]. AstraZeneca, who discovered the first PPI more
eration of PPIs might be able to overcome the limitations than 20 years ago, synthesized and started the develop-
of first-generation compounds [89]. To obtain an ideal ment of esomeprazole (the magnesium salt of the S-iso-
PPI capable of achieving full inhibition of acid control mer of omeprazole) in 1990, making it available for clin-
with the first dose, different avenues can be pursued. One ical practice worldwide at the beginning of the third mil-
way is to develop a compound whose favorable metabo- lennium.
lism would allow an enhanced delivery of the active form Taking into account that omeprazole comprises a ra-
to the proton pump with consequent better inhibition of cemic mixture of its two optical isomers, S-omeprazole
acid secretion. Exploiting the stereospecific catabolism of and R-omeprazole, and that S-omeprazole is optically
enantiomers follows that way. stable in humans with negligible inversion to the other
The extension of plasma half-life is a critical factor in isomer [96], the R&D team decided to follow the first of
improving the onset of action of PPIs and thus com- the avenues outlined above in order to obtain a PPI with
pounds with a half-life longer than that of current PPIs improved acid control. By following the methodology
may be expected to display first-day efficacy. Such an av- (the so-called ‘Sharpless Epoxidation’) discovered by
enue can be pursued through the synthesis of new chem- K. Barry Sharpless, one of the three scientists who re-
ical entities with a molecular structure different from the ceived the 2001 Nobel Prize in Chemistry [97], they set
current (i.e. substituted benzimidazole) one or designing up the stereoselective synthesis of esomeprazole [98] and
a prodrug that will release the PPI in the bloodstream were able to produce the isomer on a large scale.
[90]. Tenatoprazole, consisting of one imidazopyridine Since all the currently available PPIs are racemic mix-
ring connected to a pyridine ring by a sulfinylmethyl tures, the stereoisomers of other molecules (namely lan-
chain [91], represents an example of a new chemical en- soprazole, pantoprazole, rabeprazole and the new non-
tity with an extended half-life (about 8 h compared to benzyimidazole compound, tenatoprazole) have been
1.0–1.5 h), whereas compound D [90] is an investiga- isolated [99] and are being extensively investigated.
tional pro-PPI for very stable intravenous formulation
that displays a longer duration of action in comparison The Importance of Chirality in Pharmacology
with omeprazole. Some prodrugs synthesized recently hy- Chiral molecules were one of the great discoveries of
drolyze under physiological conditions (pH = 7.4) to pro- Louis Pasteur, who observed in the mid-1800s that two
vide PPIs with a half-life largely exceeding the 2 h and distinct crystal forms of tartaric acid would rotate polar-
are capable of providing sustained plasma concentration ized light in opposite directions. Pasteur correctly postu-
of the released PPI for a time longer than the currently lated that the two crystal forms were enantiomers, or
used compounds [92]. right- and left-handed mirror images of each other. The
The development of potassium-competitive acid Greek name kheir means ‘hand’. So, chirality indeed
blockers (P-CABs, previously called acid pump antago- means ‘handedness’. Enantiomers can be thought of as
nists, APAs) is another active area of research in control- left and right hands: although they are also mirror images
ling intragastric pH. P-CABs such as the imidazopyri- of each other, a left hand, for example, does not fit into a
dines have thus far shown fast first-day onset as well as right-handed glove. Following Louis Pasteur’s discover-
improved control of day- and nighttime intragastric pH ies, the name was coined by the Irish physicist William
with twice-daily dosing when compared to a once-daily Thomson, alias Lord Kelvin.

Update on Acid Suppression Therapy Dig Dis 2006;24:11–46 17

DDI923.indd 17 02.05.2006 16:24:53


Isomers

Structural isomers Stereoisomers


(different bonding and structure) (structurally the same but
different 3-dimensional arrangement)

Optical isomers or enantiomers Diastereoisomers


(not superimposable – mirror images) and geometric isomers

S-isomer R-isomer
Fig. 3. Different types of chemical iso-
mers.

Chirality is central to life, as many of the important and excretion) and quantitatively or qualitatively differ-
molecules come in two mirror-image forms that have very ent pharmacologic or toxicologic effects [103–105]. Ste-
different properties [100]. The fundamental molecules of reoisomers include not only the mirror-image enantio-
life, namely DNA and proteins, are, in fact, composed mers, but also geometric (cis/trans) isomers and diaste-
respectively of right- and left-handed subunits only. Na- reoisomers (isomers of drugs with more than one chiral
ture often discriminates between chiral forms of small center that are not mirror images of one another, fig. 3).
molecules, too, and what is a medicine on the one hand Diastereoisomers and geometric isomers are both chem-
can be a poison on the other. One example of this is tha- ically distinct and pharmacologically different (unless
lidomide. Thalidomide, which was given to pregnant they are interconverted in vivo) and are generally readily
women as a sedative and antinausea drug in the 1950s in separated without chiral techniques. Geometric isomers
Europe, turned out to produce debilitating birth defects and diastereoisomers therefore should, with the rare ex-
(i.e. missing limbs or phocomelia) [101]. But, in fact, only ception of cases where in vivo interconversion occurs, be
one enantiomer (i.e. the S-isomer) of the molecule is re- treated as separate drugs and developed accordingly
sponsible for these effects [102]. [100].
Stereoisomers are molecules that are identical in atom- When stereoisomers are biologically distinguishable,
ic constitution and bonding, but differ in the three-di- they might seem to be different drugs [103–105], yet it
mensional arrangement of the atoms. The stereoisomeric has been past practice to develop racemates (i.e., com-
pairs of greatest interest are those with one or more asym- pounds with 50:50 proportion of enantiomers). The prop-
metric (chiral) centers whose enantiomers (individual ste- erties of the individual enantiomers have – in the past –
reoisomers) are mirror images. According to an absolute not generally been well studied or characterized. Wheth-
convention, known as the Sequence Rule notation, when er separated enantiomers should be developed was
the peripheral groups are dimensionally arranged clock- largely an academic question because commercial separa-
wise around the chiral center, the R (rectus) configuration tion of racemates was difficult. Now that technological
is assigned to the molecule. On the contrary, if they are advances (large-scale chiral separation procedures or
arranged counterclockwise around the chiral center, the S asymmetric syntheses) permit production of many single
(sinister) configuration is allocated. enantiomers on a commercial scale, it is appropriate to
Optical isomers have essentially identical physical (ex- consider the development of the single enantiomer char-
cept for optical rotatory) and chemical (except in a chiral acterized by the most favorable profile of activity (euto-
environment) properties. Such stereoisomers usually re- mer) [104, 105]. Any decision to develop a drug as a sin-
quire specialized chiral techniques for their correct iden- gle enantiomer, however, should be made only after care-
tification, characterization, separation and measurement. ful evaluation of the cost-benefit ratio, i.e. when the
They are often readily distinguished by biological sys- advantages of the eutomer in terms of efficacy and toler-
tems, however, and may have different pharmacokinetic ability outweigh the associated increase in production
properties (absorption, distribution, biotransformation and development costs with respect to the racemic drug

18 Dig Dis 2006;24:11–46 Scarpignato /Pelosini /Di Mario

DDI923.indd 18 02.05.2006 16:24:53


[106]. Development of racemates raises issues of accept- azoles is to a larger or lesser extent (e.g. for rabeprazole)
able manufacturing control of synthesis and impurities, dependent on hepatic cytochrome P450 (CYP) isoforms,
adequate pharmacologic and toxicologic assessment, CYP3A4 and CYP2C19 [7, 73, 116]. These heme-con-
proper characterization of metabolism and distribution, taining enzymes are abundantly expressed in hepato-
and appropriate clinical evaluation [107]. cytes, where they exist in the smooth endoplasmic reticu-
From a theoretical standpoint, some stereochemical lum in association with a number of accessory proteins.
aspects of drug therapy should be taken into account [108, There, the cytochromes P450 catalyze the multistep oxida-
109]. For chiral drugs, one of the entantiomers can: tion reactions that inactivate xenobiotics by the intro-
• be inactive (e.g. S-loxiglumide) or less active (e.g. duction or exposure of a functional group on their sub-
R-warfarin), strate. The dependence of most PPIs on metabolism by
• display a quantitatively different pharmacological ef- CYP2C19 makes them susceptible to alterations in bio-
fect. The presence of the inactive or less active isomer availability due to genetic polymorphisms affecting the
is often responsible for the greater variability observed activity of this enzyme [73, 117]. In particular, omepra-
with racemates, zole is transformed into three pharmacologically inactive
• have a different metabolic pattern, metabolites, i.e. omeprazole sulfone and 5-hydroxy- plus
• have an antagonistic effect, 5-O-desmethyl-omeprazole, respectively. Studies per-
• or be, actually, toxic. formed in healthy volunteers [118], human liver micro-
Since no drug is completely devoid of adverse effects, somes and cDNA-expressed enzymes [119] have shown
recent interest has focused on the role of the different a significant stereoselectivity in the metabolism of
properties of individual drug enantiomers in causing drug omeprazole enantiomers. The R-isomer is almost exclu-
toxicity [110]. Several examples of stereoselective toxic- sively metabolized (98%) via CYP2C19, 94% transform-
ity are available in the literature. Vomiting, for instance, ing to the 5-hydroxy metabolite, and 4% to the 5-O-des-
is caused merely by the R-isomer of levamisole [111]. methyl metabolite. Only 2% of the metabolism is via the
Myasthenia gravis symptoms were no longer observed CYP3A4 isoform. The S-isomer (esomeprazole) is me-
when the R-isomer was removed from the racemic mix- tabolized primarily (73%) via the CYP2C19 isoform
ture of carnitine [112]. Only the S-isomer has been linked (46% going to the 5-O-desmethyl metabolite, 27% go-
to the teratogenic effects of thalidomide [102]. ing to the 5-hydroxy metabolite), and by 27% via the
CYP3A4 isoform to the sulfone metabolite. The intrin-
Development of Esomeprazole sic clearance (CL) of S-omeprazole is approximately one
In accordance with results of clinical trials in acid-re- third of that of R-omeprazole, because of the considerably
lated diseases [7, 74], where often different but somewhat lower CL for formation of the 5-hydroxy metabolite (15
equipotent doses of the different PPIs have been used, vs. 43 l  min/mg protein) via CYP2C19 [120]. This im-
data from experimental pharmacological studies clearly plies that the total metabolic clearance for S-omeprazole
show that – on a milligram basis – the available com- is lower than that for R-omeprazole, resulting in higher
pounds display the same potency and efficacy [76]. Since plasma levels of the S-isomer in vivo (fig. 4). These data
both R- and S-isomer of omeprazole are able to inhibit suggest that, in vivo, S-omeprazole should undergo a low-
acid secretion in isolated gastric glands to the same degree er first-pass effect thus leading to higher plasma levels and
[113], there should be no pharmacodynamic benefit in consequent better availability. Since the area under time-
using one stereoisomer instead of the racemate. Both en- concentration curve correlates with secretory inhibition
antiomers (which are chiral sulfoxides) will give rise – [121], the S-isomer should be capable of achieving a bet-
within the secretory canaliculus – to the same reactive ter acid control compared with the racemic mixture (i.e.
species, sulfenamide, which is achiral (fig. 2). There is omeprazole). The available in vivo pharmacokinetic,
therefore no pharmacodynamic reason why the racemate pharmacodynamic and clinical data all show that this is
or any of its enantiomers should differentially interact the case [122–125].
with the proton pump [9]. The stereoselective metabolism of lansoprazole [126–
Although the use of a PPI enantiomer does not confer 128], pantoprazole [129, 130] and rabeprazole [131] has
any direct advantage in vitro, the pharmacokinetic (and also been studied and consistently revealed significant
pharmacodynamic) consequences of stereoselective drug quantitative differences between isomers. The stereoiso-
metabolism might anyway affect drug activity in vivo mers of some of these PPIs are being extensively studied.
[114, 115]. The metabolism of substituted benzimid- In particular a new modified release formulation of the

Update on Acid Suppression Therapy Dig Dis 2006;24:11–46 19

DDI923.indd 19 02.05.2006 16:24:53


Chiral switching is a rational but undesirable response
1,600 of manufacturers to patent expiration. When patents on
1,400 Omeprazole commercially successful drugs approach their expiry
dates, the development of a stereoisomer enables compa-
Concentration (nmol/l)

S-omeprazole
1,200
R-omeprazole
nies to maintain market share and high prices despite
1,000
generic competition. Indeed, the zealously guarded pat-
800
ent monopoly system provides the same exclusivity and
600
market protection to an enantiomer of an existing prod-
400 uct as it does to a new chemical entity. This makes the
200 development and marketing of stereoisomers of existing
0 racemates more commercially attractive than the high
0 2 4 6 costs, uncertainties and risks of developing genuine in-
Time (h)
novative compounds [141]. It would be desirable and in
some ways ethical to explore from the very beginning the
Fig. 4. Plasma concentration vs. time at steady state (day 7) of
pharmacokinetics and pharmacodynamics of enantio-
omeprazole, S-omeprazole and R-omeprazole in four extensive mers and start the development of the more attractive
metabolizers after intake of 15 mg of each compound (from Ander- isomer (eutomer) right away.
sson et al. [120]).

Inhibition of Acid Secretion:


Where Are We Going?
R-isomer of lansoprazole (compound marked TAK-
390MR) is now in phase III development [132] and 7 Unmet Needs in Acid Suppression
clinical trials in GERD (registered at www.clinicaltrials. The dramatic success of pharmacological acid sup-
gov) are ongoing in the USA [133]. Actually, the Food pression in healing ulcers and managing patients with
and Drug Administration (FDA) granted the permission GERD has been reflected in the virtual abolition of elec-
to move directly to phase III without going through phase tive surgery for ulcer disease [142], a reduction in NSAID-
II based on the results of the phase I studies. This R- associated gastropathy [143], and the decision by most
isomer was selected either because of its higher plasma patients with reflux symptoms to continue medical ther-
concentrations [126] and lower clearance [127] and be- apy rather than undergo surgical intervention.
cause – besides being the most potent inhibitor of However, a number of challenges remain in the man-
CYP2C19 enzyme – S-lansoprazole also inhibits CY2C9 agement of acid-related disorders. These include manage-
and CYP2D6 activities [134]. These properties make the ment of patients with gastroesophageal symptoms who do
S-isomer an unsuitable drug for the treatment of acid-re- not respond adequately to PPI therapy, treatment of pa-
lated diseases for its large drug interaction potential. By tients with nonvariceal upper gastrointestinal (GI) bleed-
the way, NSAIDs – amongst other drugs – represent one ing, prevention of stress-related mucosal bleeding, opti-
of the most important substrates of CYP2C9 [135]. Being mal treatment and prevention of NSAID-related GI in-
NSAID-associated symptoms and mucosal lesions are jury, and optimal combination of antisecretory and
one of the clinical indications of PPIs, the use of S-lanso- antibiotic therapy for the eradication of H. pylori infec-
prazole concomitantly with NSAIDs would need great tion [144, 145].
caution or be actually contraindicated. As outlined before, PPIs have a number of shortcom-
While chirality with the elegance of its underlying con- ings, which stem from their pharmacology. However,
cepts is certainly fascinating, the demonstration that chi- there is increasing evidence of both inappropriate pre-
ral switching provides superior clinical benefits (either in scribing and inappropriate use of these drugs. An expand-
terms of efficacy, safety or economy) is difficult. Although ing proportion of patients have indeed poor indication to
esomeprazole is considered by some [124, 125, 136] a true acid suppression. Not uncommonly, empiric therapy in
therapeutic advantage and a cost-effective treatment for nonexplored and functional dyspepsia is based upon PPI
acid-related diseases [137], others [138–140] have ques- therapy, standard or high dose for 2–4 weeks. If symptom
tioned its superiority over other PPIs, including omepra- resolution is limited and patient dissatisfaction contin-
zole. ues, often the dose is doubled. Upon sudden arrest of such

20 Dig Dis 2006;24:11–46 Scarpignato /Pelosini /Di Mario

DDI923.indd 20 02.05.2006 16:24:54


therapy because of insufficient symptom relief, rebound nologies (Vénissieux, France) and applied in cooperation
acid secretion may reactivate GERD-like/dyspeptic with TAP Pharmaceutical Products Inc. (Lake Forest, Ill.,
symptoms leading to a vicious circle [144]. There is a stag- USA) (a joint venture between two global pharmaceutical
gering lack of clear instructions in practice concerning leaders, Abbott and Takeda Pharmaceutical Co. Ltd) to
empirical therapy. That PPIs are often taken inappropri- lansoprazole [151]. This technology consists of a multi-
ately is revealed by a US survey [146] showing that only ple-dose system containing 5,000–10,000 microparticles
27% of GERD patients dosed their PPI correctly (i.e. up per capsule or tablet. The 200- to 500-m diameter-sized
to 60 min before any meal of the day) and only 9.7% dosed microparticles are released in the stomach and then pass
it optimally (i.e. 15–60 min before the first meal of the into the small intestine, where each microparticle, operat-
day). Furthermore, in a study of 1,046 primary care phy- ing as a miniature delivery system, releases the compound
sicians across the USA [147], only 36% of them did give by osmotic pressure at an adjustable rate and over an ex-
their patients advice on when and how to take their med- tended period of time. The design of the Micropump™
ication. microparticles allows an extended transit time in the
Although a proper education can improve PPI use and small intestine with a plasma mean residence time ex-
effectiveness, currently available drugs have their own tended up to 24 h. Thanks to the microparticles’ size,
shortcomings. To overcome these limitations, novel for- these formulations are easy to swallow, well tolerated and
mulations of currently available PPIs [92, 148] and nov- taste masking.
el PPIs [149] are being developed. In addition, new ave- Another proprietary controlled-release formulation
nues (i.e., competitive blockade of proton pump at K+ (the so-called ChroNAB delivery technology) has been
exchange sites [93] or blockade of CCK2, e.g. gastrin re- developed by AGI Therapeutics Ltd (Dublin, Ireland)
ceptors on the parietal and ECL cells [150]) are being ex- and applied to currently available PPIs in order to mod-
plored. ify their pharmacokinetics allowing for a better acid con-
In this section, the new PPI formulations currently trol, especially during the night when the NAB occurs
under development or recently introduced in clinical [152]. One such formulation, namely AGI 010, is cur-
practice as well as the novel acid-lowering drugs, which rently undergoing clinical testing.
have already reached clinical testing, will be discussed. Currently available PPIs are orally administered as
gastroprotected preparations. The different enteric coat-
New PPI Formulations ings, which are necessary to protect the acid-labile PPI
Some interesting new PPI formulations have recently from acid degradation within the stomach, have the po-
been patented by AstraZeneca [92]. An enteric-coated, tential disadvantage of delaying PPI absorption and, as a
extended-release dosage form of a PPI provided an ex- consequence, the available PPI formulations are consid-
tended plasma concentration profile of the drug for a pe- ered delayed-release (DR) preparations.
riod of up to 12 h. This pharmacokinetic profile is ob- DR oral dosage forms are supplied as enteric-coated
tained with a formulation comprising a core material of granules encapsulated in a gelatine shell (e.g. omeprazole
a hydrophilic or hydrophobic matrix containing the PPI capsules and lansoprazole capsules) or as enteric-coated
protected by enteric coating. An additional patent de- tablets (e.g. pantoprazole tablets, rabeprazole tablets and
scribes a multilayered tablet capable of releasing the PPI omeprazole multiple-unit pellet system [MUPS]). An al-
in discrete pulses, separated in time (2 pulses of drug re- ternative oral formulation of lansoprazole – the lansopra-
lease at least 0.5 h apart). The dosage form, which has one zole orally disintegrating tablet (ODT) – has recently been
fraction with instant release of the drug and at least an- developed and introduced into clinical practice. It is easy
other fraction with a pulsed delayed release, is intended to swallow and can be taken orally with or without water.
for once-daily administration [92]. The bioavailability This feature greatly improves compliance when patients
and pharmacokinetic profile of these formulations have are away from home (and water is not readily available)
not yet been studied in humans and it is presently un- and in those patients with odynophagia or dysphagia as
known whether they will assure a better and longer lasting well as swallowing disorders. In addition, when this for-
control of intragastric pH compared with the currently mulation is dispersed in water, it provides a less expen-
available PPI formulations. sive alternative to intravenous PPIs for patients with na-
A Micropump™ technology, which permits either ex- sogastric or gastric tubes in ICU or long-term care settings
tended or both delayed and extended delivery of drugs to [153]. Some studies (reviewed by Horn and Howden
the small intestine, has been developed by Flamel Tech- [154]) have evaluated PPI absorption from intact nonen-

Update on Acid Suppression Therapy Dig Dis 2006;24:11–46 21

DDI923.indd 21 02.05.2006 16:24:54


istration unit, be it a tablet or a granule-containing cap-
sule, is relatively large. The lansoprazole ODT contains
7 much smaller microgranules than lansoprazole capsules
(330 vs. 1,100 g). The tablet disintegrates rapidly (with-
4
3
in 30 s) in the mouth without water. Unlike the granules
1 in lansoprazole capsules, which have a core, a lanso-
2
5
prazole layer and a gastroresistant enteric coating, each
6 lansoprazole ODT microgranule comprises seven layers
(fig. 5). The active lansoprazole layer surrounds an inert
core, followed by an inert under the coating layer that
improves stability in high humidity. Three enteric-coat-
ing layers prevent dissolution in the stomach, improve
Fig. 5. Structure/composition of enteric-coated microgranules used
to make LFDT (thickness of each layer is not to scale). 1 = Inert
stability, reduce damage during compression and neutral-
core; 2 = lansoprazole layer; 3 = undercoating layer – improves sta- ize the taste of the microgranule. An outer layer increases
bility in high humidity; 4 = first enteric-coating layer – improves the hardness of the tablet [156, 157]. When lansoprazole
stability of lansoprazole; 5 = second enteric-coating layer – prevents ODT disintegrates in the mouth the microgranules are
breakage of the enteric coats during tablet compression; 6 = third swallowed with the patient’s saliva. The enteric-coated
enteric-coating layer – neutralizes taste of microgranule; 7 = over-
coating layer – improves tablet hardness (from Baldi and Malfert-
microgranules dissolve in the neutral condition of the
heiner [156]). small intestine and lansoprazole is absorbed into the
bloodstream. However, absorption kinetics still depends
on the enteric coating; the oral pharmacokinetics of lan-
soprazole after dosing with the ODT is essentially identi-
capsulated omeprazole or lansoprazole as well as their cal to that observed after dosing with capsules of enteric-
suspension in sodium bicarbonate (8.4%) solution. Al- coated granules [158]. The bioequivalence between these
though, with exception of the so-called simplified omepra- two formulations will translate into a similar pharmaco-
zole suspension (SOS) whose bioavailability was im- dynamics and clinical efficacy. And indeed, despite that
paired, the other ‘extemporary’ formulations allowed a the formulation was preferred by a higher proportion of
proper PPI absorption, predisintegrated lansoprazole patients, no significant difference in symptom relief be-
ODT would represent the most appropriate choice for tween lansoprazole ODT and other PPIs (namely esome-
administration via nasogastric or gastrostomy tube. prazole) was found in NERD [159].
Most PPI tablets (e.g. rabeprazole) are formulated as Conversely from lansoprazole ODT, the recently
an enteric-coated, single-unit system, that is, a tablet with FDA-approved immediate-release (IR) omeprazole for-
a gastric acid-resistant layer only on its outer surface. mulation displays a different pharmacokinetics and phar-
There is a risk that the enteric coat of such tablets can macodynamics compared with the standard, DR prepa-
become damaged during gastric emptying. As PPIs are ration [148]. This formulation consists of pure, nonen-
acid-labile, a small crack or damage to a single-unit sys- teric-coated omeprazole powder (40 or 20 mg per unit
tem may, therefore, affect the entire dose. Multiple-unit dose) along with 1,680 mg of sodium bicarbonate (con-
systems have been developed to overcome this problem taining 460 mg of sodium). It is flavored with peach and
[155]. Common formulations of multiple-unit systems in- peppermint and is designed be constituted with water to
clude both capsules and tables containing enteric-coated be drunk.
granules. The very small size and multiple coatings of Besides sachets, capsules containing 40 or 20 mg of
these granules mean that they are less likely to be dam- omeprazole are also available, while a chewable tablet
aged during gastric emptying, and absorption issues are formulation has been developed and an NDA (new
therefore minimized. If a granule is damaged and a crack drug application) already submitted to FDA [160]. It is
develops on the surface, only a small portion of active important to distinguish IR omeprazole (IR-OME) for-
substance is likely to be affected. The lansoprazole cap- mulation from the SOS (see above). While IR-OME has
sule and omeprazole MUPS are examples of multiple- no form of enteric coating, SOS is prepared by opening
unit systems. While enteric coating of oral formulations standard capsules of DR omeprazole (DR-OME), drop-
is beneficial for ensuring that PPI agents are effectively ping the granular contents into 8.4% sodium bicarbonate
delivered, it does mean that the individual dose admin- solution and agitating the mixture until the enteric coat-

22 Dig Dis 2006;24:11–46 Scarpignato /Pelosini /Di Mario

DDI923.indd 22 02.05.2006 16:24:54


900 8 IR-OME 40 mg
Omeprazole concentration (ng/ml)

IR-OME (n = 7)
800 DR-OME (n = 10) Pantoprazole 40 mg
7
700 Median pH 4.7 ✽
6
600

Gastric pH
5
500
400 4

300 3
Median pH 2.0
200 2
100 1
0
0
0 1 2 3 4 5 6
23:00 24:00 02:00 04:00 06:00
a Time post-dose (h)
Time (24-hour clock)

Dose Meal

8 Day 7 Fig. 7. Eight-hour nighttime period gastric pH on day 6 demon-


7
Baseline strated that once-daily dosing with IR-OME produced significantly
better nocturnal gastric acid control than once-daily DR pantopra-
6 zole in a crossover study in 32 GERD patients. * p ! 0.001 IR-OME
vs. pantoprazole (from Castell et al. [164]).
Gastric pH

2
teers. Within the first 30 min, DR-OME had no measur-
1
able activity on intragastric pH, whereas the IR formula-
0 tion caused an immediate increase in pH. From 4 to 6 h
0 3 6 9 12 15 18 21 24
b Time (h) after ingestion, there was a 27% reduction from baseline
in intragastric activity with DR-OME and 65% decrease
with the IR formulation (fig. 6) [162]. Therefore, the an-
Fig. 6. Pharmacokinetics and pharmacodynamics of IR-OME in tisecretory effect of IR-OME was quicker than that ob-
healthy volunteers. a Mean plasma concentration from fasting sub- served with the classical DR formulation while the dura-
jects with IR-OME (40 mg, n = 7) and DR capsules (40 mg, n = 10)
tion of the acid-lowering activity was similar. The early
(from Hepburn and Goldlust [161]). b Effect of 7 days of once-
daily dosing with the IR formulation (40 mg, n = 24) on 24-hour increase in intragastric pH is likely due to the neutralizing
intragastric pH (from Howden [162]). capacity of sodium bicarbonate, which also accelerates
and enhances absorption of omeprazole whose increased
bioavailability translates into a more profound acid sup-
pression.
ing of the granules disintegrates and a suspension formed. As already discussed, NAB is a common physiological
Due to the impaired absorption of omeprazole from this event, observed in approximately 70% of both healthy
extemporary preparation [154], a lower than expected an- subjects and GERD patients treated with up to twice-
tisecretory effect can be predicted. daily administration of DR PPIs, regardless of whether
The pharmacokinetics of IR-OME has been studied in the PPI is administered in the morning, prior to dinner,
healthy volunteers given 40 mg of both formulations in a or at bedtime [163]. Bedtime dosing with 40 mg of IR-
crossover fashion [161]. While the AUC was not signifi- OME suspension provides better control of nocturnal
cantly different, the Cmax was higher and the Tmax shorter gastric acidity than once-daily dosing with all DR PPIs
with the IR formulation (fig. 6). As with the DR capsules, (fig. 7) [164, 165] and also decreases NAB more effec-
both Cmax and AUC but not Tmax increased between 1 tively than with dosing of DR-OME 20 mg b.d. and lan-
and 7 days of once-daily dosing with IR-OME, indicating soprazole 30 mg b.d. or pantoprazole 40 mg b.d. Night-
an increase of bioavailability over time. time control of gastric acidity with IR-OME suspension
In a crossover trial, the effect of both formulations on is comparable with that achieved by twice-daily dosing of
intragastric acidity was investigated in healthy volun- DR esomeprazole 20 mg [164, 165].

Update on Acid Suppression Therapy Dig Dis 2006;24:11–46 23

DDI923.indd 23 02.05.2006 16:24:54


N NH CH3 H3CO NH CH3
OCH3 N OCH3
N S N S

a O N b O N
CH3

Ilaprazole (IY-81149) Tenatoprazole (TU-199)


2-(RS)[4-methoxy-3-methyl)-2-pyridinyl]methyl- (RS)-5-methoxy-2-[[(4-methoxy-3,5-dimethylpyrid-2-yl)
sulfinyl]-5-(1H-pyrol-1-yl)-1H-benzimidazole methyl]sulfinyl]-1H-imidazo[4,5b]pyridine
[CAS Registry No. 172152-36-2] [CAS Registry No. 113712-98-4]

Fig. 8. Chemical structure of ilaprazole (a) and tenatoprazole (b), two novel PPIs, currently under active develop-
ment. Note that ilaprazole belongs – like the other PPIs – to benzimidazole compounds whereas tenatoprazole
represents a different chemical entity.

Being easy to administer through a nasogastric tube, Novel PPIs


IR-OME was also studied for its ability to control intra- Several new PPIs have been synthesized since the dis-
gastric pH in critically ill patients [166, 167]. A random- covery of omeprazole. The majority of them are still in
ized, double-blind trial [168] found this formulation more preclinical development or have been abandoned. This is
effective than intravenous cimetidine (the only FDA-ap- for instance the case of leminoprazole [171], whose de-
proved drug for this indication) in maintaining gastric pH velopment has been interrupted despite the interesting
14 and similarly effective in preventing upper GI bleed- pharmacological profile showing also a peculiar mucosal-
ing without increasing the incidence of pneumonia. protective activity of the drug [172, 173]. Only two drugs
Several studies [162] have shown that antacids do not are actively being studied in humans, namely ilaprazole
have a major effect on the absorption or disposition of and tenatoprazole, and will be reviewed in detail.
DR PPIs. On the contrary, addition of sodium bicarbon- Ilaprazole (compound marked IY-81149, fig. 8) is a
ate to uncoated omeprazole clearly affects its pharmaco- benzimidazole compound synthesized at Il-Yang (South
kinetics and pharmacodynamics. The putative mecha- Korea) and presently developed by TAP Pharmaceutical
nism of action to explain these observations is as follows Products Inc. (Lake Forest, Ill., USA). Although the drug
[162]. Following ingestion of IR-OME as an oral suspen- is already on the market in South Korea, phase II clinical
sion, the sodium bicarbonate causes a prompt rise in in- trials will start soon in the USA.
tragastric pH. While this serves the primary function of Studies performed on rabbit isolated parietal cells
protecting the uncoated omeprazole from acid degrada- [174] have shown that ilaprazole irreversibly inhibits
tion within the stomach, it may also provide a temporary H+,K+-ATPase and aminopyrine accumulation in a dose-
stimulus to gastrin release from antral G cells. Sodium dependent manner with a potency comparable to that of
bicarbonate solution has previously been shown to raise omeprazole. In vivo investigations [174–176] evaluated
circulating gastrin levels within 30 min of oral ingestion the antisecretory activity of the compound, which proved
[169, 170]. The rise in circulating gastrin may stimulate to be 2–3 times stronger than that of omeprazole. In a
the parietal cell mass and promote the insertion of func- model of experimentally-induced esophagitis in the rat
tioning molecules of H+,K+-ATPase into the secretory [176], both ilaprazole and omeprazole prevented the de-
canaliculi. Since the peak plasma concentration of velopment of esophagitis and reduced gastric acid secre-
omeprazole occurs around 30 min after ingestion of IR- tion in a dose-dependent manner. The antisecretory ac-
OME, this allows for the rapid uptake of circulating tivity of this novel PPI was confirmed in GERD patients
omeprazole by activated parietal cells leading to irrevers- where the drug, at 10 and 20 mg daily, provided a great-
ible inhibition of a large proportion of available proton er and prolonged suppression of acid secretion compared
pumps. This sequence of events may explain both the ra- to omeprazole [177]. Since a half-life of 3.6 h (i.e. longer
pidity of onset of the antisecretory effect and its pro- than that of omeprazole) has been reported in healthy
longed duration. volunteers [178], the improved pharmacodynamic activ-

24 Dig Dis 2006;24:11–46 Scarpignato /Pelosini /Di Mario

DDI923.indd 24 02.05.2006 16:24:55


ity could reflect the improved pharmacokinetics of the
drug. It should be pointed out, however, that ilaprazole 4.00

displays an inhibitory effect on cytochrome P450 enzymes, 3.50

TPZ bound (nmol/mg)


as revealed by its dose-dependent ability to prolong hexo- 3.00

barbital-induced sleeping time in mice [175]. 2.50

Tenatoprazole (compound also marked TU-199) has 2.00

been developed by Mitsubishi Pharma in Japan [91] and 1.50

is now under active development by Negma-Gild (France). 1.00

Conversely from all the other PPIs, this compound is not 0.50

a benzimidazole derivative, consisting of one imidazo- 0


0 4 8 12 16 20 24
pyridine ring connected to a pyridine ring by a sulfinyl- Time (h)
methyl chain (fig. 8). It represents therefore a new chem-
ical entity. In vitro studies, performed in hog and dog
gastric microsomes, have shown that tenatoprazole dis- Fig. 9. Time course of loss of 14C-tenatoprazole binding to the
plays an inhibitory action on proton pump comparable H+,K+-ATPase. Radioactive tenatoprazole was administered intra-
venously at 0.1 mCi/kg of animal with a dosage of 20 mol/kg
to that of omeprazole and lansoprazole [179]. The inhib-
through the tail vein. The rats were sacrificed at 1, 2, 6, 12 and
itory activity of this novel compound on gastric H+,K+- 24 h. Each point refers to the mean (8SD) of the values obtained
ATPase has been thoroughly characterized by George from 4 to 6 animals. After maximum binding of tenatoprazole, te-
Sachs’ team [180, 181]. Like the other PPIs, tenatoprazole natoprazole binding up to 24 h post-dose in vivo could be described
is a prodrug (pKa = 4.04), which is converted to the ac- by a first exponential equation: C(TPZ) = 1.824e–0.176t + 1.141. This
first component is mainly from a fast decay with a half-life 3.9 h
tive sulfenamide or sulfenic acid by acid in the secretory and the plateau of 1.141 at 24 h post-dose is related to binding that
canaliculus of the stimulated parietal cell of the stomach. decays with a half-life corresponding to enzyme turnover (54 h)
This active species binds to luminally accessible cysteines (from Shin et al. [180]).
of the gastric H+,K+-ATPase resulting in disulfide forma-
tion and acid secretion inhibition. Tenatoprazole binds
at the catalytic subunit of the gastric acid pump with a
stoichiometry of 2.6 nmol/mg of the enzyme in vitro. In tenatoprazole was unable to affect emptying rate [183].
vivo, maximum binding of tenatoprazole was 2.9 nmol/ In addition, experiments carried out in dogs with gastric
mg of the enzyme at 2 h after intravenous administration. fistula revealed an elevation of intragastric pH lasting lon-
The binding sites of tenatoprazole were at cysteine 813 ger (by 20%) than that observed with omeprazole or lan-
and cysteine 822 as shown by tryptic and thermolysin soprazole [184].
digestion of the ATPase labeled by tenatoprazole. Both Pharmacokinetic studies, performed in Japanese sub-
of these sites are located in the proton transport pathway, jects, showed a 7-fold longer half-life (7.6 and 13.7 h after
though cysteine 822 is found deeper in the TM5/6 mem- single and repeated administration of 20 mg, respective-
brane domain than cysteine 813. Decay of tenatoprazole ly), compared to the other PPIs. As a consequence, the
binding on the gastric enzyme consisted of two compo- area under the concentration-time curves, that reflect tis-
nents. One was relatively fast, with a half-life 3.9 h due sue exposure, was about 20-fold higher after single oral
to reversal of binding at cysteine 813 and the other was a administration of 20 and 40 mg of the drug (24.50 and
plateau phase corresponding to ATPase turnover reflect- 67.76 mg  h/ml, respectively) [185]. These pharmacoki-
ing binding at cysteine 822 that also results in sustained netic properties translated into a long-lasting antisecre-
inhibition in the presence of reducing agents in vitro tory action. Indeed, inhibition of basal and tetragastrin-
(fig. 9). stimulated acid output in healthy volunteers was still sig-
The in vivo antisecretory potency of tenatoprazole, nificant 2 days after tenatoprazole administration.
evaluated in different animal models (Shay rat prep- Intragastric pH recordings, performed after repeated ad-
aration, acute fistula rats and Heidenhain pouch dogs), ministration of a dose as low as 10 mg of the drug, did
proved to be 2–4 times higher than that observed with show that the time spent above pH 3 and pH 4 reached
omeprazole. As a consequence, the healing activity of the 96.7 and 88.0%, respectively. The most interesting find-
compound on experimentally-induced gastric and duode- ing was the prolonged effect of tenatoprazole during
nal ulcers appeared similarly higher [179]. Conversely nighttime, since the % of pH holding time above 3 and 4
from omeprazole, which delays gastric emptying [182], was almost identical during this period compared to day-

Update on Acid Suppression Therapy Dig Dis 2006;24:11–46 25

DDI923.indd 25 02.05.2006 16:24:55


14,000 y = –191.17 + 149.01x 300,000 y = –2.5215e+4 + 2953.7x
12,000 r2 = 0.998 r2 = 0.983
250,000

AUC (ng  h/ml)


10,000

Cmax (ng/ml)
200,000
8,000
150,000
6,000
Fig. 10. Pharmacokinetics of tenatoprazole 100,000
after repeated (7 days) oral administration 4,000
in healthy volunteers. Both Cmax and AUC 2,000 50,000
show a linear correlation with the dose ad-
0 0
ministered. Each point refers to the mean 0 20 40 60 80 100 0 20 40 60 80 100
(8SEM) of the values obtained from 8 sub- Dose (mg) Dose (mg)
jects (drawn from data in Domagala and
Ficheux [188]).

Table 2. Comparison between pharmacokinetic parameters ob- either single and repeated administration). A linear cor-
tained in Japanese or European healthy volunteers after single oral relation between tenatoprazole dose and AUC was found,
administration of 20 mg tenatoprazole (from data in the Investiga-
thus showing a linear pharmacokinetic pattern (fig. 10).
tor’s Brochure [185] and Domagala and Ficheux [188])
The long half-life of tenatoprazole was confirmed for ev-
Pharmacokinetic Japanese subjects Caucasian subjects ery dose, reaching 8.7 8 2.6 h for repeated administra-
parameter (n = 6) (n = 8) tion of 40 mg [188]. A comparison of pharmacokinetic
parameters between Asian and European subjects is
Cmax, g/ml 4.2=0.6 2.980.8
shown in table 2.
tmax, h 2.580.2 2.581.0
t½, h 13.783.0 7.882.0 Pharmacodynamic studies on the same subjects
AUC, g  h/ml 59.2813.0 31.0813.0 showed an increase of intragastric pH with tenatoprazole
40 mg daily for 7 days significantly higher (p ! 0.05) than
Each value represents the mean (8SD) of the values obtained that observed with the same regimen of esomeprazole, the
in 6–9 subjects.
median pH being 4.6 8 0.9 and 4.2 8 0.8, respectively
[189]. In addition, the time spent above pH 4 during
nighttime after tenatoprazole administration was signifi-
cantly longer than that observed with esomeprazole (table
3). The intragastric pH during the night was similarly
time (94.7 vs. 98.3% and 86.3 vs. 89.3%, respectively) higher (4.7 8 1.1 units with tenatoprazole and 3.6 8 1.4
[185]. units with esomeprazole, p ! 0.01) [189]. The better con-
The compound was originally discovered by Mitsubi- trol of intragastric acidity achieved with tenatoprazole
shi Pharma and was therefore first studied in Japanese during the night was already evident from the first 24 h
subjects. Pharmacokinetic (and consequently pharmaco- of dosing [190].
dynamic) differences between East Asians and Cauca- A recent pharmacodynamic and pharmacokinetic in-
sians are well known, being mainly due to genetic poly- vestigation [191] confirmed and extended previous data
morphism of several phase I enzymes such as CYP2D6 showing the prolonged duration of acid suppression with
and the CYP2C subfamily [186]. Since the pharmacoki- tenatoprazole (fig. 11). The proportion of healthy volun-
netics and pharmacodynamics of PPIs depend on teers spending at least 16 h above pH 4 in the 24-hour
CYP2C19 genotype status, CYP2C19 genotype-depen- period was remarkably higher with tenatoprazole than
dent differences in pharmacokinetics and pharmacody- with esomeprazole (81.5 vs. 34.5%, p ! 0.001), while the
namics of PPIs influence the cure rates for the GERD and proportion of subjects with NAB was lower (73.1 vs.
H. pylori infection by PPI-based therapies [187]. Phar- 93.1%, p = 0.06), although the difference fell short of sta-
macokinetics of tenatoprazole was thus re-investigated in tistical significance. Even 3 days after treatment was dis-
H. pylori-negative healthy Caucasian subjects in a ran- continued, mean 24 h pH, and percentage of time at pH
domized, double-blind, dose-ranging study (10–80 mg, 13 and pH 14 were significantly higher with tenatopra-

26 Dig Dis 2006;24:11–46 Scarpignato /Pelosini /Di Mario

DDI923.indd 26 02.05.2006 16:24:56


pH
8

7 Tenatoprazole 40 mg Esomeprazole 40 mg
6

1 pH = 4

0
8 10 12 14 16 18 20 22 24 2 4 6 8
Time (h)

Fig. 11. Mean 24-hour intragastric pH profiles (on day 7) in 30 H. pylori-negative healthy volunteers giving te-
natoprazole (40 mg) or esomeprazole (40 mg) orally once a day. It is evident that the mean duration of NAB is
shorter on tenatoprazole than on esomeprazole (4.3 vs. 6.5 h, p ! 0.0001) (from Hunt et al. [191]).

Table 3. Intragastric acidity in healthy


volunteers given esomeprazole or Esomeprazole Tenatoprazole Tenatoprazole
tenatoprazole once daily for 7 days. 40 mg 20 mg 40 mg
Percentage of time spent above pH 4
on day 7. Each value refers to the mean 24 h (08:00–08:00 h) 56.2816.5 50.9811.1 63.3817.8*
(8SEM) of the values obtained from Daytime (08:00–20:00 h) 65.6817.7*** 47.2812.0 62.2818.5*
18 H. pylori-negative subjects (from Nighttime (20:00–08:00 h) 46.8819.7 54.6814.0 64.3821.7**
Galmiche et al. [189])
Tenatoprazole 40 mg vs. tenatoprazole 20 mg: * p < 0.01.
Tenatoprazole 40 mg vs. esomeprazole 40 mg: ** p < 0.01.
Esomeprazole 40 mg vs. tenatoprazole 20 mg: *** p < 0.01.

zole, indicating a sustained control of intragastric acidity of a phase II dose-ranging (10, 20 and 40 mg daily) Ca-
with this novel PPI compared to esomeprazole. After 7 nadian study, performed in patients with reflux esopha-
days of repeated dosing the maximal plasma concentra- gitis (grade A–C according to Los Angeles classification),
tion of tenatoprazole was almost 6 times higher than that demonstrated that this novel PPI effectively heals muco-
of esomeprazole, while AUC was 32 times higher. A sig- sal lesions regardless of the dose [Alan B. Thomson, pers.
nificant correlation between AUC and percentage of time commun.]. After 4 weeks of treatment with whatever dose
intragastric pH 14 was observed with tenatoprazole not of tenatoprazole, the healing rate was larger than that re-
only during but also after stopping treatment [191]. ported with esomeprazole (40 mg daily) in published clin-
All these data clearly show that – compared to the ex- ical trials.
isting PPIs – tenatoprazole has a longer half-life and a Like the other PPIs, tenatoprazole is a racemic mixture
longer duration of the antisecretory action, in agreement of two stereoisomers which derive from the chiral nature
with the current knowledge according to which the anti- of the sulfur atom of the sulfinyl group [193]. As a conse-
secretory effect is proportional to the AUC [121, 192]. quence, in order to exploit the features of stereoselective
Although these properties should theoretically translate catabolism, the S-isomer was selected for further develop-
into a better therapeutic efficacy, no randomized clinical ment [194]. As expected, the in vitro binding of both S-
trials in acid-related diseases are available as yet. It is and R-tenatoprazole to hog gastric H+,K+-ATPase was
worth mentioning, however, that a preliminary analysis virtually the same and overlapped that observed with the

Update on Acid Suppression Therapy Dig Dis 2006;24:11–46 27

DDI923.indd 27 02.05.2006 16:24:56


200,000 (–)TU-199 Parietal
Plasma concentration of TU-199

180,000 (–)TU-199 Na cell canaliculus


160,000
140,000 K+ Cl –
120,000
(ng/ml)

100,000
80,000 H+
60,000
40,000
20,000
0 Parietal
0 1 2 3 4 5 6 7 8 9 cell cytoplasm
Time (h)
K+

Na + K+ Cl –
Fig. 12. Plasma levels of S-tenatoprazole (TU-199) after oral
administration in dogs. Either the sodium salt or the free from
(100 mg/kg) were administered using a gelatine capsule containing
the crystallized powder of each compound. Each point represents
the mean of the values obtained from 3 to 4 animals (from Shin et
al. [180]). (–)TU-199 = S-tenatoprazole-free form; (–)TU-199 Na = K+ HCO 3–
S-tenatoprazole sodium salt hydrate. Na + K+

racemate [181]. Similarly, there was no difference in Fig. 13. A simplified model for the secretion of gastric acid by the
binding reversibility kinetics amongst the different forms parietal cell. The H+,K+-ATPase located on the apical membrane
of the parietal cell exchanges H+ for K+. K+ is recycled from the
of this PPI [181]. Preliminary experiments in laboratory
canaliculus into the cytoplasm by K+ channels in the apical mem-
animals did confirm the antisecretory and ulcer healing brane. The combined actions of K+ channels and enzymes on the
activity of S-tenatoprazole and did point out that this iso- basolateral membrane regulate cytoplasmic K+ levels. Cl– enters the
mer is more effective than R-tenatoprazole [Ficheux et cell cytoplasm via a Cl–/HCO3– exchanger and moves from the cy-
al., unpubl. observations]. toplasm into the canaliculus via a Cl– channel (most likely ClC-2)
(from Geibel [94]).
A careful pharmacokinetic and structural study by
George Sachs’ team [181] showed that the oral bioavail-
ability of S-tenatoprazole sodium salt hydrate is almost
twice that of S-tenatoprazole-free form (fig. 12). The dif-
ference in bioavailability can be explained by the better markedly increased and remained over 5 or above
solubility of the sodium salt due to its peculiar crystal throughout most of the 24-hour recording period, both
structure. The crystal form of S-tenatoprazole sodium salt during day- and nighttime [Galmiche, pers. commun.].
hydrate is indeed quite different from that of S-tenato- Some pharmacodynamic and healing studies with S-te-
prazole-free form. In the crystal of S-tenatoprazole sodi- natoprazole sodium have already been planned and will
um salt hydrate there is a loose packing structure of mol- start in the near future.
ecules, which results in rapid water access and hence In summary, the available studies point out both phar-
greater solubility. The pharmacokinetics of S-tenatopra- macokinetic and pharmacodynamic advantages of te-
zole sodium was then studied in healthy volunteers and natoprazole over esomeprazole. Since this last compound
proved to be linear with a dose-related increase in both provides – amongst the members of the class – the most
Cmax and AUC [Ficheux et al., unpubl. observations]. The effective control of intragastric pH whatever the param-
antisecretory effect of this stereoisomer (80 mg orally eter considered [195], it is conceivable that tenatoprazole
once a day over 1 week) was investigated in H. pylori- could similarly be better than the other existing PPIs. Te-
negative healthy subjects. On day 7, intragastric pH was natoprazole (as well as its S-isomer) then appears a prom-

28 Dig Dis 2006;24:11–46 Scarpignato /Pelosini /Di Mario

DDI923.indd 28 02.05.2006 16:24:57


Table 4. P-CABs already studied in clinical trials

P-CAB Chemical class Clinical Company


development phase

AZD0865* Imidazopyridine Phase II AstraZeneca


CS526 (R105266)* Pyrrolopyridazine Phase I Sankyo and Ube/Novartis
Revaprazan (YH1885, Revanex) Pyrimidine Phase III Yuhan
Soraprazan (BY359) Imidazonaphthyridine Phase II Altana

* Discontinued in July 2005.

ising PPI for the treatment of acid-related diseases, where lished in 1982, this compound was shown to inhibit gas-
it has the potential to address unmet clinical needs. tric acid secretion in humans [197], although its mecha-
nism of action was not fully understood at the time. Sub-
Potassium-Competitive Acid Blockers sequently, SCH28080 was shown to block gastric
The next generation of drugs which suppress gastric H+,K+-ATPase by competing with K+ [198]. The develop-
acidity will most likely be P-CABs which are K+-com- ment of SCH28080 was discontinued because of hepatic
petitive inhibitors of the ATPase [93]. While the PPIs toxicity. Since then, several other compounds with this
have a unique mechanism of action based on their chem- peculiar mechanism of action have been synthesized and
istry, P-CABs have a structural specificity for their target, studied, but only few did reach clinical development.
the K+-binding region of the H+,K+-ATPase. Four representative members of the P-CAB class have
Although it can also be activated by NH4+ in vitro, the been studied in humans with the aim of finding a suitable
proton pump is highly selective for K+ [196]. In common drug for the treatment of acid-related diseases (table 4,
with many other cells, the level of K+ in the parietal cell fig. 14). Despite a number of papers presented at the re-
is higher than that in the plasma. The higher intracellular cent DDW (Chicago 2005) and UEGW (Copenhagen
K+ level is dependent on H+,K+-ATPase. This enzyme, 2005) and two clinical trials on GERD (registered at
located on the basolateral membrane of the cell, ex- www.clinicaltrials.gov) that were successfully completed,
changes intracellular H+ for extracellular K+ [10]. The AZD0865 was discontinued [199]. The same holds true
level of K+ within the cell is also regulated by K+ chan- for CS526. However, a follow-up compound of sorapra-
nels, which allow ion movement across the basolateral zan is currently in phase I [200]. Therefore, there are still
membrane (fig. 13). These channels have a particularly three molecules under active development.
important role in generating negative cell membrane po- P-CABs are lipophilic, weak bases that have high pKa
tential. Given the importance of the cation for enzyme values and are stable at low pH. This combination of
function, agents that compete with the binding of K+ properties allows them to concentrate in acidic environ-
have the potential to block acid secretion. P-CABs in- ments. For example, the concentration of a P-CAB with
hibit H+,K+-ATPase by binding ionically to the enzyme a pKa of 6.0 would theoretically be expected to be 100,000-
and thus prevent its activation by the K+ cation. Since fold higher in the parietal cell canaliculus (pH = 1) than
these molecules are larger than K+, it is likely that they in the plasma (pH = 7.4). The concentration of P-CABs
compete by preventing the access of the cation to its in the gastric mucosa is demonstrated by in vitro and in
binding site rather than occupying the ion-binding site vivo studies with AZD0865 and revaprazan [201, 202].
directly. On entering an acidic environment, P-CABs are instant-
P-CABs, despite sharing the same mechanism of ac- ly protonated and it is in this form that it is thought to
tion, represent a heterogeneous class of drugs. Indeed, bind to and inhibit the enzyme. The protonated form of
they belong to four different chemical classes, namely a P-CAB inhibits H+,K+-ATPase by binding ionically to
imidazopyridines, pyrimidines, imidazonaphthyridines it, as illustrated by the recovery of enzymatic activity af-
and quinolones [93, 94]. ter washout of AZD0865 and revaprazan [201, 202]. This
The prototype P-CAB, SCH28080, was developed by family of compounds binds to the outward-facing confor-
Schering-Plough more than 20 years ago. In a study pub- mation of the pump on the luminal side and do not re-

Update on Acid Suppression Therapy Dig Dis 2006;24:11–46 29

DDI923.indd 29 02.05.2006 16:24:57


Fig. 14. Chemical structures of P-CABs under clinical investigation.

quire its activation. This would suggest that these agents for PPIs [207], the pH-rising effect of P-CABs seems to
will produce more rapid acid inhibition and will be able be increased in the presence of H. pylori infection [208].
to elevate gastric pH to a higher level than PPIs. Animal studies have shown a close correlation be-
Both animal and human studies have shown that P- tween maximum inhibition of acid output and the loga-
CABs achieve rapidly peak plasma concentrations after rithm of Cmax [93], thus suggesting that the duration of
oral administration. This is partly due to their stability at action of P-CABs will depend on their half-life. One draw-
low pH allowing their administration as IR formulations. back of these agents is the need for twice-daily dosing
All the compounds studied to date exhibit a linear phar- since they inhibit the pump only for the duration of their
macokinetic pattern [93]. The rapid absorption of P- presence in the blood; however, since these compounds
CABs is mirrored by a fast onset of acid inhibition. In are acid-stable, their plasma half-life may be readily pro-
healthy volunteers, a single oral dose of pumaprazole (an longed with extended-release formulations [92]. The
imidazopyridine compound marked BY841) [203] or re- main differences between P-CABs and PPIs are summa-
vaprazan [204] increased intragastric pH to about 6 with- rized in table 5. It is evident that P-CABs offer a more
in 30–60 min, whereas high doses of AZD0865 resulted rapid elevation in intragastric pH than a PPI (and similar
in over 95% inhibition of acid secretion within 1 h after to that achieved by an H2-RA) while maintaining the
oral dosing [205]. Both the degree [206] and the duration same degree of antisecretory action, whose duration is
[205] of the antisecretory action are dose-dependent. As dependent on half-life and can easily be prolonged by ap-

30 Dig Dis 2006;24:11–46 Scarpignato /Pelosini /Di Mario

DDI923.indd 30 02.05.2006 16:24:58


Fig. 15. Chemical structure of the novel H2-receptor antagonists ebrotidine and lafutidine. Besides S- and R-iso-
mers, cis- and trans-isomers of lafutidine have also been isolated.

Table 5. P-CABs and PPIs: main


differences in the mechanism of action P-CABs PPIs

Acts directly on the H+,K+-ATPase enzyme Requires transformation to the active form
Super-concentrates in parietal cell acid space Concentrate in parietal cell acid space
(100,000-fold higher than in plasma) (1,000-fold higher than in plasma)
P-CABs binds competitively to the potassium Sulfenamide binds covalently
binding site of H+,K+-ATPase to H+,K+-ATPase
Duration of effect related to half-life Duration of effect related to half-life
of drug in plasma of the sulfenamide-enzyme complex
Full effect from first dose Full effect after repeated doses

propriate formulations. Whether these favorable phar- on the way in Europe [209]. A combination of H2-RA
macodynamic properties will translate into clinical ben- with the novel PPI tenatoprazole has also been patented
efits is unknown. Results of phase III clinical trials with [210].
one of such compound (namely revaprazan) are eagerly Although the last decade has been dominated by the
awaited. growing use of PPIs, several new H2-RAs have been syn-
thesized. While the majority of them have been discon-
New H2-Receptor Antagonists tinued, few molecules reached clinical development and
As already discussed, despite all the intrinsic limita- two (namely ebrotidine and lafutidine, fig. 15) have actu-
tions, there is still a place for H2-RAs in the era of PPIs. ally been marketed. These drugs belong to a new genera-
A recent survey of Castell’s team [71] found that the ma- tion of H2-RAs which combine the antisecretory effect
jority of GERD patients report a persistent improvement with a mucosal-protective activity.
of nighttime symptoms from bedtime H2-RA use and sug- Ebrotidine has been developed by the Ferrer group in
gested that a possible clinically important tolerance does Spain [211]. The drug is able to displace 3H-thiotidine-
occur in a small number of patients. In this connection a specific binding to H2-receptors with an affinity (Ki
fast-dissolving oral tablet containing a fixed dose combi- 127.5 nmol/l) significantly (p ! 0.05) higher than that
nation of a PPI and an H2-RA (product marked OX 17) of ranitidine (Ki 190.0 nmol/l) and cimetidine (Ki
has been developed by Orexo AB (Uppsala, Sweden) and 246.1 nmol/l) [212]. Its antisecretory efficacy is similar to
phase II clinical trials with this formulation are presently that of ranitidine and 10 times higher than that of cimeti-

Update on Acid Suppression Therapy Dig Dis 2006;24:11–46 31

DDI923.indd 31 02.05.2006 16:24:58


inhibition of tiotidine binding and histamine-induced cy-
clic-3,5-adenosine monophosphate production in Chi-
Lafutidine 10 mg nese hamster ovary cells expressing human H2-receptors
100 Rabeprazole 20 mg
[221]. Animal experiments [222] have pointed out that
pH holding time (%)

80
✽✽
lafutidine has a potent and long-lasting antisecretory ef-
✽✽ ✽✽ ✽✽
fect, a finding confirmed by human investigations [223]
60 ✽✽
✽✽ showing that this drug inhibits acid secretion more
40 ✽✽
strongly than conventional H2-RAs. Conversely from ra-
20 nitidine and famotidine, lafutidine increases both day-
0
and nighttime intragastric pH in H. pylori-negative sub-
jects [224], and conversely from omeprazole (and other
1 2 3 3.5 4 5 6 7 8
PPIs) its efficacy is not influenced by the CYP2C19 gen-
pH otype status [223]. When used in combination with anti-
biotics (namely clarithromycin and amoxicillin) it
achieves an eradication rate which is comparable to that
of the same PPI-based triple therapy [225]. A recent cross-
Fig. 16. Time spent above a given pH in the postprandial period
over study [226] in healthy volunteers did show that a
after administration of a single dose of lafutidine (10 mg) or rabe-
prazole (20 mg) in healthy volunteers. Each point refers to the mean single dose (10 mg) of this novel H2-RA is able to increase
(8SD) of the values obtained from 10 subjects. ** Significant (p ! intragastric pH more quickly than a single dose (20 mg)
0.01) difference between means (from Inamori et al. [226]). of rabeprazole, the fastest amongst the available PPIs
[227]. Both in fasting conditions and in the postprandial
state, the duration of the antisecretory action was longer
dine [211]. The mucosal-protective properties of ebroti- than that of the PPI since the drug maintained the pH
dine, possibly due to enhanced mucosal synthesis of both over a given threshold for a sustained period of time (fig.
endogenous prostaglandins (PGs) and nitric oxide (NO) 16). Its terminal half-life (3.30 8 0.39 and 3.79 8 1.02
[213, 214], stem from its ability to improve the physico- h in fasting and fed conditions, respectively [228]) is lon-
chemical characteristics of mucus gel (i.e. increase in mu- ger than that of cimetidine, ranitidine and nizatidine [55,
cus gel dimension, viscosity, hydrophobicity and hydrogen 229] and shorter than that of roxatidine, but not dissimi-
ion retardation capacity). Improvements in mucus gel-pro- lar from that of famotidine [55].
tective qualities with ebrotidine are directly related to the Conversely from other H2-RAs [230], lafutidine does
ability of the drug to enhance the synthesis and secretion increase SST release, which may contribute to its antise-
of sulfo- and sialomucins and phospholipids of gastric mu- cretory effect [231]. Similarly, the observed rise in calci-
cus and to promote mucin macromolecular assembly tonin gene-related peptide (CGRP) levels after drug ad-
[215]. This drug also exhibits an inhibitory activity against ministration could account for both its acid-lowering and
H. pylori that is synergistic with a number of antibacterial cytoprotective activities [232].
agents; it inhibits the urease enzyme and the proteolytic Lafutidine displays mucosal protection against a vari-
and mucolytic activities of H. pylori, and counteracts the ety of noxious agents, and structure-activity relationships
inhibitory effects of H. pylori lipopolysaccharide. revealed that the presence of furfurylsulfinyl, pyridyl and
Although ebrotidine proved to be as effective as ra- amide moiety (fig. 15) are chemically important for the
nitidine for the treatment of patients with PU or erosive gastroprotective activity. The mucosal-protective effect
reflux esophagitis [211], it achieved a significantly better of lafutidine is attenuated by pretreatment with the an-
ulcer healing rate (94 vs. 86%, p ! 0.05) than ranitidine tagonist of CGRP (CGRP8-37) and the blocker of NO
treatment in smokers [216]. Unfortunately, the drug was production (NG-nitro-L-arginine), as well as chemical ab-
prematurely withdrawn from the Spanish market because lation of capsaicin-sensitive sensory neurons, suggesting
of serious hepatotoxicity [217–219]. that this action appears through capsaicin-sensitive affer-
Another peculiar H2-RA endowed with both antisecre- ent neurons and is mediated by CGRP and NO [233].
tory and mucosal-protective activities is lafutidine. This Interestingly enough, lafutidine also exhibits a protective
drug, originally developed by Fujirebio Inc. [220], is cur- activity against the experimentally-induced mucosal le-
rently marketed by UCB and Taiho Pharmaceutical in sions in digestive tissues other than the stomach, i.e. acid-
Japan. This compound showed a potent and long-lasting induced reflux esophagitis [234], indomethacin-induced

32 Dig Dis 2006;24:11–46 Scarpignato /Pelosini /Di Mario

DDI923.indd 32 02.05.2006 16:248


small intestinal lesions [235] and colonic inflammation As a consequence, they both bind to the same receptors
induced by dextran sulfate sodium [236]. Finally, this H2- located on parietal cells, and CCK also stimulates acid
RA inhibits H. pylori-induced IL-8 release from gastric secretion both in vitro and in vivo. CCK however stimu-
epithelial cells and impairs cellular adhesion of the mi- lates fundic D cells to produce SST, which in turn inhib-
croorganism, protecting in this way against the mucosal its both gastrin release and acid secretion [244].
inflammation associated with the infection [238]. Receptors for CCK are a family of G protein-coupled
Provided lafutidine is available worldwide, it will rep- receptors, which were first characterized on pancreatic
resent a quick and strong means of inhibiting acid secre- acinar cells and identified as CCK type A receptors (now
tion which is particularly suitable for on-demand use in called CCK1), with the subsequent discovery in the same
PUD and GERD, where it could also be useful to control year of a second receptor with a different pharmacology
NAB. It could also be particularly effective in the preven- in the brain, i.e. CCK type B receptors (now CCK2)1 [244,
tion and treatment of NSAID-associated GI lesions. In 245]. These two types of CCK receptors could be phar-
this setting, its marked and long-lasting antisecretory ac- macologically distinguished on the basis of their affinity
tivity, coupled with the mucosal-protective properties, for the agonists CCK and gastrin and by recently devel-
will probably allow the drug to go beyond the intrinsic oped subtype-specific antagonists. CCK1 receptors are
limitations of conventional H2-RAs, which only protects highly selective (500- to 1,000-fold) for sulfated analogs
the duodenum [35, 37]. In this connection, the use of la- of CCK, whereas CCK2 receptors have similarly high af-
futidine and its isomers [238] in the prevention or treat- finity for both sulfated and nonsulfated peptide analogs
ment of inflammatory enteropathy has recently been pat- of CCK and gastrin peptides [245, 246]. However, al-
ented [239]. though binding the same receptors, gastrin is mainly a
According to R&D Focus, some other H2-receptor an- peripheral ligand, while CCK is considered a brain-gut
tagonists are being developed. Among them, CP-66,948 peptide thanks to its effects on the CNS. Formerly, the
developed by Pfizer Inc. (Groton, Conn., USA), a com- gastrin receptor mediating acid secretion in the stomach
pound [2-(N-pentyl-N-guanidino)-4-(2-methylimidazol- was thought to constitute a third type of high-affinity re-
4-yl)thiazole] possessing both antisecretory and mucosa- ceptor on the basis of its location, small differences in
protective properties [240] entered phase I trials. Prelim- affinity for CCK and gastrin-like peptides, and the rever-
inary results have shown that a single oral bedtime dose sal in relative affinity for receptor subtype-selective an-
(50 mg) of CP-66,948 produced the same inhibition of tagonists in canine gastric glands. Subsequent cloning of
acid secretion achieved by 150 mg of ranitidine. However, gastrin receptors from canine stomach and CCK2 recep-
no further development is currently reported. A similar tors from canine brain revealed their molecular identity,
compound (TRM 115 by Terumo Co., Tokyo, Japan) was leading to the classification of gastrin receptors as CCK2
recently discontinued after completing phase I studies. receptors [245]. These receptor subtypes have been found
not only on parietal cells [246] but also on ECL cells [247],
Gastrin (CCK2) Antagonists fundic D cells [248] and the vagus nerve [246].
Gastrin is a major endocrine regulator of gastric acid The known targets for gastrin-mediated acid secretion
secretion and its release stimulates an estimated mean of are histamine-releasing ECL cells and acid-secreting pa-
90% of postprandial secretion [1]. This hormonal peptide rietal cells, which both possess a CCK2 receptor [249].
is produced by the antral G cells in response to food pro- The current concept of gastrin regulation of acid secretion
teins and their digestion by-products as well as upon stim- involves a complex but incompletely delineated mecha-
ulation by gastrin-releasing peptides from postganglionic nism. The feedback loop is constituted by at least three
fibers of the vagus nerve [241]. Besides its central role in key elements: the G cell, the ECL cell, and the parietal
the regulation of acid secretion, gastrin also affects GI
motility [242] and stimulates epithelial cell proliferation
throughout the GI tract [243]. 1
The initial nomenclature of the receptors as CCK-A (A for alimentary)
Gastrin displays a structural similarity with cholecys- and CCK-B (B for brain) receptors is generally accepted by pharmacologists
and molecular biologists. Based on the guidelines defined by the International
tokinin (CCK), another GI peptide released by lipids Union of Pharmacology (IUPHAR) Committee on Receptor Nomenclature
from the upper small bowel I cells [244]. Both peptides and Drug Classification, receptors should be named after their endogenous
share indeed the same C-terminal pentapeptide amide ligands and identified by a numerical subscript corresponding to the chrono-
logical order of the formal demonstration of their existence by cloning and se-
sequence although they differ in sulfation at the sixth quencing. Because the CCK-A receptor was the first to be cloned, it should be
(gastrin) and seventh (CCK) tyrosyl residues [241, 244]. renamed CCK1, and the CCK-B receptor should become CCK2.

Update on Acid Suppression Therapy Dig Dis 2006;24:11–46 33

DDI923.indd 33 02.05.2006 16:24:59


Fig. 17. Chemical structure of CCK2 antagonists under clinical investigation.

cell, and their products gastrin, histamine and hydrochlo- presumed to play a role in subsequent histamine release
ric acid, respectively. Gastrin has a direct secretory effect [249]. The released histamine is a potent stimulant of acid
on both parietal cells and ECL cells, although the latter secretion, exerting a direct stimulatory effect on the H2-
effect is probably far more significant. The peptide medi- receptor of the parietal cell. The parietal cell secretory
ates histamine release from ECL cells via activation of its product, hydrochloric acid, causes antral inhibition of gas-
CCK2 receptors. The initial signal transduction events in- trin release from the G cell, probably via a SST-regulated
volve a transient rise in intracellular calcium, followed by mechanism. It also seems probable that SST by a direct
histamine granule exocytosis [249]. This process occurs action on ECL cells is able to inhibit histamine release,
maximally within 5 min. Thereafter, extracellular calcium since the direct effect of paracrine peptide on parietal cells
influx and histidine decarboxylase (HDC) activation are is not pronounced in vitro [4]. Indeed, ECL cells have been

34 Dig Dis 2006;24:11–46 Scarpignato /Pelosini /Di Mario

DDI923.indd 34 02.05.2006 16:24:59


20,000 y = –1609.9 + 24.956x 70 y = 15.471 + 7.1376e2x
r2 = 0.945 60 r2 = 1.000

Acid inhibition (%)


15,000

AUC (ng/ml  h)
50

40
10,000
Fig. 18. Pharmacokinetics and pharmaco- 30
dynamics of itriglumide after single admin- 20
5,000
istration in healthy volunteers. a Plot of
10
AUC vs. dose, showing a linear pharmaco-
kinetic pattern. b Dose-dependent inhibi- 0 0
tion of G-17 (32 pmol/kg  h) induced acid 0 100 200 300 400 500 600 700 0 100 200 300 400 500 600 700
secretion (drawn from data in Beglinger et a Single dose (by oral route) (mg) b Single dose (by oral route) (mg)
al. [264]).

reported to express SST receptors, suggesting a dual regu- Amongst the amino acid derivatives, proglumide,
lation of the ECL cell by both gastrin and SST [250]. which has long been used as an antiulcer drug [256], was
Whilst gastrin appears to be a major stimulant for hista- considered the prototype CCK antagonist [257]. Its low
mine release, there is also significant evidence for the role potency and specificity (the compound binds to both
of other gut neurotransmitters, namely acetylcholine, va- types of receptors) stimulated the synthesis of glutaramic
soactive intestinal peptide, PYY and corticotropin gene- acid derivatives, the most interesting ones being lorglu-
related peptide (CGRP) in this process [251]. mide and loxiglumide (as selective CCK1-receptor antag-
Given the important physiological role of gastrin in onists) and spiroglumide and itriglumide (as CCK2-re-
the stimulation of gastric acid secretion, selective CCK2- ceptor antagonists). These compounds (developed at Rot-
receptor antagonists offer a potential approach to regulate ta Research Laboratorium, Milan, Italy) are active after
acid production. High-affinity nonpeptide antagonists for oral administration and able to antagonize the effects of
both types of CCK receptors were synthesized more than both endogenous and exogenous peptides (i.e. CCK and
15 years ago. Their availability has stimulated a broad gastrin) [258, 259].
array of investigations into the physiologic actions of Animal studies [260, 261] have shown that spiroglu-
CCK-like peptides [252], including gastrin [253]. mide (compound marked CR-2194) is a selective CCK2
At least 12 chemical classes of CCK-receptor antago- antagonist, capable of inhibiting dose-dependently penta-
nists have been described [254]. Amongst them, several gastrin- but not carbachol- or histamine-stimulated acid
ligands with high affinity for CCK2 receptors have been secretion. Studies performed on healthy volunteers [262]
identified and characterized by binding and pharmaco- confirmed the inhibitory activity against gastrin-induced
dynamic studies [246]. However, only few have been test- acid secretion and also showed its ability to inhibit post-
ed in humans for their effect on acid secretion or their prandial gastric secretion. Despite these promising re-
anxiolytic activity2. The 5 compounds, whose antisecre- sults, the development of spiroglumide was ended because
tory effect was studied in clinical trials, will be reviewed more potent and selective derivatives became available.
in detail. They belong to amino acid (i.e. spiroglumide Itriglumide (compound marked CR-2945) is a follow-
and itriglumide) or benzodiazepine (i.e. L-365,260, YF- up selective antagonist endowed with a good oral bio-
476 and Z-360) derivatives (fig. 17). availability [263]. After intraduodenal administration,
the drug proved to be more potent as an antisecretory
compound than either ranitidine or omeprazole [263]. Its
healing activity of experimentally-induced gastric and
2
The involvement of CCK in human anxiety is well documented. Exog- duodenal ulcer was comparable to that of ranitidine. The
enous administration of CCK2-receptor agonists, such as cholecystokinin-tet-
rapeptide and pentagastrin, provoke panic attacks in man. Patients with panic
pharmacodynamics and pharmacokinetics of itriglumide
disorder are hypersensitive to CCK2-receptor stimulation compared to healthy were recently studied in humans, where the compound
volunteers and patients with other anxiety disorders, and they differ from inhibited gastrin-stimulated acid secretion in a dose-de-
healthy subjects in CCK metabolism and genetic characteristics of the CCK2-
receptor system. As a consequence, some CCK2 antagonists (e.g. L-365,260 or pendent manner (fig. 18). The pharmacokinetics proved
CI-988) are being developed as anxiolytics [254, 255]. to be linear in the dose range of 30–600 mg and drug was

Update on Acid Suppression Therapy Dig Dis 2006;24:11–46 35

DDI923.indd 35 02.05.2006 16:24:59


well tolerated at any dose level [264]. Itriglumide there- famotidine and omeprazole, respectively [279]. The po-
fore appears to be a promising CCK2 antagonist that de- tent and long-lasting antisecretory effect of YF-476 was
serves further clinical investigations in acid- and gastrin- confirmed in human studies after single administration
related disorders. of the drug [280], which also found a linear pharmacoki-
The other major chemical class of CCK2 antagonists netics with a half-life of 7.1 h after oral administration of
has exploited a benzodiazepine template present in 100 mg. However, the effect of repeated (7 days) doses of
asperlicin, which was initially discovered in a natural this compound on intragastric pH was disappointing,
product screen for CCK receptor antagonists. The struc- with the 200-mg daily dose giving a significant inhibition
turally related benzodiazepines L-365,260 and YF-476 of acid secretion only in the postprandial period [281].
are selective antagonists of the CCK2 receptor subtype. The adaptation of the antisecretory effect of YF-476 after
Their in vitro pharmacological profiles have been charac- short-term administration is unlikely to be due to endog-
terized using the human CCK2 receptor expressed in Chi- enous gastrin increase [282] and might reflect up-regula-
nese hamster ovary cells. Conversely from other benzodi- tion of gastrin (CCK2) and/or histamine receptors.
azepine derivatives (L-740,093 and YM022), which dis- According to R&D Focus, a third benzodiazepine de-
play an insurmountable antagonism, L-365,260 (devel- rivative, presently in a phase I study in UK, is the com-
oped at Merck Research Laboratories, West Point, Pa., pound marked Z-360, developed at Zeria Pharmaceutical
USA) behaves as a competitive antagonist [265]. Animal Co., Japan. Preclinical studies [283] have shown a very
experiments [266–270] have shown that this compound high (615-fold) selectivity toward human CCK2 receptors
inhibits pentagastrin-stimulated acid secretion and that compared to CCK1 ones. The drug dose-dependently in-
higher doses also affect basal gastric secretion as well as hibited pentagastrin- but not histamine- or carbachol-
the response to other secretagogues. Thanks to its antise- stimulated acid secretion with a potency 50 times higher
cretory activity, the drug proved to be capable of prevent- than that of L-365,260. In Pavlov pouch dogs, postpran-
ing aspirin-associated gastric damage and cysteamine-in- dial acid secretion was inhibited by 70% by Z-360 while
duced duodenal ulcer. In humans, L-325,260 confirmed other CCK2 antagonists (namely L-365,260 and YM 022)
its selectivity towards CCK2 receptors being able to in- proved to be ineffective [284].
hibit pentagastrin-stimulated acid secretion [271] but not Being CCK2 receptors expressed in the regenerative
fat-induced gallbladder emptying [272]. The drug does mucosa adjacent to the ulcer margin they can mediate the
not inhibit – at the clinically tested doses – basal acid se- gastrin-enhanced cell proliferation underlying ulcer heal-
cretion and has poor bioavailability. Its development as ing [285]. Therefore, despite their acid-lowering activity,
an antisecretory compound is therefore unlikely. Never- CCK2 antagonists might delay mucosal healing. Because
theless, since L-325,260 crosses the blood-brain barrier of this concern and because of the development of toler-
[273], reaching and binding central CCK2 receptors, it ance (see above), it is unlikely that these compounds will
represents a useful tool to assess the physiology and patho- be used as antiulcer drugs. Similarly, it is difficult to imag-
physiology of CCK in the CNS. A placebo-controlled tri- ine these agents as an alternative to PPIs in GERD. On
al in patients with panic disorders [274] gave however the contrary, their use together with long-term acid sup-
disappointing results. A back-up compound (L-368,935) pression would prevent the consequences of PPI-induced
with more water solubility [275] and less CNS penetra- hypergastrinemia (e.g. ECL cell hyperplasia or gastrin-
tion [276] and thus more suitable for peripheral receptor driven GI malignancies) [286–289]. As a matter of fact,
blockade has entered phase I in the USA. a preclinical study using the benzodiazepine CCK2-recep-
YF-476 is a potent and orally active CCK2 antagonist tor antagonist Z-360 seems to confirm this hypothesis.
synthesized at Ferring Research Institute (Southampton, Rats were given repeated doses of Z-360, omeprazole or
UK) [277] and co-developed with Yamanouchi Pharma- a combination of the two compounds for 4 weeks. Al-
ceutical Co. (Tsukuba, Japan). In vitro and vivo experi- though both drugs caused hypergastrinemia, omeprazole
ments [278, 279] have confirmed its high selectivity by but not Z-360 caused ECL hyperplasia. In addition, Z-
showing that its affinity for the rat brain CCK2 receptors 360 significantly reduced omeprazole-induced hyper-
is 4,100 times higher than that for rat pancreatic CCK1 plasia [283], thus suggesting that CCK2-receptor block-
receptors and that the drug is able to inhibit pentagastrin- ade could prevent ECL cell proliferation stimulated by
but not histamine-stimulated acid secretion in rats and long-term treatment with PPIs. In this connection, sev-
dogs. In dogs with gastric fistula its antisecretory activity eral patents concerning this particular application of
was found to be 7 and 40 times more potent than that of CCK2 antagonists have been granted to the James Black

36 Dig Dis 2006;24:11–46 Scarpignato /Pelosini /Di Mario

02.05.2006
DDI923.indd
16:25:00
36
Foundation [290]. An additional logic use of this class of
drugs is the treatment of GI and pancreatic cancer. In- G-17DT immunogen
deed, it is becoming increasingly apparent that gastrin
and gastrin (CCK2) receptors are expressed in a number Synthesized Peptide N-terminal
gastrin peptide spacer end
of tumor sites (GI and non-GI) throughout the body
[291]. It is noteworthy that in these neoplastic cells post-
translational processing of gastrin is not as well developed
as in endocrine cells and other less completely processed
DT
forms of gastrin, such as glycine-extended gastrin, may
also have a role [292]. Despite being attractive from a
theoretical point of view, targeting CCK2 receptors in
cancer however has not provided optimal beneficial ef-
fects, as yet [293]. Diphtheria toxoid (DT)

Antigastrin Vaccine
G-17DT is an immunoconjugate of the amino-termi- Fig. 19. Antigastrin immunogen. G-17DT consists of: (a) A syn-
nal sequence of gastrin 17 (G-17) linked by means of a thetic peptide which is similar to a portion of the targeted hormone
spacer peptide to diphtheria toxoid (fig. 19) [294]. It has gastrin 17. (b) A ‘carrier’, diphtheria toxoid (DT), to which a num-
ber of these synthetic gastrin peptides are chemically bound. DT,
been developed by Aphton Corp. (Miami, Fla., USA),
which is recognized as foreign, provokes the body into initiating an
originally labeled as Gastrimmune™ and later known as immune response. Since the body normally will not mount an im-
Insegia™. Given as an intramuscular vaccination, it has mune response against ‘self’ proteins and molecules, the coupling
been shown to induce the formation of antibodies that of DT to the synthetic peptide is critical for provoking an immune
can neutralize not only G-17 but also its precursor gly- response against the synthetic peptide. Since the peptides resemble
a portion of the targeted hormone, the antibodies produced also
cine-extended G-17, which also stimulates the growth of
bind and neutralize natural gastrin 17. (c) A proprietary, slow-re-
human gastric and possibly other GI cancers [295]. These lease emulsion which contains (a) and (b).
antigastrin antibodies can inhibit the proliferation of
pancreatic cancer cell lines. In a phase II study of 30 pa-
tients with pancreatic cancer, 67% of them were shown
to mount an immune response to the vaccine. Eighty-two able mean of achieving acid suppression while avoiding
percent of patients given the highest dose of the vaccine hypergastrinemia. A phase II clinical trial in GERD has
(250 g) achieved a response. There was indeed a signif- been started although not currently recruiting patients
icant difference between the median survival times noted [302]. Aphton has also patented the use of G-17DT to-
for patients who produced an immune response and for gether with antisecretory agents for the treatment of
those who did not [296]. While preliminary, these results GERD [303]. Combination therapy is claimed to be a
are promising and three phase III clinical trials (registered more effective method of controlling gastric acid secretion
at www.clinicaltrials.gov) with G-17DT, alone or in com- since two independent mechanisms will operate. In addi-
bination with chemotherapy, are ongoing [297]. tion, the gastrin immunogen will block PPI-induced hyper-
In addition to the treatment of GI and pancreatic can- gastrinemia, thus reducing mucosal hyperplasia [290].
cer, the use of G-17DT has been proposed for the manage-
ment of GERD. Indeed, besides stimulating acid secretion,
G-17 also affects lower esophageal sphincter function. In- Conclusions and Perspectives for the Future
deed, continuous intravenous infusion of this peptide to
healthy volunteers significantly increases gastroesophageal The discovery of gastrin by John Edkins [304] initi-
reflux and the number of transient lower esophageal sphinc- ated the scientific examination of the regulation of gastric
ter relaxations associated with reflux [298]. Active immu- acid secretion and led to elucidation of the pathogenic
nization against G-17 should not only neutralize its acid- basis of PU and its subsequent cure. During the course of
stimulating properties but also reduce postprandial gastro- the century after this breakthrough, the identification of
esophageal reflux. Animal studies [299–301] have indeed the cellular regulators of acid secretion culminated in the
shown that this antigastrin vaccine strongly inhibits gastric development of novel pharmacotherapeutic agents,
acid secretion. Therefore, G-17DT would represent a reli- namely H2-RAs and PPIs, which allowed the effective

Update on Acid Suppression Therapy Dig Dis 2006;24:11–46 37

DDI923.indd 37 02.05.2006
02.05.2006 16:25:00
16:25:00
and safe treatment not only of PU but also of GERD and bination with PPIs will be attempted with the aim of re-
other acid-related disorders. Although antisecretory ther- ducing the long-term consequences of hypergastrinemia
apy has advanced dramatically since the introduction of [288, 289]. This combination would be particularly useful
cimetidine in the mid-1970s, there are several identifiable in patients with ZES, where the elevated gastrin levels
unmet needs especially in the management of GERD, [307] may also be associated with reduced absorption of
where an antisecretory therapy with rapid onset of action sodium and water in the gut as well as with the increased
and sustained antisecretory effect would be desirable. occurrence of carcinoids [308].
This is also true in the management and prevention of While H2-receptor antagonists (especially soluble or
nonvariceal upper GI bleeding and may be increasingly OTC formulations) will become the ‘antacids of the third
important in patients taking NSAIDs. millennium’ and will be particularly useful for on-de-
A number of new drugs are currently being investigated mand symptom relief, clinicians will continue to rely on
to provide a significant advance on current treatments. PPIs to control acid secretion in GERD and other acid-
Some of them (namely P-CABs and CCK2-receptor an- related diseases. In this connection, new formulations,
tagonists) have already reached clinical testing while some novel compounds and better acid-suppressing regimens
others (like the antigastrin vaccine, H3-receptor ligands or are welcome.
gastrin-releasing peptide (GRP) receptor antagonists) are The recently introduced IR-OME formulation (cur-
still in preclinical development and need the proof of con- rently available only in the USA) quickly increases intra-
cept in human beings. An increasing body of evidence sug- gastric pH and, given at bedtime, seems to achieve a bet-
gests that H3-receptors are involved in the regulation of ter control of nocturnal acidity. IR formulations of other
acid secretion as well as mucosal protection [for review, PPIs (including the investigational ones) will probably be
see 150] and several potent and selective H3-receptor ago- available in the future and will enlarge our therapeutic
nists and antagonists have been synthesized [305]. None armamentarium.
of these compounds has however been tested in humans, As far as the novel PPIs are concerned, tenatoprazole
as yet. Similarly, although endogenous GRP is involved in appears to be a true advance in the acid suppression ther-
the regulation of acid secretion, GRP antagonists (like, for apy. Its long half-life (the longest among the available
instance, BIM-26226 or BW2258U89) have been used in compounds) and long duration of antisecretory action,
men only as a pharmacological tool to gain new insights with no difference between day and night, will allow the
into the physiological role of the peptide rather than as an- drug to go beyond the intrinsic limitations of currently
tisecretory drugs [150]. However, the potential therapeutic available compounds. Thanks to its favorable pharmaco-
applications of these antagonists would be limited by the kinetics, the sodium salt of S-tenatoprazole is being de-
fact that blockade of GRP receptors also affects other GI veloped and the preliminary results indicate that this
functions, such as pancreatic secretion, gallbladder con- drug has the potential to address unmet clinical needs.
traction and gastric as well as intestinal motility [306]. Although some decades have elapsed from the intro-
Of the current approaches to reduce acid secretion, P- duction of an effective and safe antisecretory drug in the
CABs and CCK2-receptor antagonists hold the greatest clinical practice and the therapeutic use of acid suppres-
promise, with several compounds already in clinical tri- sion has stood the test of time, a large number of basic
als. Although the quick onset of action of P-CABs (i.e. a and clinical publications on the topic appear every year
full effect from the first dose) is appealing, the results of in the medical literature. All this ongoing research clearly
phase II studies with one such agent (namely AZD0865) shows that the final chapter on the pharmacological treat-
did not show any advantages over esomeprazole (Sohtell ment of acid-related diseases has not yet been written.
et al., pers. commun.). Furthermore, the development of
this class of drugs, which started in the early of 1990s, has
been very slow, with many compounds abandoned over Acknowledgements
the years for lack of efficacy or safety reasons. The results
We are indebted to Prof. Marcello Tonini (Chairman, Depart-
of clinical trials with soraprazan are therefore eagerly
ment of Physiological and Pharmacological Sciences, University of
awaited to understand whether such agents will have a Pavia, Italy) for the critical reading of the manuscript and his use-
role in the future management of acid-related disorders. ful suggestions. We would also like to thank Dr. Elena Losi (Depart-
Thanks to their limited efficacy and the development of ment of Pharmaceutical Chemistry, University of Parma) for the
tolerance, it is unlikely that CCK2 antagonists will be used invaluable help with the molecular structures of investigational new
drugs.
alone as antisecretory compounds but, rather, their com-

38 Dig Dis 2006;24:11–46 Scarpignato /Pelosini /Di Mario

DDI923.indd 38
References

1 Hersey SJ, Sachs G: Gastric acid secretion. 16 Gisbert JP, Gonzalez L, Calvet X, Roque M, 28 Metz DC, Jensen RT: Advances in gastric an-
Physiol Rev 1995;75:155–189. Gabriel R, Pajares JM: Proton pump inhibitor, tisecretory therapy in Zollinger-Ellison. Front
2 Aihara T, Nakamura E, Amagase K, Tomita clarithromycin and either amoxicillin or nitro- Gastrointest Res 1995;23:240–257.
K, Fujishita T, Furutani K, Okabe S: Pharma- imidazole: a meta-analysis of eradication of 29 Meijer JL, Jansen JB, Lamers CB: Omeprazole
cological control of gastric acid secretion for Helicobacter pylori. Aliment Pharmacol Ther in the treatment of Zollinger-Ellison syndrome
the treatment of acid-related peptic disease: 2000;14:1319–1328. and histamine H2-antagonist refractory ulcers.
past, present, and future. Pharmacol Ther 17 Vallve M, Vergara M, Gisbert JP, Calvet X: Digestion 1989;44(suppl 1):31–39.
2003;98:109–127. Single vs. double dose of a proton pump in- 30 Bell NJ, Burget D, Howden CW, Wilkinson J,
3 Lindstrom E, Chen D, Norlen P, Andersson K, hibitor in triple therapy for Helicobacter pylori Hunt RH: Appropriate acid suppression for
Hakanson R: Control of gastric acid secretion: eradication: a meta-analysis. Aliment Pharma- the management of gastro-oesophageal reflux
the gastrin-ECL cell-parietal cell axis. Comp col Ther 2002;16:1149–1156. disease. Digestion 1992;51(suppl 1):59–67.
Biochem Physiol A Mol Integr Physiol 2001; 18 Malfertheiner P, Mégraud F, O’Morain C, 31 Bell NJ, Hunt RH: Role of gastric acid suppres-
128:505–514. Hungin APS, Jones R, Axon A, Graham DY, sion in the treatment of gastro-oesophageal re-
4 Makhlouf GM, Schubert ML: Gastric somato- Tytgat G, the European Helicobacter pylori flux disease. Gut 1992;33:118–124.
statin: a paracrine regulator of acid secretion. Study Group (EHPSG): Current concepts in 32 Chiba N, De Gara CJ, Wilkinson JM, Hunt
Metabolism 1990;39(suppl 2):138–142. the management of Helicobacter pylori infec- RH: Speed of healing and symptom relief in
5 Chey WY, Chang TM: Neural control of the tion – the Maastricht 2-2000 Consensus Re- grade II–IV gastroesophageal reflux disease: a
release and action of secretin. J Physiol Phar- port. Aliment Pharmacol Ther 2002; 16: 167– meta-analysis. Gastroenterology 1997; 112:
macol 2003;54(suppl 4):105–112. 180. 1798–1810.
6 Olbe L, Carlsson E, Lindberg P: A proton- 19 Malfertheiner P, Mégraud F, O’Morain C: 33 Van Pinxteren B, Numans ME, Bonis PA, Lau
pump inhibitor expedition: the case histories Guidelines for the management of Helico- J: Short-term treatment with proton pump in-
of omeprazole and esomeprazole. Nat Rev bacter pylori infection – summary of the Maas- hibitors, H2-receptor antagonists and prokinet-
Drug Discov 2003;2:132–139. tricht-3 2005 Consensus Report. Eur Gastro- ics for gastro-oesophageal reflux disease-like
7 Stedman CAM, Barclay ML: Comparison of enterol Rev 2005, pp 59–62 [http://www. symptoms and endoscopy negative reflux dis-
the pharmacokinetics, acid suppression and ef- touchbriefings.com/cdps/cditem.cfm?nid= ease (Cochrane Review). Cochrane Database
ficacy of proton pump inhibitors. Aliment 1489] Syst Rev 2004;4:CD002095.
Pharmacol Ther 2000;14:963–978. 20 Marshall BJ, Warren JR: Unidentified curved 34 Scarpignato C, Pelosini I: Prevention and
8 Sachs G: Proton pump inhibitors and acid-re- bacilli in the stomach of patients with gastritis treatment of non-steroidal anti-inflammatory
lated diseases. Pharmacotherapy 1997;17:22– and peptic ulceration. Lancet 1984;i:1311– drug-induced gastro-duodenal damage: ratio-
37. 1315. nale for the use of antisecretory compounds.
9 Kromer W: Relative efficacies of gastric pro- 21 Marshall BJ: The 1995 Albert Lasker Medical Ital J Gastroenterol Hepatol 1999;31(suppl 1):
ton-pump inhibitors on a milligram basis: de- Research Award. Helicobacter pylori. The etio- S63–S72.
sired and undesired SH reactions. Impact of logic agent for peptic ulcer. JAMA 1995; 274: 35 Koch M, Dezi A, Ferrario F, Capurso I: Pre-
chirality. Scand J Gastroenterol 2001;36(suppl 1064–1066. vention of nonsteroidal anti-inflammatory
234):3–9. 22 Tytgat GNJ, Noach LA, Rauws EAJ: Helico- drug-induced gastrointestinal mucosal injury.
10 Sachs G, Shin JM, Briving C, Wallmark B, bacter pylori infection and duodenal ulcer dis- A meta-analysis of randomized controlled clin-
Hersey S: The pharmacology of the gastric acid ease. Gastroenterol Clin North Am 1993; 22: ical trials. Arch Intern Med 1996; 156: 2321–
pump: the H+,K+-ATPase. Annu Rev Pharma- 127–139. 2332.
col Toxicol 1995;35:277–305. 23 Leodolter A, Kulig M, Brasch H, Meyer-Sa- 36 Leandro G, Pilotto A, Franceschi M, Bertin T,
11 Wolfe MM, Sachs G: Acid suppression: opti- bellek W, Willich SN, Malfertheiner P: A meta- Lichino E, Di Mario F: Prevention of acute
mizing therapy for gastroduodenal ulcer heal- analysis comparing eradication, healing and NSAID-related gastroduodenal damage: a
ing, gastroesophageal reflux disease, and stress- relapse rates in patients with Helicobacter py- meta-analysis of controlled clinical trials. Dig
related erosive syndrome. Gastroenterology lori-associated gastric or duodenal ulcer. Ali- Dis Sci 2001;46:1924–1936.
2000;118(suppl 1):S9–S31. ment Pharmacol Ther 2001;15:1949–1958. 37 Rostom A, Dube C, Wells G, Tugwell P, Welch
12 Burget DW, Chiverton SG, Hunt RH: Is there 24 Ford AC, Delaney BC, Forman D, Moayyedi V, Jolicoeur E, McGowan J: Prevention of
an optimal degree of acid suppression for heal- P: Eradication therapy in Helicobacter pylori- NSAID-induced gastroduodenal ulcers. Co-
ing of duodenal ulcers? A model of the relation- positive peptic ulcer disease: systematic review chrane Database Syst Rev 2002;4:CD002296.
ship between ulcer healing and acid suppres- and economic analysis. Am J Gastroenterol 38 Hooper L, Brown TJ, Elliott R, Payne K, Rob-
sion. Gastroenterology 1990;99:345–351. 2004;99:1833–1855. erts C, Symmons D: The effectiveness of five
13 Poynard T, Lemaire M, Agostini H: Meta- 25 Bazzoli F, BianchiPorro G, Bianchi MG, Mol- strategies for the prevention of gastrointestinal
analysis of randomized clinical trials compar- teni M, Pazzato P, Zagari RM: Treatment of toxicity induced by non-steroidal anti-inflam-
ing lansoprazole with ranitidine or famotidine Helicobacter pylori infection. Indications and matory drugs: systematic review. BMJ 2004;
in the treatment of acute duodenal ulcer. Eur J regimens: an update. Dig Liver Dis 2002; 34: 329:948.
Gastroenterol Hepatol 1995;7:661–665. 70–83. 39 Berstad A: Does profound acid inhibition im-
14 Salas M, Ward A, Caro J: Are proton pump 26 Gisbert JP, Pajares JM: Systematic review and prove haemostasis in peptic ulcer bleeding?
inhibitors the first choice for acute treatment meta-analysis: is one-week proton pump inhib- Scand J Gastroenterol 1997;32:396–398.
of gastric ulcers? A meta analysis of random- itor-based triple therapy sufficient to heal pep- 40 Geun WP: Are there indications for intrave-
ized clinical trials. BMC Gastroenterol 2002;2: tic ulcer? Aliment Pharmacol Ther 2005; 21: nous acid inhibition in the prevention and
17–23. 795–804. treatment of upper GI bleeding? Scand J Gas-
15 Scarpignato C, Pelosini I: Antisecretory drugs 27 Metz DC, Pisegna JR, Fishbeyn VA, Benya troenterol 2000;35(suppl 232):10–20.
for eradication of Helicobacter pylori: antibac- RV, Jensen RT: Control of gastric acid hyper- 41 Zed PJ, Loewen PS, Slavik RS, Marra CA:
terial activity and synergism with antimicro- secretion in the management of patients with Meta-analysis of proton pump inhibitors in
bial agents. Progr Basic Clin Pharmacol 1999; Zollinger-Ellison syndrome. World J Surg treatment of bleeding peptic ulcers. Ann Phar-
11:136–180. 1993;17:468–480. macother 2001;35:1528–1534.

Update on Acid Suppression Therapy Dig Dis 2006;24:11–46 39

DDI923.indd 39 02.05.2006 16:25:01


42 Gisbert JP, Gonzalez L, Calvet X, Roque M, 55 Scarpignato C, Bruley des Varannnes S: Traite- 68 Guda N, Mueller R, Vakil N: The effect of
Gabriel R, Pajares JM: Proton pump inhibi- ment médical du RGO par les antagonistes des over-the-counter ranitidine (75 mg) on night-
tors versus H2-antagonists: a meta-analysis of récepteurs H2 de l’histamine. Gastroenterol time heartburn in patients with erosive esoph-
their efficacy in treating bleeding peptic ulcer. Clin Biol 1999;23:S78–S89. agitis on daily proton pump inhibitor mainte-
Aliment Pharmacol Ther 2001;15:917–926. 56 Wilder-Smith CH, Merki HS: Tolerance dur- nance therapy. Gastroenterology 2003; 124:
43 Levine JE, Leontiadis GI, Sharma VK, How- ing dosing with H2-receptor antagonists. An 539A.
den CW: Meta-analysis: the efficacy of intrave- overview. Scand J Gastroenterol 1992;27(sup- 69 Orr WC: Night-time gastro-oesophageal reflux
nous H2-receptor antagonists in bleeding pep- pl 193):14–19. disease: prevalence, hazards, and manage-
tic ulcer. Aliment Pharmacol Ther 2002; 16: 57 Sandvik AK, Brenna E, Waldum HL: Review ment. Eur J Gastroenterol Hepatol 2005; 17:
1137–1142. article: the pharmacological inhibition of gas- 113–120.
44 Selby NM, Kubba AK, Hawkey CJ: Acid sup- tric acid secretion: tolerance and rebound. Ali- 70 Tutuian R, Katz PO, Castell DO: Nocturnal
pression in peptic ulcer haemorrhage: a ‘meta- ment Pharmacol Ther 1997;11:1013–1018. acid breakthrough: pH, drugs and bugs. Eur J
analysis’. Aliment Pharmacol Ther 2000; 14: 58 Scarpignato C: Antisecretory drugs, Helico- Gastroenterol Hepatol 2004;16:441–443.
1119–1126. bacter pylori infection and symptom relief in 71 Rackoff A, Agrawal A, Hila A, Mainie I, Tutu-
45 Leontiadis GI, Sharma VK, Howden CW: Sys- GORD: still an unexplored triangle. Dig Liver ian R, Castell DO: Histamine-2 receptor an-
tematic review and meta-analysis of proton Dis 2005;37:468–474. tagonists at night improve gastroesophageal
pump inhibitor therapy in peptic ulcer bleed- 59 Redstone HA, Barrowman N, Veldhuyzen reflux disease symptoms for patients on proton
ing. BMJ 2005;330:568. Van Zanten SJ: H2-receptor antagonists in the pump inhibitor therapy. Dis Esophagus 2005;
46 Leontiadis GI, Sharma VK, Howden CW: Sys- treatment of functional (nonulcer) dyspepsia: 18:370–373.
tematic review and meta-analysis: proton- a meta-analysis of randomized controlled clin- 72 Katz PO, Tutuian R: Histamine receptor an-
pump inhibitor treatment for ulcer bleeding ical trials. Aliment Pharmacol Ther 2000; 15: tagonists, proton pump inhibitors and their
reduces transfusion requirements and hospital 1291–1299. combination in the treatment of gastro-oe-
stay – results from the Cochrane Collabora- 60 Zacny J, Zamakhshary M, Sketris I, Veld- sophageal reflux disease. Best Pract Res Clin
tion. Aliment Pharmacol Ther 2005; 22: 169– huyzen van Zanten S: Systematic review: the Gastroenterol 2001;15:371–384.
174. efficacy of intermittent and on-demand thera- 73 McColl KEL, Kennerley P: Proton pump in-
47 Bardou M, Toubouti Y, Benhaberou-Brun D, py with histamine H2-receptor antagonists or hibitors – differences emerge in the hepatic
Rahme E, Barkun AN: Meta-analysis: proton- proton pump inhibitors for gastro-oesophageal metabolism. Digest Liver Dis 2002; 34: 461–
pump inhibition in high-risk patients with reflux disease patients. Aliment Pharmacol 467.
acute peptic ulcer bleeding. Aliment Pharma- Ther 2005;21:1299–1312. 74 Thomson ABR: Are the orally administered
col Ther 2005;21:677–686. 61 Earnest D, Robinson M, Rodriguez-Stanley S, proton pump inhibitors equivalent? A compar-
48 Khuroo MS, Khuroo MS, Farahat KL, Kagevi Ciociola AA, Jaffe P, Silver MT, Kleoudis CS, ison of lansoprazole, omeprazole, pantopra-
IE: Treatment with proton pump inhibitors in Murdock RH: Managing heartburn at the zole, and rabeprazole. Curr Gastroenterol Rep
acute non-variceal upper gastrointestinal ‘base’ of the GERD ‘iceberg’: effervescent ra- 2000;2:482–493.
bleeding: a meta-analysis. J Gastroenterol nitidine 150 mg b.d. provides faster and better 75 Langman MJS: Which PPI? Gut 2001;49:309–
Hepatol 2005;20:11–25. heartburn relief than antacids. Aliment Phar- 310.
49 Leontiadis GI, Sharma VK, Howden CW: Sys- macol Ther 2000;14:911–918. 76 Kromer W, Horbach S, Lühmann R: Relative
tematic review and meta-analysis: enhanced 62 Robinson M, Rodriguez-Stanley S, Ciociola efficacies of gastric proton pump inhibitors:
efficacy of proton-pump inhibitor therapy for AA, Filinto J, Zubaidi S, Miner PB Jr, Gardner their clinical and pharmacological basis. Phar-
peptic ulcer bleeding in Asia – a post-hoc anal- JD: Synergy between low-dose ranitidine and macology 1999;59:57–77.
ysis from the Cochrane Collaboration. Aliment antacid in decreasing gastric and oesophageal 77 Guidance on the use of proton pump inhibitors
Pharmacol Ther 2005;21:1055–1061. acidity and relieving meal-induced heartburn. in the treatment of dyspepsia: NICE, UK:
50 Chong E, Ensom MH: Pharmacogenetics of the Aliment Pharmacol Ther 2001; 15: 1365– Technology Appraisal Guidance No 7, 2000
proton pump inhibitors: a systematic review. 1374. [http://www. NICE.org.uk]
Pharmacotherapy 2003;23:460–471. 63 Peghini PL, Katz PO, Bracy NA, Castell DO: 78 Bardhan KD: Is there any acid peptic disease
51 Furuta T, Shirai N, Sugimoto M, Ohashi K, Nocturnal recovery of gastric acid secretion that is refractory to proton pump inhibitors?
Ishizaki T: Pharmacogenomics of proton with twice-daily dosing of proton pump inhib- Aliment Pharmacol Ther 1993;7(suppl 1):13–
pump inhibitors. Pharmacogenomics 2004; 5: itors. Am J Gastroenterol 1998;9:763–767. 31.
181–202. 64 Xue S, Katz PO, Banerjee P, Tutuian R, Castell 79 Amin MR, Postma GN, Johnson P, Digges N,
52 Andriulli A, Annese V, Caruso N, Pilotto A, DO: Bedtime H2 blockers improve nocturnal Koufman JA: Proton pump inhibitor resis-
Accadia L, Niro AG, Quitadamo M, Merla A, gastric acid control in GERD patients on pro- tance in the treatment of laryngopharyngeal
Fiorella S, Leandro G: Proton-pump inhibitors ton pump inhibitors. Aliment Pharmacol Ther reflux. Otolaryngol Head Neck Surg 2000;125:
and outcome of endoscopic hemostasis in 2001;15:1351–1356. 374–378.
bleeding peptic ulcers: a series of meta-analy- 65 Fackler WK, Ours TM, Vaezi MF, Richter JE: 80 Sontag SJ: Persistent gastroesophageal reflux
ses. Am J Gastroenterol 2005;100:207–219. Long-term effect of H2RA therapy on noctur- disease symptoms on standard proton-pump
53 Gisbert J, Khorrami S, Carballo F, Calvet X, nal gastric acid breakthrough. Gastroenterol- inhibitor therapy. Gastroenterol Clin North
Gene E, Dominguez-Munoz J: H. pylori eradi- ogy 2002;122:625–632. Am 2002;31(suppl 4):S77–S84.
cation therapy vs. antisecretory non-eradica- 66 Cross LB, Justice LN: Combination drug ther- 81 Galmiche JP, Sacher-Huvelin S: Reflux gastro-
tion therapy (with or without long-term main- apy for gastroesophageal reflux disease. Ann œsophagien résistant aux inhibiteurs de la
tenance antisecretory therapy) for the Pharmacother 2002;36:912–916. pompe à protons. Hépato-Gastro 2002;9:159–
prevention of recurrent bleeding from peptic 67 Pan T, Wang Y, Guo Z, Wang Q: Additional 163.
ulcer. Cochrane Database Syst Rev 2003; 4: bedtime H2-receptor antagonist for the control 82 Ashida K, Sakaguchi M, Tanaka M, Takiuchi
CD004062. of nocturnal gastric acid breakthrough. Co- H, Egashira Y, Katsu K: Clinical study on the
54 Colin-Jones DG: The role and limitations of chrane Database Syst Rev 2004;4:CD004275. pathophysiology and treatment of PPI-resis-
H2-receptor antagonists in the treatment of tant ulcers. J Clin Gastroenterol 1995;20(suppl
gastro-oesophageal reflux disease. Aliment 2):S67–S71.
Pharmacol Ther 1995;9(suppl 1):9–14. 83 Katz PO: Lessons learned from intragastric pH
monitoring. J Clin Gastroenterol 2001; 33:
107–113.

40 Dig Dis 2006;24:11–46 Scarpignato /Pelosini /Di Mario

DDI923.indd 40 02.05.2006 16:25:02


84 Hatlebakk JG, Katz PO, Castell DO: Medical 101 Stern L: The lesson of thalidomide. Clin 120 Andersson T, Bredberg E, Sunzel M, Antons-
therapy. Management of the refractory pa- Pharmacol Ther 1973;14:660–661. son M, Weidolf L: Pharmacokinetics (PK)
tient. Gastroenterol Clin North Am 1999;28: 102 Richardson P, Hideshima T, Anderson K: and effect on pentagastrin stimulated peak
847–860. Thalidomide: emerging role in cancer medi- acid output of omeprazole and its two optical
85 Leite L, Lambrecht N, Sachs G, Castell D, cine. Annu Rev Med 2002;53:629–657. isomers, S-omeprazole/esomeprazole (E) and
Lagerstrom P: Is omeprazole resistance due 103 Williams K, Lee E: Importance of drug enan- R-omeprazole. Gastroenterology 2000; 118
to mutations of cysteine 813 or 822 in the acid tiomers in clinical pharmacology. Drugs (suppl 2):A1210.
pump? Gastroenterology 1995;108:A147. 1985;30:333–354. 121 Junghard O, Hassan-Alin M, Hasselgren G:
86 Leite LP, Just RJ, Castell DO, Lagerstrom 104 Lam Y: Stereoselectivty: an issue of signifi- The effect of the area under the plasma con-
PO: Control of gastric acid with high dose H2- cant importance in clinical pharmacology. centration vs. time curve and the maximum
receptor antagonists after omeprazole failure: Pharmacotherapy 1988;8:147–157. plasma concentration of esomeprazole on in-
report of two cases. Am J Gastroenterol 1995; 105 Hutt AJ, Tan SC: Drug chirality and its clinical tragastric pH. Eur J Clin Pharmacol 2002;58:
90:1874–1877. significance. Drugs 1996;52(suppl 57):1–12. 453–458.
87 Huber R, Kohl B, Sachs G, Senn-Bilfinger J, 106 Pifferi G, Perucca E: The cost-benefit ratio of 122 Tonini M, Vigneri S, Savarino V, Scarpignato
Simon WA, Sturm E: Review article: the con- enantiomeric drugs. Eur J Drug Metab Phar- C: Clinical pharmacology and safety profile of
tinuing development of proton pump inhibi- macokinet 1995;20:15–25. esomeprazole, the first enantiomerically pure
tors with particular reference to pantopra- 107 Eichelbaum M: Enantiomers: implications proton pump inhibitor. Dig Liver Dis 2001;
zole. Aliment Pharmacol Ther 1995; 9: and complications in developmental pharma- 33:600–606.
363–378. cology. Dev Pharmacol Ther 1992; 18: 131– 123 Vigneri S, Tonini M, Scarpignato C, Savarino
88 Tytgat GN: Shortcomings of the first-genera- 134. V: Improving opportunities for effective
tion proton pump inhibitors. Eur J Gastroen- 108 Lee EJ, Williams KM: Chirality. Clinical management of gastro-esophageal reflux dis-
terol Hepatol 2001;13(suppl 1):S29–S33. pharmacokinetic and pharmacodynamic ease. Dig Liver Dis 2001;33:720–730.
89 Robinson M: New-generation proton pump considerations. Clin Pharmacokinet 1990; 124 Scott LJ, Dunn CJ, Mallarkey G, Sharpe M:
inhibitors: overcoming the limitations of ear- 18:339–345. Esomeprazole. A review of its use in the man-
ly-generation agents. Eur J Gastroenterol 109 Brocks DR, Jamali F: Stereochemical aspects agement of acid-related disorders. Drugs
Hepatol 2001;13(suppl 1):S43–S47. of pharmacotherapy. Pharmacotherapy 1995; 2002;62:1503–1538.
90 Sachs G: Improving on PPI-based therapy of 15:551–564. 125 Johnson DA: Review of esomeprazole in the
GORD. Eur J Gastroenterol Hepatol 2001; 110 Scott AK: Stereoisomers and drug toxicity. treatment of acid disorders. Expert Opin
13(suppl 1):S35–S41. The value of single stereoisomer therapy. Pharmacother 2003;4:253–264.
91 Anonymus: Tenatoprazole. Benatoprazole, Drug Saf 1993;8:149–159. 126 Kim KA, Shon JH, Park JY, Yoon YR, Kim
TU-199. Drugs RD 2002;3:276–277. 111 Hsu WH: Toxicity and drug interactions of MJ, Yun DH, Kim MK, Cha IJ, Hyun MH,
92 Tonini M, De Giorgio R, De Ponti F: Novel levamisole. J Am Vet Med Assoc 1980; 176: Shin JG: Enantioselective disposition of lan-
therapeutic strategies in acid-related disor- 1166–1169. soprazole in extensive and poor metabolizers
ders. Expert Opin Ther Patents 2003;13:639– 112 Marzo A, Cardace G, Arrigoni Martelli E: En- of CYP2C19. Clin Pharmacol Ther 2002;72:
649. zymes in stereoselective pharmacokinetics of 90–99.
93 Andersson K, Carlsson E: Potassium-com- endogenous substances. Chirality 1992; 4: 127 Kim KA, Kim MJ, Park JY, Shon JH, Yoon
petitive acid blockade: a new therapeutic 247–251. YR, Lee SS, Liu KH, Chun JH, Hyun MH,
strategy in acid-related diseases. Pharmacol 113 Erlandsson P, Isaksson R, Lorentzon P, Lind- Shin JG: Stereoselective metabolism of lan-
Ther 2005;108:294–307. berg P: Resolution of the enantiomers of soprazole by human liver cytochrome P450
94 Geibel JP: Role of potassium in acid secre- omeprazole and some of its analogues by liq- enzymes. Drug Metab Dispos 2003;31:1227–
tion. World J Gastroenterol 2005; 11: 5259– uid chromatography on a trisphenylcarbamo- 1234.
5265. ylcellulose-based stationary phase. The effect 128 Miura M, Tada H, Yasui-Furukori N, Uno T,
95 Parsons ME, Garner A: Drug development in of the enantiomers of omeprazole on gastric Sugawara K, Tateishi T, Suzuki T: Effect of
gastroenterology – the changing view of in- glands. J Chromatogr 1990;532:305–319. clarithromycin on the enantioselective dispo-
dustry. Aliment Pharmacol Ther 1995; 9: 114 Eichelbaum M: Pharmacokinetic and phar- sition of lansoprazole in relation to CYP2C19
457–463. macodynamic consequences of stereoselec- genotypes. Chirality 2005;17:338–344.
96 Hassan-Alin M, Niazi M, Röhss K, Sthenoff tive drug metabolism in man. Biochem Phar- 129 Tanaka M, Yamazaki H, Hakusui H, Naka-
H: Esomeprazole, the S-isomer of omepra- macol 1988;37:93–96. michi N, Sekino H: Differential stereoselec-
zole, is optically stable in humans. Gastroen- 115 Tucker GT, Lennard MS: Enantiomer-spe- tive pharmacokinetics of pantoprazole, a pro-
terology 2000;118(suppl 2):A1244–A1245. cific pharmacokinetics. Pharmacol Ther ton pump inhibitor in extensive and poor
97 The Nobel Foundation: Information for the 1990;45:309–329. metabolizers of pantoprazole – a preliminary
public. The 2001 Nobel Prize in Chemis- 116 Andersson T: Pharmacokinetics, metabolism study. Chirality 1997;9:17–21.
try [www.nobel.se/chemistry/laureates/2001/ and interactions of acid pump inhibitors – fo- 130 Cao H, Wang MW, Sun LX, Ikejima T, Hu
public.htlm]. cus on omeprazole, lansoprazole and panto- ZQ, Zhao WH: Pharmacodynamic compari-
98 Cotton H, Elebring T, Larsson M, Li L, Sö- prazole. Clin Pharmacokinet 1996;31:9–28. son of pantoprazole enantiomers: inhibition
rensen H, von Unge S: Asymmetric synthesis 117 Egan LJ, Murray JA: New perspectives in gas- of acid-related lesions and acid secretion in
of esomeprazole. Tetrahedron Asymmetry tric acid suppression: genetic polymorphisms rats and guinea-pigs. J Pharm Pharmacol
2000;11:3819–3825. predict the efficacy of proton pump inhibi- 2005;57:923–927.
99 Del Nozal MJ, Toribio L, Bernal JL, Alonso tors. Dig Dis 2000;18:58–63. 131 Mano N, Oda Y, Takakuwa S, Chiku S, Na-
C, Jimenez JJ: Chiral separation of omepra- 118 Tybring G, Bottiger Y, Widen J, Bertilsson L: kata H, Asakawa N: Plasma direct injection
zole and several related benzimidazoles using Enantioselective hydroxylation of omeprazole high-performance liquid chromatographic
supercritical fluid chromatography. J Sep Sci catalyzed by CYP2C19 in Swedish white sub- method for simultaneously determining
2004;27:1023–1029. jects. Clin Pharmacol Ther 1997;62:129–137. E3810 enantiomers and their metabolites by
100 Lough JW, Wainer I: Chirality in Natural and 119 Äbelö A, Andersson TB, Bredberg U, Skan- using flavoprotein-conjugated column. J
Applied Sciences. Oxford, Blackwell Publish- berg I, Weidolf L: Stereoselective metabolism Pharm Sci 1996;85:903–907.
ing, 2002, pp 1–336. by human liver CYP enzymes of a substituted
benzimidazole. Drug Metab Dispos 2000;28:
58–64.

Update on Acid Suppression Therapy Dig Dis 2006;24:11–46 41

DDI923.indd 41 02.05.2006 16:25:02


132 TAP 2005 Press Release: TAP initiates phase 148 Anonymous: Omeprazole/antacid-powder 164 Castell D, Bagin R, Goldlust B, Major J, Hep-
III studies for investigational new gastroen- suspension-Santarus: omeprazole/sodium bi- burn B: Comparison of the effects of immedi-
terology product [http://www.tap.com/npr_ carbonate powder-Santarus, SAN 05. Drugs ate-release omeprazole powder for oral sus-
2005_8_30.asp] R&D 2004;5:349–350. pension and pantoprazole delayed-release
133 ClinicalTrials.gov. Ongoing Trials with TAK- 149 Scarpignato C, Pelosini I, Molina E: Acid sup- tablets on nocturnal acid breakthrough in pa-
390MR [http://www.clinicaltrials. gov/ct/ pression in Barrett’s esophagus: why and tients with symptomatic gastro-oesophageal
search;jsessionid=A1CC1E29324CED0D3 how? In Giuli R, Siewert JR, Couturier D, reflux disease. Aliment Pharmacol Ther
E0263C6AD8B0243?term = TAK-390MR]. Scarpignato C (eds): Barrett’s Esophagus. 250 2005;21:1467–1474.
134 Liu KH, Kim MJ, Shon JH, Moon YS, Seol Questions, 250 Answers. Paris, John Libbey 165 Castell D: Review of immediate-release
SY, Kang W, Cha IJ, Shin JG: Stereoselective Eurotext, 2003, vol 2, pp 437–455. omeprazole for the treatment of gastric acid-
inhibition of cytochrome P450 forms by lan- 150 Lehmann F, Hildebrand P, Beglinger C: New related disorders. Expert Opin Pharmacother
soprazole and omeprazole in vitro. Xeno- molecular targets for treatment of peptic ulcer 2005;6:2501–2510.
biotica 2005;35:27–38. disease. Drugs 2003;63:1785–1797. 166 Stollman N, Metz DC: Pathophysiology and
135 Rettie AE, Jones JP: Clinical and toxicologi- 151 Flamel Technologies: Micropump® platform prophylaxis of stress ulcer in intensive care
cal relevance of CYP2C9: drug-drug interac- [http://www.flamel.com/techAndProd/mi- unit patients. J Crit Care 2005;20:35–45.
tions and pharmacogenetics. Annu Rev Phar- cropump.shtml] 167 Metz DC: Preventing the gastrointestinal
macol Toxicol 2005;45:477–494. 152 AGI Therapeutics: Product AG-010 [http:// consequences of stress-related mucosal dis-
136 Andersson T: Single-isomer drugs. True ther- www.agitherapeutics.com/agi010.html] ease. Curr Med Res Opin 2005;21:11–18.
apeutic advances. Clin Pharmacokinet 2004; 153 Freston JW, Kukulka MJ, Lloyd E, Lee C: A 168 Conrad SA, Gabrielli A, Margolis B, Quartin
43:279–285. novel option in proton pump inhibitor dos- A, Hata JS, Frank WO, Bagin RG, Rock JA,
137 Raghunath AS, Green JR, Edwards SJ: A re- ing: lansoprazole orally disintegrating tablet Hepburn B, Laine L: Randomized, double-
view of the clinical and economic impact of dispersed in water and administered via na- blind comparison of immediate-release
using esomeprazole or lansoprazole for the sogastric tube. Aliment Pharmacol Ther omeprazole oral suspension versus intrave-
treatment of erosive esophagitis. Clin Ther 2004;20:407–411. nous cimetidine for the prevention of upper
2003;25:2088–2101. 154 Horn JR, Howden CW: Similarities and dif- gastrointestinal bleeding in critically ill pa-
138 Anonymous: Do single stereoisomer drugs ferences among delayed-release proton-pump tients. Crit Care Med 2005;33:760–765.
provide value? Ther Initiative 2002; 45 inhibitor formulations. Aliment Pharmacol 169 Zimbaro G, Ferraù O, Familiari L, Consolo
(June–Sept letter) [http://www.ti.ubc.ca/pag- Ther 2005;22(suppl 3):20–24. F: Endogenous gastrin and insulin secretion
es/letter45.htm] 155 Maroni A, Zema L, Cerea M, Sangalli ME: in humans: effect of gastric lavage with sodi-
139 Anonymous: Prilosec, Nexium and stereoiso- Oral pulsatile drug delivery systems. Expert um bicarbonato (in Italian). Boll Soc Ital Biol
mers. Med Lett 2003;45:49–52. Opin Drug Deliv 2005;2:855–871. Sper 1973;49:896–901.
140 Mansfield P, Henry D, Tonking A: Single-en- 156 Baldi F, Malfertheiner P: Lansoprazole fast 170 Feurle GE: The action of antacids on serum
antiomer drugs. Elegant science, disappoint- disintegrating tablet: a new formulation for gastrin concentrations in man. Klin Wochen-
ing results. Clin Pharmacokinet 2004; 43: an established proton pump inhibitor. Diges- schr 1977;55:1039–1042.
287–290. tion 2003;67:1–5. 171 Saito E, Matsuo Y: Inhibitory effect of lemi-
141 Henry DA, Lexchin J: Medicines, health and 157 Baldi F: Lansoprazole oro-dispersible tablet: noprazole on acid secretion in parietal cells
commerce: a global perspective of the phar- pharmacokinetics and therapeutic use in isolated from guinea pig gastric mucosa. Jpn
maceutical industry: industry as medicine acid-related disorders. Drugs 2005;65:1419– J Pharmacol 1995;68:19–23.
provider. Lancet 2002;360:1590–1595. 1426. 172 Takahashi S, Okabe S: The cytoprotective ef-
142 Bardhan KD, Williamson M, Royston C, 158 Freston JW, Chiu YL, Mulford DJ, Ballard fect of leminoprazole on indomethacin-in-
Lyon C: Admission rates for peptic ulcer in ED: Comparative pharmacokinetics and duced damage to rabbit gastric mucosal cells.
the Trent region, UK, 1972–2000. Changing safety of lansoprazole oral capsules and orally J Pharmacol Exp Ther 1996;279:975–982.
pattern, a changing disease? Dig Liver Dis disintegrating tablets in healthy subjects. Ali- 173 Takahashi S, Okabe S: Mechanism by which
2004;36:577–588. ment Pharmacol Ther 2003;17:361–367. orally administered leminoprazole stimulates
143 Fries JF, Murtagh KN, Bennett M, Zatarain 159 Baldi F, Ghersi S, Cavoli C, Torresan F: A mucus synthesis in rats. Pharmacology 1998;
E, Lingala B, Bruce B: The rise and decline of comparison of the new lansoprazole orally 57:47–56.
nonsteroidal anti-inflammatory drug-associ- disintegrating tablets with esomeprazole in 174 Kwon D, Chae JB, Park CW, Kim YS, Lee
ated gastropathy in rheumatoid arthritis. Ar- patients with non-erosive reflux disease. Gas- SM, Kim EJ, Huh IH, Kim DY, Cho KD: Ef-
thritis Rheum 2004;50:2433–2440. troenterology 2005; 128(suppl 2):A523– fects of IY-81149, a newly developed proton
144 Tytgat GN: Are there unmet needs in acid A524. pump inhibitor, on gastric acid secretion in
suppression? Best Pract Res Clin Gastroen- 160 Santarus Inc. Corporate [http://www.santa- vitro and in vivo. Arzneimittelforschung
terol 2004;18(suppl):67–72. rus.com/corporate]. 2001;51:204–213.
145 Hunt RH: The unmet needs in delayed-re- 161 Hepburn B, Goldlust B: Comparative effects 175 Kim EJ, Lee RK, Lee SM, Kim DY: General
lease proton-pump inhibitor therapy in 2005. of an omeprazole antacid complex-immedi- pharmacology of IY-81149, a new proton
Aliment Pharmacol Ther 2005; 22(suppl ate release and omeprazole delayed-release pump inhibitor. Arzneimittelforschung 2001;
3):10–19. on omeprazole pharmacokinetics and gastric 51:51–59.
146 Pezanoski J, Guanaratnam N, Cowen M: pH in healthy subjects. Gastroenterology 176 Kil BJ, Kim IW, Shin CY, Jeong JH, Jun CH,
Correct and incorrect dosing of proton pump 2003;124(suppl):A228. Lee SM, Kim DY, Huh IH, Sohn UD: Com-
inhibitors and its impact on GERD symp- 162 Howden CW: Review article: immediate-re- parison of IY-81149 with omeprazole in rat
toms. Gastroenterology 2003; 124(suppl): lease proton-pump inhibitor therapy – poten- reflux oesophagitis. J Auton Pharmacol 2000;
A228. tial advantages. Aliment Pharmacol Ther 20:291–296.
147 Chey WD, Inadomi JM, Booher AM, Sharma 2005;22(suppl 3):25–30. 177 Periclou AP, Goldwater R, Lee SM, Park
VK, Fendrick AM, Howden CW: Primary- 163 Tutuian R, Castell DO: Nocturnal acid break- DW, Kim DY, Cho KD, Boileau F, Jung WT:
care physicians’ perceptions and practices on through – approach to management. Med- A comparative pharmacodynamic study of
the management of GERD: results of a na- GenMed 2004; 6: 11 [http://www.medscape. IY-81149 versus omeprazole in patients with
tional survey. Am J Gastroenterol 2005;100: com/viewarticle/490723]. gastroesophageal reflux disease. Clin Phar-
1237–1242. macol Ther 2000;68:304–311.

42 Dig Dis 2006;24:11–46 Scarpignato /Pelosini /Di Mario

DDI923.indd 422
178 Goldwater R, Lee SM, Chung GJ, Kim DY, 190 Galmiche JP, Sacher-Huvelin S, Bruley des 204 Yu KS, Bae KS, Shon JH, Cho JY, Yi SY,
Cho KD, Boileau F: A phase I, randomized, Varannes S, Vavasseur F, Taccoen A, Fioren- Chung JY, Lim HS, Jang IJ, Shin SG, Song
parallel placebo-controlled safety, tolerance tini P, Homerin M: A comparative study of KS, Moon BS: Pharmacokinetic and pharma-
and pharmacokinetic study of multiple doses the early effects of tenatoprazole 40 mg and codynamic evaluation of a novel proton
of IY-8119 in fasting male volunteers. Clin esomeprazole 40 mg on intragastric pH in pump inhibitor, YH1885, in healthy volun-
Pharmacol Ther 1999;65:126(abstr PI-35). healthy volunteers. Aliment Pharmacol Ther teers. J Clin Pharmacol 2004;44:73–82.
179 Uchiyama K, Wakatsuki D, Kakinoki B, 2005;21:575–582. 205 Nilsson C, Albrektson E, Rydholm H, Rohss
Takeuchi Y, Araki T, Morinaka Y: Effects of 191 Hunt RH, Armstrong D, James C, Chowd- K, Hassan-Alin M, Hasselgren G: Tolerabil-
TU-199, a novel H+,K+-ATPase inhibitor, on hury SK, Yuan Y, Fiorentini P, Taccoen A, ity, pharmacokinetics and effects on gastric
gastric acid secretion and gastroduodenal ul- Cohen P: Effect on intragastric pH of a PPI acid secretion after single oral doses of the
cers in rats. Methods Find Exp Clin Pharma- with a prolonged plasma half-life: compari- potassium-competitive acid blocker
col 1999;21:115–122. son between tenatoprazole and esomeprazole AZD0865 in healthy male subjects. Gastro-
180 Shin JM, Homerin M, Domagala F, Ficheux on the duration of acid suppression in healthy enterology 2005;128(suppl 2):A528.
H, Sachs G: Characterization of the inhibi- male volunteers. Am J Gastroenterol 2005; 206 Park S, Song K, Moon BS, Joo S, Choi M,
tory activity of tenatoprazole on the gastric 100:1949–1956. Kim J, Choi K, Chung I, Sun H: The effect of
H+,K+-ATPase in vitro and in vivo. Biochem 192 Lind T, Rydberg L, Kyleback A, Jonsson A, 7 days of dosing with YH1885 on 24-hour
Pharmacol 2006;71:837–849. Andersson T, Hasselgren G, Holmberg J, intragastric acidity and plasma gastrin con-
181 Shin JM, Cho YM, Sachs G: Chemistry of Rohss K: Esomeprazole provides improved centration in healthy male subjects. Gut
covalent inhibition of the gastric (H+,K+)- acid control vs. omeprazole in patients with 2002;51(suppl 3):A167.
ATPase by proton pump inhibitors. J Am symptoms of gastro-oesophageal reflux dis- 207 Scarpignato C, Pelosini I: Antisecretory drugs
Chem Soc 2004;126:7800–7811. ease. Aliment Pharmacol Ther 2000;14:861– for eradication of Helicobacter pylori: anti-
182 Waldum HL, Lehy T, Brenna E, Sandvik AK, 867. bacterial activity and synergism with antimi-
Petersen H, Sognen BS, Bonfils S, Lewin MJ: 193 Bentley R: Role of sulfur chirality in the crobial agents. Progr Basic Clin Pharmacol
Effect of the histamine-1 antagonist astem- chemical processes of biology. Chem Soc Rev 1999;11:135–178.
izole alone or with omeprazole on rat gastric 2005;34:609–624. 208 Martinek J, Blum AL, Stolte M, Hartmann
mucosa. Scand J Gastroenterol 1991;26: 23– 194 Charbit S, Cohen A, Ficheux H, Homerin M, M, Verdu EF, Luhmann R, Dorta G, Wiesel
35. Schutze F, Taccoen A: Enantiomer (–) of te- P: Effects of pumaprazole (BY841), a novel
183 Kakinoki B, Ono C, Yamazaki N, Chikamat- natoprazole and the therapeutic use thereof. reversible proton pump antagonist, and of
su N, Wakatsuki D, Uchiyama K, Morinaka Patent No WO060891, 2004. omeprazole, on intragastric acidity before
Y: General pharmacological properties of the 195 Miner PB Jr, Katz PO, Chen Y, Sostek M: and after cure of Helicobacter pylori infec-
new proton pump inhibitor (+/–)-5-methoxy- Gastric acid control with esomeprazole, lan- tion. Aliment Pharmacol Ther 1999; 3: 27–
2-[[(4-methoxy-3,5-dimethylpyrid-2-yl) soprazole, omeprazole, pantoprazole, and ra- 34.
methyl]sulfi-nyl]-1H-imidazo[4,5-b]pyri- beprazole: a five-way crossover study. Am J 209 Orexo AB: Product Portfolio [http://www.
dine. Methods Find Exp Clin Pharmacol Gastroenterol 2003;98:2616–2620. orexo.com/DynPage.aspx?id = 14994&mn1=
1999;21:179–187. 196 Lorentzon P, Sachs G, Wallmark B: Inhibi- 1542&mn2=1619].
184 Uchiyama K, Wakatsuki D, Kakinoki B, tory effects of cations on the gastric H+,K+- 210 Schulze F, Charby S, Ficheux H, Homerin M,
Takeuchi Y, Araki T, Morinaka Y: The long- ATPase. A potential-sensitive step in the K+ Taccoen A, Inaba Y: Pharmaceutical compo-
lasting effect of TU-199, a novel H+,K+- limb of the pump cycle. J Biol Chem 1988; sition combining tenatoprazole and an hista-
ATPase inhibitor, on gastric acid secretion in 263:10705–10710. mine H2-receptor antagonist. Patent No
dogs. J Pharm Pharmacol 1999;51:457–464. 197 Ene MD, Khan-Daneshmend T, Roberts CJ: W037256, 2004.
185 Negma-Lerads: Tenatoprazole (TU-199) In- A study of the inhibitory effects of SCH 211 Patel SS, Wilde MI: Ebrotidine. Drugs 1996;
vestigator’s Brochure, Sept 2001. 28080 on gastric secretion in man. Br J Phar- 51:974–980.
186 Kim K, Johnson JA, Derendorf H: Differ- macol 1982;76:389–391. 212 Agut J, Sanchez JC, Sacristan A, Ortiz JA:
ences in drug pharmacokinetics between East 198 Beil W, Hackbarth I, Sewing KF: Mechanism Action of ebrotidine, ranitidine and cimeti-
Asians and Caucasians and the role of genet- of gastric antisecretory effect of SCH 28080. dine on the specific binding to histamine H1-
ic polymorphisms. J Clin Pharmacol 2004; Br J Pharmacol 1986;88:19–23. and H2-receptors. Arzneimittelforschung
44:1083–1095. 199 ClinicalTrials.gov. Ongoing trials with AZD 1997;47(suppl 4A):447–449.
187 Furuta T, Shirai N, Sugimoto M, Nakamura 0865 [http://www.clinicaltrials.gov/ct/search; 213 Konturek PC, Brzozowski T, Konturek SI,
A, Hishida A, Ishizaki T: Influence of jsessionid=AAD3E3D625382610CF52EE75 Marquez M, Torres J, Ortiz JA: Studies on
CYP2C19 pharmacogenetic polymorphism 696E5703?term=AZD0865&submit=Search] the cytoprotective and antisecretory activity
on proton pump inhibitor-based therapies. 200 Altana Group: Gastrointestinal projects of ebrotidine. A review. Arzneimittelfor-
Drug Metab Pharmacokinet 2005; 20: 153– [http://www.altana.com/root/index.php? schung 1997;47(suppl 4A):578–589.
167. page_id=2025] 214 Slomiany BL, Piotrowski J, Slomiany A: Gas-
188 Domagala F, Ficheux H, Houin G, Barre J: 201 Park S, Ahn B, Lee B, Kang H, Song K: Phar- troprotective properties of ebrotidine. A re-
Pharmacokinetics of tenatoprazole, a newly macokinetics of YH1885. Gut 2003;52:(sup- view. Arzneimittelforschung 1997; 47(suppl
synthesized proton pump inhibitor, in heal- pl VI):A62. 4A):459–467.
thy male Caucasian volunteers. Arzneimittel- 202 Briving C, Svensson K, Maxvall I, Andersson 215 Slomiany BL, Piotrowski J, Slomiany A:
forschung 2006;56:33–39. K: Mechanism of action of AZD0865, an Anti-Helicobacter pylori activities of ebroti-
189 Galmiche JP, Bruley des Varannes S, Du- H+,K+-ATPase selective, potassium competi- dine. A review of biochemical and animal ex-
crotte P, Sacher-Huvelin S, Vavasseur F, Tac- tive acid blocker. Gastroenterology 2004; perimental studies and data. Arzneimittel-
coen A, Fiorentini P, Homerin M: Tenatopra- 126(suppl):A333. forschung 1997;47(suppl 4A):475–482.
zole, a novel proton pump inhibitor with a 203 Wurst W, Hartmann M: Current status of 216 Matov V, Metchkov G, Krastev Z, Tchernev
prolonged plasma half-life: effects on intra- acid pump antagonists (reversible PPIs). Yale K, Mitova R, Marquez M, Torres J, Herrero
gastric pH and comparison with esomepra- J Biol Med 1996;69:233–243. E, Fillat O, Ortiz JA: Ebrotidine versus ra-
zole in healthy volunteers. Aliment Pharma- nitidine in the treatment of acute duodenal
col Ther 2004;19:655–662. ulcer. A multicentre, randomized, double-
blind, controlled clinical trial. Arzneimittel-
forschung 1997;47(suppl 4A):555–559.

Update on Acid Suppression Therapy Dig Dis 2006;24:11–46 43

DDI923.indd 43 02.05.2006 16:25:03


217 Andrade RJ, Lucena MI, Martin-Vivaldi R, 227 Pantoflickova D, Dorta G, Ravic M, Jornod 240 Hakkinen JP, Holt WF, Goddard CJ, Oates
Fernandez MC, Nogueras F, Pelaez G, Go- P, Blum AL: Acid inhibition on the first day PJ, Murphy WR, Maciejko JJ, Reiter LA: CP-
mez-Outes A, Garcia-Escano MD, Bellot V, of dosing: comparison of four proton pump 66,948: an antisecretory histamine H2-recep-
Hervas A, Cardenas F, Bermudez F, Romero inhibitors. Aliment Pharmacol Ther 2003;17: tor antagonist with mucosal protective prop-
M, Salmeron J: Acute liver injury associated 1507–1514. erties. Dig Dis Sci 1991;36:1721–1728.
with the use of ebrotidine, a new H2-receptor 228 Haruki S, Matsumoto H, Kodama K, Takebe 241 Dockray GJ: Gastrin. Best Pract Res Clin En-
antagonist. J Hepatol 1999;31:641–646. M, Takayanagi H, Cyong J: Pharmacokinetic docrinol Metab 2004;18:555–568.
218 Pineda JA, Larrauri J, Macias J, Hernandez studies of lafutidine (FRG-8813) in man (in 242 Bertaccini G: Peptides: gastrointestinal hor-
A, Guijarro J, Sayago M, Gavilan F, Aguilar Japanese). Jpn Pharmacol Ther 1995; 23: mones. Gastrin; in Bertaccini G (ed): Gastro-
J, Lissen E: Rapid progression to liver cirrho- 179–189. intestinal Motility: Mediators and Drugs.
sis of toxic hepatitis due to ebrotidine. J Hep- 229 Lin JH: Pharmacokinetic and pharmacody- Berlin, Springer, 1982, vol 2, pp 11–40.
atol 1999;31:777–778. namic properties of histamine H2-receptor 243 Koh TJ, Chen D: Gastrin as a growth factor
219 Castillo JR, Torello J, Hernandez A: Liver antagonists. Relationship between intrinsic in the gastrointestinal tract. Regul Pept 2000;
injury caused by ebrotidine: a new example potency and effective plasma concentrations. 93:37–44.
of the utility of the postmarketing surveil- Clin Pharmacokinet 1991;20:218–236. 244 Rehfeld JF: Cholecystokinin. Best Pract Res
lance. Eur J Clin Pharmacol 2000; 56: 187– 230 Takeyama M, Itoh H, Hayashi T, Nagano T: Clin Endocrinol Metab 2004;18:569–586.
189. Effects of histamine H2-receptor antagonists 245 Wank SA: G protein-coupled receptors in gas-
220 Hirakawa N, Matsumoto H, Hosoda A, Se- on human plasma levels of motilin, gastrin, trointestinal physiology. I. CCK receptors: an
kine A, Yamaura T, Sekine Y: A novel hista- secretin and somatostatin. Pharm Pharmacol exemplary family. Am J Physiol 1998; 274:
mine 2 (H2) receptor antagonist with gastro- Commun 1998;4:591–594. G607–G613.
protective activity. II. Synthesis and 231 Itoh H, Naito T, Takeyama M: Lafutidine 246 Noble F, Wank SA, Crawley JN, Bradwejn J,
pharmacological evaluation of 2-furfuryl-thio changes levels of somatostatin, calcitonin Seroogy KB, Hamon M, Roques BP: Interna-
and 2-furfurylsulfinyl acetamide derivatives gene-related peptide, and secretin in human tional Union of Pharmacology. XXI. Struc-
with heteroaromatic rings. Chem Pharm Bull plasma. Biol Pharm Bull 2002;25:379–382. ture, distribution, and functions of cholecys-
(Tokyo) 1998;46:616–622. 232 Taché Y, Raybould H, Wei JY: Central and tokinin receptors. Pharmacol Rev 1999; 51:
221 Fukushima Y, Otsuka H, Ishikawa M, Asano peripheral actions of calcitonin gene-related 745–781.
T, Anai M, Katsube T, Ogawa K, Kajiwara T, peptide on gastric secretory and motor func- 247 Waldum HL, Kleveland PM, Sandvik AK,
Ohkawa S, Ishikawa T, Omata M, Saitoh T: tion. Adv Exp Med Biol 1991;298:183–198. Brenna E, Syversen U, Bakke I, Tommeras K:
Potent and long-lasting action of lafutidine on 233 Onodera S, Nishida K, Takeuchi K: Unique The cellular localization of the cholecystoki-
the human histamine H2 receptor. Digestion profile of lafutidine, a novel histamine H2-re- nin 2 (gastrin) receptor in the stomach. Phar-
2001;64:155–160. ceptor antagonist. Mucosal protection macol Toxicol 2002;91:359–362.
222 Shibata M, Yamaura T, Inaba N, Onodera S, throughout gastrointestinal tract mediated by 248 Zavros Y, Fleming WR, Hardy KJ, Shulkes
Chida Y, Ohnishi H: Gastric antisecretory ef- capsaicin-sensitive afferent neurons. Drug A: Regulation of fundic and antral somato-
fect of FRG-8813, a new histamine H2 recep- Des Rev 2004;1:133–144. statin secretion by CCK and gastrin. Am J
tor antagonist, in rats and dogs. Eur J Phar- 234 Nagahama K, Yamato M, Kato S, Takeuchi Physiol 1998;274:G742–G750.
macol 1993;235:245–253. K: Protective effect of lafutidine, a novel H2- 249 Kidd M, Modlin IM, Tang LH: Gastrin and
223 Shimatani T, Inoue M, Kuroiwa T, Horikawa receptor antagonist, on reflux esophagitis in the enterochromaffin-like cell: an acid up-
Y, Mieno H, Nakamura M: Effect of omepra- rats through capsaicin-sensitive afferent neu- date. Dig Surg 1998;15:209–217.
zole 10 mg on intragastric pH in three differ- rons. J Pharmacol Sci 2003;93:55–61. 250 Reubi JC, Waser B, Horisberger U, Halter F,
ent CYP2C19 genotypes, compared with 235 Tanaka A, Mizoguchi H, Hase S, Miyazawa Soroka CJ, Kumar RR, Goldenring JR, Mod-
omeprazole 20 mg and lafutidine 20 mg, a T, Takeuchi K: Intestinal protection by lafu- lin IM: Identification of somatostatin and
new H2-receptor antagonist. Aliment Phar- tidine, a histamine H2-receptor antagonist, gastrin receptors on enterochromaffin-like
macol Ther 2003;18:1149–1157. against indomethacin-induced damage in cells from Mastomys gastric tumors. Endocri-
224 Saitoh T, Fukushima Y, Otsuka H, Hirakawa rats – role of endogenous nitric oxide. Med nology 1992;131:166–172.
J, Ishikawa M, Takahashi H, Mori H, Ohka- Sci Monit 2001;7:869–877. 251 Sandor A, Kidd M, Lawton GP, Miu K, Tang
wa S: Lafutidine, a novel histamine H2 recep- 236 Okayama M, Tsubouchi R, Kato S, Takeuchi LH, Modlin IM: Neurohormonal modulation
tor antagonist, inhibits daytime acid secre- K: Protective effect of lafutidine, a novel his- of rat enterochromaffin-like cell histamine se-
tion in both H. pylori infected and non tamine H2-receptor antagonist, on dextran cretion. Gastroenterology 1996; 110: 1084–
infected human subjects. J Gastroenterol sulfate sodium-induced colonic inflamma- 1092.
Hepatol 2002;17(suppl):A828. tion through capsaicin-sensitive afferent neu- 252 Scarpignato C: Cholecystokin antagonists
225 Isomoto H, Inoue K, Furusu H, Nishiyama rons in rats. Dig Dis Sci 2004; 49: 1696– and motilides. Pharmacology and potential in
H, Shikuwa S, Omagari K, Mizuta Y, Murase 1704. the treatment of gastroesophageal reflux dis-
K, Murata I, Kohno S: Lafutidine, a novel 237 Nozawa Y, Nishihara K, Akizawa Y, Orimo- ease and other digestive motor disorders.
histamine H2-receptor antagonist, vs. lanso- to N, Nakano M, Uji T, Ajioka H, Kanda A, Front Gastrointest Res 1992;20:90–128.
prazole in combination with amoxicillin and Matsuura N, Kiniwa M: Lafutidine inhibits 253 Häkanson R, Ding XQ, Norlen P, Lindstrom
clarithromycin for eradication of Helico- Helicobacter pylori-induced interleukin-8 E: CCK2 receptor antagonists: pharmacologi-
bacter pylori. Helicobacter 2003;8:111–119. production in human gastric epithelial cells. cal tools to study the gastrin-ECL cell-parietal
226 Inamori M, Togawa J, Iwasaki T, Ozawa Y, J Gastroenterol Hepatol 2004;19:506–511. cell axis. Regul Pept 1999;80:1–12.
Kikuchi T, Muramatsu K, Chiguchi G, Mat- 238 Pan C, Xu X, He H, Cai X, Zhang X: Separa- 254 Herranz R: Cholecystokinin antagonists:
sumoto S, Kawamura H, Abe Y, Kirikoshi H, tion and identification of cis and trans iso- pharmacological and therapeutic potential.
Kobayashi N, Shimamura T, Kubota K, Sa- mers of 2-butene-1,4-diol and lafutidine by Med Res Rev 2003;23:559–605.
kaguchi T, Saito S, Ueno N, Nakajima A: HPLC and LC-MS. J Zhejiang Univ Sci B 255 Chambers MS, Fletcher SR: CCK-B antago-
Early effects of lafutidine or rabeprazole on 2005;6:74–78. nists in the control of anxiety and gastric acid
intragastric acidity: which drug is more suit- 239 Takeuchi K, Kato S: Prophylactic or treating secretion. Prog Med Chem 2000;37:45–81.
able for on-demand use? J Gastroenterol agent for inflammatory enteropathy. Patent
2005;40:453–458. No JP 321366, 2003.

44 Dig Dis 2006;24:11–46 Scarpignato /Pelosini /Di Mario

DDI923.indd 44 02.05.2006 16:25:03


256 Weiss H: Proglumide after 10 years: a review 268 Nishida A, Yuki H, Tsutsumi R, Miyata K, 279 Takemoto Y, Yuki H, Nishida A, Ito H, Ko-
of clinical results; in Weiss J, Miederer SE Kamato T, Ito H, Yamano M, Honda K: L- bayashi-Uchida A, Takinami Y, Akuzawa S,
(eds): Proglumide and Other Gastrin Antago- 365,260, a potent CCK-B/gastrin receptor an- Ohta M, Satoh M, Semple G, Miyata K: Ef-
nists. Amsterdam, Elsevier, 1979, pp 113– tagonist, suppresses gastric acid secretion in- fects of YF476, a potent and selective gastrin/
131. duced by histamine and bethanechol as well cholecystokinin-B receptor antagonist, on
257 Hahne WF, Jensen RT, Lemp GF, Gardner as pentagastrin in rats. Jpn J Pharmacol 1992; gastric acid secretion in beagle dogs with gas-
JD: Proglumide and benzotript: members of 58:137–145. tric fistula. Arzneimittelforschung 1998; 48:
a different class of cholecystokinin receptor 269 Hirst BH, Elliott KJ, Ryder H, Szelke M: In- 403–407.
antagonists. Proc Natl Acad Sci USA 1981; hibition of gastrin- and histamine-stimulated 280 Boyce M, Warrington S, Johnston A, Harris
78:6304–6308. gastric acid secretion by gastrin and cholecys- A: Effect on gastric pH of single doses of
258 Rovati LC: Pentanoic acid derivatives; in tokinin antagonists in the rat. Aliment Phar- YF476, a new gastrin antagonist, compared
Adler G, Beglinger C (eds): Cholecystokinin macol Ther 1991;5:31–39. with ranitidine and placebo. Br J Clin Phar-
Antagonists in Gastroenterology. Basic and 270 Varga G, Campbell DR, Bussjaeger LJ, Solo- macol 2000;49:493P.
Clinical Status. Berlin, Springer, 1991, pp mon TE: Role of gastrin and cholecystokinin 281 Boyce M, Warrington S, Johnston A, Harris
65–69. receptors in regulation of peptone-stimulated A: Effect on gastric pH of repeated doses of
259 Revel L, Makovec F: Discovery of new CCK2 gastric acid secretion in conscious rats. Eur J YF476, a new gastrin antagonist, compared
receptor antagonists. A review of pharmaco- Pharmacol 1993;250:37–42. with omeprazole and placebo. Br J Clin Phar-
logical studies. I. Development of potential 271 Murphy MG, Sytnik B, Kovacs TO, Mertz H, macol 2000;50:383P–384P.
therapeutic tools for gastrin-related gastroin- Ewanik D, Shingo S, Lin JH, Gertz BJ, Walsh 282 Boyce M, Warrington S, Lewis Y, Nentwich
testinal diseases. Methods Find Exp Clin JH: The gastrin-receptor antagonist L- H, Harris A: Adaptation to the antisecretory
Pharmacol 1999;21:375–383. 365,260 inhibits stimulated acid secretion in effect of YF476, a new gastrin antagonist, in
260 Scarpignato C, Kisfalvi I, D’Amato M, Varga humans. Clin Pharmacol Ther 1993;54:533– healthy men. Br J Clin Pharmacol 2001; 53:
G: Effect of dexloxiglumide and spiroglu- 539. 437P.
mide, two new CCK-receptor antagonists, on 272 Grasing K, Freedholm D, Murphy MG, Swi- 283 Miura N, Yoneta T, Ukawa H, Fikuda Y, Eta
gastric emptying and secretion in the rat: gar M, Russer T, Nosher J, Lin J, Frame V, R, Mera Y, Omata T, Kinomoto T, Kurimo-
evaluation of their receptor selectivity in Clarke L, Seibold JR: Selective blockade of to T, Itoh Z: Pharmacological profile of Z-
vivo. Aliment Pharmacol Ther 1996;10:411– cholecystokinin type B receptors with L- 360, a novel CCK-B/gastrin (CCK2) antago-
419. 365,260 does not impair gallbladder contrac- nist with excellent oral potency. Gastroen-
261 Revel L, Ferrari F, Makovec F, Rovati LC, tion in normal humans. Am J Gastroenterol terology 2001;120(suppl 1):A311.
Impicciatore M: Characterization of antigas- 1996;91:569–573. 284 Morita H, Miura N, Hori Y, Matsunaga Y,
trin activity in vivo of CR-2194, a new R-4- 273 Grasing K, Murphy MG, Lin J, Swigar M, Ukawa H, Suda H, Yoneta T, Kurimoto T,
benzamido-5-oxo-pentanoic acid derivative. Freedholm D, Clarke L, Zhang J, Wolkowitz Itoh Z: Effects of Z-360, a novel CCK-B/gas-
Eur J Pharmacol 1992;216:217–224. OM, Weingartner H, Putnam K, Seibold JR: trin (CCK2) receptor antagonist, on meal-in-
262 Beltinger J, Hildebrand P, Drewe J, Christ A, Human pharmacokinetics and tolerability of duced acid secretion and experimental ulcer
Hlobil K, Ritz M, D’Amato M, Rovati L, Beg- L-365,260, a novel cholecystokinin-B antago- models in dogs and rats. Gastroenterology
linger C: Effects of spiroglumide, a gastrin re- nist. J Clin Pharmacol 1996;36:292–300. 2001;120(suppl 1):A331.
ceptor antagonist, on acid secretion in hu- 274 Kramer MS, Cutler NR, Ballenger JC, Pat- 285 Schmassmann A, Reubi JC: Cholecystokinin-
mans. Eur J Clin Invest 1999;29:153–159. terson WM, Mendels J, Chenault A, et al: A B/gastrin receptors enhance wound healing in
263 Makovec F, Revel L, Letari O, Mennuni L, placebo-controlled trial of L-365,260, a CCK- the rat gastric mucosa. J Clin Invest 2000;
Impicciatore M: Characterization of antise- B antagonist, in panic disorder. Biol Psychia- 106:1021–1029.
cretory and antiulcer activity of CR-2945, a try 1995;37:462–466. 286 Jensen RT: Involvement of cholecystokinin/
new potent and selective gastrin/CCK(B) re- 275 Bock MG, DiPardo RM, Mellin EC, Newton gastrin-related peptides and their receptors in
ceptor antagonist. Eur J Pharmacol 1999; RC, Veber DF, Freedman SB, Smith AJ, Pa- clinical gastrointestinal disorders. Pharmacol
369:81–90. tel S, Kemp JA, Marshall GR, et al: Second- Toxicol 2002;91:333–350.
264 Beglinger C, Degen L, Schroller S, D’Amato generation benzodiazepine CCK-B antago- 287 Aly A, Shulkes A, Baldwin GS: Gastrins, cho-
M, Persiani S: Oral itriglumide, a specific nists. Development of subnanomolar analogs lecystokinins and gastrointestinal cancer.
CCK2/gastrin receptor antagonist, inhibits with selectivity and water solubility. J Med Biochim Biophys Acta 2004;1704:1–10.
gastrin stimulated gastric acid secretion in Chem 1994;37:722–724. 288 Waldum HL, Fossmark R, Bakke I, Martin-
humans. Gut 2005;54(suppl VII):A36. 276 Patel S, Chapman KL, Heald A, Smith AJ, sen TC, Qvigstad G: Hypergastrinemia in
265 Dunlop J: CCK receptor antagonists. Gen Freedman SB: Measurement of central ner- animals and man: causes and consequences.
Pharmacol 1998;31:519–524. vous system activity of systemically adminis- Scand J Gastroenterol 2004;39:505–509.
266 Lotti VJ, Chang RS: A new potent and selec- tered CCKB receptor antagonists by ex vivo 289 Waldum HL, Gustafsson B, Fossmark R,
tive non-peptide gastrin antagonist and brain binding. Eur J Pharmacol 1994; 253: 237– Qvigstad G: Antiulcer drugs and gastric can-
cholecystokinin receptor (CCK-B) ligand: 244. cer. Dig Dis Sci 2005;50(suppl 1):S39–S44.
L365,260. Eur J Pharmacol 1989; 162: 273– 277 Semple G, Ryder H, Rooker DP, Batt AR, 290 Steel K: Gastrin and gastrin receptor ligands
280. Kendrick DA, Szelke M, Ohta M, Satoh M, – a review of recent patent literature. IDrugs
267 Pendley CE, Fitzpatrick LR, Ewing RW, Mo- Nishida A, Akuzawa S, Miyata K: (3R)-N-(1- 2002;5:689–695.
lino BF, Martin GE: The gastrin/cholecysto- (tert-butylcarbonylmethyl)-2,3-dihydro-2- 291 Reubi JC, Schaer JC, Waser B: Cholecystoki-
kinin B receptor antagonist L-365,260 reduc- oxo-5-(2-pyridyl)-1H-1,4-benzodiazepin-3- nin (CCK)-A and CCK-B/gastrin receptors in
es basal acid secretion and prevents gastro- yl)-N-(3-(methylamino)phenyl)urea (YF476): human tumors. Cancer Res 1997; 57: 1377–
intestinal damage induced by aspirin, ethanol a potent and orally active gastrin/CCK-B an- 1386.
and cysteamine in the rat. J Pharmacol Exp tagonist. J Med Chem 1997;40:331–341. 292 Ferrand A, Wang TC: Gastrin and cancer: A
Ther 1993;265:1348–1354. 278 Takinami Y, Yuki H, Nishida A, Akuzawa S, review. Cancer Lett 2006 (in press).
Uchida A, Takemoto Y, Ohta M, Satoh M, 293 Rengifo-Cam W, Singh P: Role of progastrins
Semple G, Miyata K: YF476 is a new potent and gastrins and their receptors in GI and
and selective gastrin/cholecystokinin-B re- pancreatic cancers: targets for treatment.
ceptor antagonist in vitro and in vivo. Ali- Curr Pharm Des 2004;10:2345–2358.
ment Pharmacol Ther 1997;11:113–120.

Update on Acid Suppression Therapy Dig Dis 2006;24:11–46 45

DDI923.indd 45 02.05.2006 16:25:04


294 Anonymous: Gastrin 17 vaccine – Aphton: 299 Makishima R, Larkin D, Michaeli D, Gagi- 304 Modlin IM, Sachs G, Wright N, Kidd M: Ed-
anti-gastrin 17 immunogen, G17DT. Bio- nella T: Active immunization against gastrin- kins and a century of acid suppression. Diges-
drugs 2003;17:223–225. 17 with an N-terminal derived immunogen tion 2005;72:129–145.
295 Iwase K, Evers BM, Hellmich MR, Guo YS, inhibits gastric and duodenal lesions in rats. 305 Leurs R, Bakker RA, Timmerman H, de Esch
Higashide S, Kim HJ, Townsend CM Jr: Reg- Gastroenterology 1995;106:A824. IJ: The histamine H3 receptor: from gene
ulation of growth of human gastric cancer by 300 Justin T, Watson S, Michaeli D, Hardcastle cloning to H3 receptor drugs. Nat Rev Drug
gastrin and glycine-extended progastrin. Gas- J, Steele R: Gastric acid suppression using Discov 2005;4:107–120.
troenterology 1997;113:782–790. anti-gastrin 17 antibodies produced by a gas- 306 Degen LP, Peng F, Collet A, Rossi L, Ket-
296 Brett BT, Smith SC, Bouvier CV, Michaeli D, trin immunogen, Gastrimmune, in an in vivo terer S, Serrano Y, Larsen F, Beglinger C, Hil-
Hochhauser D, Davidson BR, Kurzawinski pig model. Gastroenterology 1995; 108: debrand P: Blockade of GRP receptors inhib-
TR, Watkinson AF, Van Someren N, Pound- A125. its gastric emptying and gallbladder con-
er RE, Caplin ME: Phase II study of anti-gas- 301 Smith AM, Morris T, Justin T, Michaeli D, traction but accelerates small intestinal tran-
trin-17 antibodies, raised to G17DT, in ad- Watson SA: Gastrimmune-induced antigas- sit. Gastroenterology 2001;120:361–368.
vanced pancreatic cancer. J Clin Oncol 2002; trin-17 antibodies inhibit acid secretion in a 307 Gibril F, Jensen RT: Zollinger-Ellison syn-
20:4225–4231. rat fistula model. Aliment Pharmacol Ther drome revisited: diagnosis, biologic markers,
297 ClinicalTrials.gov. Ongoing Trials with 2001;15:1981–1988. associated inherited disorders, and acid hy-
G-17DT [http://www.clinicaltrials.gov/ct/sea 302 Aphton Corp: Products [http://www.aphton. persecretion. Curr Gastroenterol Rep 2004;6:
rch;jsessionid=65546F66B71B7A09A601C6 com/products.asp?aph=2] 454–463.
258731D626?term=G-17DT] 303 Gevas P, Grimes S, Karr S, Michaelu D: 308 Haga Y, Nakatsura T, Shibata Y, Sameshima
298 Straathof JW, Lamers CB, Masclee AA: Ef- Preparation of a medicament for the treat- H, Nakamura Y, Tanimura M, Ogawa M:
fect of gastrin-17 on lower esophageal sphinc- ment of gastroesophageal reflux disease. Human gastric carcinoid detected during
ter characteristics in man. Dig Dis Sci 1997; EP1077716, 2001. long-term antiulcer therapy of H2 receptor
42:2547–2551. antagonist and proton pump inhibitor. Dig
Dis Sci 1998;43:253–257.

46 Dig Dis 2006;24:11–46 Scarpignato /Pelosini /Di Mario

DDI923.inddView46
publication stats 02.05.2006 16:25:04

You might also like