You are on page 1of 18

Adv Ther

DOI 10.1007/s12325-016-0345-2

REVIEW

Characteristics of the Novel Potassium-Competitive


Acid Blocker Vonoprazan Fumarate (TAK-438)
Kazuyoshi Otake . Yuuichi Sakurai . Haruyuki Nishida . Hideo Fukui .
Yoshihiko Tagawa . Hitomi Yamasaki . Masatoshi Karashima .
Keiichi Otsuka . Nobuhiro Inatomi

Received: February 24, 2016


 Springer Healthcare 2016

Abstract: Proton pump inhibitors (PPIs) are due to genetic polymorphisms of cytochrome
widely prescribed as first-line therapy for the P450 (CYP) 2C19 metabolism; (3) they have a
treatment of acid-related diseases, such as relatively slow onset of pharmacological action
peptic ulcers and gastro-esophageal reflux and may require several doses to achieve
disease, and for the eradication of Helicobacter optimal acid suppression and symptom relief;
pylori. However, the therapeutic efficacy of and (4) they often do not provide
conventional PPIs is considered limited stable suppression of gastric acid secretion
because: (1) they are unstable under acidic over 24 h. Vonoprazan fumarate (TAK-438,
conditions and require an enteric-coated hereinafter referred to as ‘‘vonoprazan’’) is a
formulation in clinical use; (2) they show high new potassium-competitive acid blocker
interindividual variability in pharmacokinetics (P-CAB) developed to resolve the above
limitations of conventional PPIs. Various
physicochemical data have shown that
Enhanced content To view enhanced content for this
article go to www.medengine.com/Redeem/92D4F060 vonoprazan has a high solubility and stability
50CF12A4.

K. Otake (&) Y. Tagawa  H. Yamasaki


Global Medical Affairs Japan Department, Takeda Drug Metabolism and Pharmacokinetics Research
Pharmaceutical Co., Ltd., Tokyo, Japan Laboratories, Pharmaceutical Research Division,
e-mail: Kazuyoshi.otake@takeda.com Takeda Pharmaceutical Co., Ltd., Kanagawa, Japan

Y. Sakurai M. Karashima
Clinical Science, Takeda Development Center Japan, Analytical Development Laboratories, CMC Center,
Takeda Pharmaceutical Co., Ltd., Osaka, Japan Takeda Pharmaceutical Co., Ltd., Kanagawa, Japan

H. Nishida K. Otsuka
Medicinal Chemistry Research Laboratories, Analytical Development Laboratories, CMC Center,
Pharmaceutical Research Division, Takeda Takeda Pharmaceutical Co., Ltd., Osaka, Japan
Pharmaceutical Co., Ltd., Kanagawa, Japan
N. Inatomi
H. Fukui Extra Value Generation Drug Discovery Unit,
Drug Safety Research Laboratories, Pharmaceutical Pharmaceutical Research Division, Takeda
Research Division, Takeda Pharmaceutical Co., Ltd., Pharmaceutical Co., Ltd., Kanagawa, Japan
Kanagawa, Japan
Adv Ther

over a broad pH range in aqueous conditions. In peptic ulcers caused by gastrin-producing


addition, vonoprazan has a more potent and tumors (Zollinger–Ellison syndrome) [1].
longer-lasting acid suppression effect than the Gastric acid is secreted from parietal cells of
conventional PPI, lansoprazole. Preclinical the gastric glands, situated in the mucosa of the
pharmacokinetic studies have shown that gastric fundus and the gastric corpus (Fig. 1).
vonoprazan is accumulated and retained in Histamine, acetylcholine, and gastrin stimulate
the stomach for more than 24 h, even after it gastric acid secretion through histamine (H2)
is eliminated from the plasma. From these receptors, muscarinic (M3) receptors, and
findings, we propose that vonoprazan, which cholecystokinin (CCK2) receptors, respectively,
possesses a novel mode of action, can improve which are present on the basolateral membranes
on the outcomes seen with conventional of parietal cells. Histamine activates adenylate
PPI-based treatments for acid-related diseases. cyclase by binding with H2 receptors, increasing
Funding: This review project, including the the intracellular cyclic adenosine
publication of this article, was funded by monophosphate concentration in cells.
Takeda Pharmaceutical Company Limited. Acetylcholine and gastrin increase intracellular
Ca2? concentration via the inositol
Keywords: Acid-related diseases; phospholipid pathway by binding with M3
Gastroenterology; Helicobacter pylori receptors and CCK2 receptors, respectively.
eradication; Lansoprazole; Stimulation of enterochromaffin-like cells by
Potassium-competitive acid blockers; PPI; gastrin also leads to histamine secretion. These
Proton pump inhibitor; Reflux esophagitis; intracellular messengers ultimately promote
TAK-438; Vonoprazan gastric acid secretion by activating relevant
protein kinases and by phosphorylating
proteins (Fig. 2) [2, 3].
INTRODUCTION: ACID-RELATED In the final step of gastric acid secretion, the
DISEASES AND THERAPEUTIC proton pump, H?,K?-ATPase induces the
STRATEGIES secretion of acid into the inner cavity of the
gastric gland (Fig. 1) [4]. H?,K?-ATPase is a
Acid-related diseases are a family of diseases membrane-bound protein and belongs to the
related to gastric acid, for which clinical effects P-type ATPase family. It forms heterodimers
are generally obtained by suppression of gastric consisting of a subunits and b subunits [5].
acid secretion. While this was previously Using the energy of ATP-hydrolysis, it transports
understood to be centered on peptic ulcers, H? to the inner cavity of the gastric gland against
such as gastric ulcers and duodenal ulcers, a concentration gradient of approximately 1
reflux esophagitis has also come to be million-fold (106), in exchange for K? into the
recognized as a significant pathological cytoplasm of parietal cells [6]. In response to the
condition due to adoption of Western-style movement of H?, HCO3- is transported out of
diet. Suppression of gastric acid secretion has the cytoplasm of the parietal cell by Cl-/HCO3-
become a common therapeutic strategy, as exchangers in the basolateral cell membrane in
evidenced by treatments for gastric mucosal exchange for Cl- into the cytoplasm. Cl- is
injury induced by continuous use of transported to the inner cavity of the gastric
non-steroidal anti-inflammatory drugs or gland through the Cl- channel in the apical
Adv Ther

Fig. 1 Parietal cell and proton pump. Reproduced novel mode of action—physicochemical properties,
with permission from K. Otake, Y. Sakurai, H. Nishida non-clinical data, and a proposed mode of action. Prog
et al. Vonoprazan fumarate (TAK-438), a new Med 2014;34:2183–2194
potassium-competitive acid blocker (P-CAB) with a

membrane of the parietal cell (Fig. 2). Although apical membrane, and functions as a proton
the expression of H?,K?-ATPase a subunit mRNA pump. In the secretory canaliculus,
in humans has been observed in the stomach, H?,K?-ATPase is activated, because there is an
adrenal glands, cerebellum, pancreas, and adequate supply of K?, but it does not function
kidneys, the expression of the b subunit mRNA as a proton pump in tubulovesicles due to the
has only been confirmed in the stomach. lack of K? permeability [9].
Furthermore, with respect to the expression of It is known that the therapeutic effects for
protein, both the a subunit and the b subunit acid-related diseases are enhanced in proportion
have only been observed in the stomach, to the rise in intragastric pH. The target pH for
suggesting that the stomach is the only organ acid secretion suppression varies depending on
in humans with significant levels of the acid-related disease. A meta-analysis showed
H?,K?-ATPase [7]. Kraut reported the that following 24-h pH monitoring and clinical
H?,K?-ATPase expression in human kidney in test results of acid secretion inhibitors, the
2001 [8]; however, as Herrmann’s investigation optimal conditions for duodenal ulcer healing
showed that the amount of enzyme expression is are to maintain an intragastric pH of 3 or above
considered to be very small in kidney [7]. for 18–20 h of the day [10]. In patients with
The morphology of the parietal cell reflux esophagitis, it has been shown that an
conspicuously differs between a resting state intragastric pH[4 is the target standard for
and an active acid-secreting state. During the treatment [11, 12].
resting state, H?,K?-ATPase mainly exists in There is a long history of drugs used to
cytoplasmic tubulovesicles, but in the active suppress gastric acid secretion including
state, when the tubulovesicles and the secretory anticholinergic agents and non-selective
canaliculi fuse, H?,K?-ATPase is exposed on the muscarinic receptor antagonists, although
Adv Ther

correlation between H. pylori infection and


diseases including gastric cancer was reported.
To demonstrate adequate antimicrobial potency
in H. pylori eradication therapy, it is necessary to
have an environment of pH[5.0 in order to
promote the transition of H. pylori into the
proliferative phase [14]. Regimens including the
combination of an antimicrobial agent such as
clarithromycin and amoxicillin along with a
PPI, is the standard therapy in patients with H.
pylori [15].
However, the limits of treatment with PPIs
have become clear with the accumulation of
clinical experience.
The reflux symptoms are not adequately
controlled after the first dose of a PPI in
approximately two-thirds of symptomatic
Fig. 2 Gastric acid secretion. Adapted with permission gastroesophageal reflux disease (GERD)
from K. Otake, Y. Sakurai, H. Nishida et al. Vonoprazan
patients, and about half of patients still suffer
fumarate (TAK-438), a new potassium-competitive
acid blocker (P-CAB) with a novel mode of action— symptoms even after a few days of initiating
physicochemical properties, non-clinical data, and a pro- therapies [16]. More than half of patients on
posed mode of action. Prog Med 2014;34:2183–2194. PPIs are dissatisfied with treatment [17]. In
cAMP cyclic adenosine monophosphate, CCK2 cholecys-
addition, the so-called the nocturnal acid
tokin2 receptor, 106 histamine2 receptor, M3 muscarinic3
receptor, ECL enterochromaffin-like cell breakthrough is a significant issue and difficult
to control as it may be receiving PPI twice daily
[18–20]. The ‘‘Japanese Guidelines for Diagnosis
their clinical effects have been unsatisfactory. In and Treatment of Gastroesophageal Reflux
the latter half of the 1970s, the development of Disease 2009’’, states that: ‘‘PPIs demonstrate
H2 receptor antagonists (H2RA) led to a effects that are superior to H2RA, but they
dramatic improvement in the management of should still be improved with respect to acid
peptic ulcer disease, due to their potent acid secretion suppression. In other words, a drug
secretion inhibitory effect. The introduction of that inhibits acid secretion promptly after
proton pump inhibitors (PPIs) during the 1990s administration and controls acid secretion
revolutionized the treatment of acid-related over a 24-h period does not yet exist’’ [21].
diseases. This class of drugs demonstrated For H. pylori eradication therapy, although
significant therapeutic effects not only for the an eradication rate of 80% or more is considered
treatment of peptic ulcer disease, but also for desirable [22], the eradication rate with the
reflux esophagitis. Thus, PPIs became the conventional regimens using a PPI has declined
first-line of therapy for treatment of to 70% due to an increase in
acid-related diseases [13]. antimicrobial-resistant bacteria [23].
The significance of Helicobacter pylori (H. This article is based on previously conducted
pylori) eradication was recognized when the studies and does not involve any new studies of
Adv Ther

human or animal subjects performed by any of are weakly basic, highly lipophilic, and stable at
the authors. a low pH. In clinical terms, it is anticipated from
these characteristics that the onset of the acid
secretion inhibitory effect will be rapid and a
LIMITATIONS OF PPIS
near-maximum effect will be obtained from the
AND DISCOVERY OF POTASSIUM-
first dose. Several pharmaceutical companies
COMPETITIVE ACID BLOCKERS
had proceeded with clinical development of
The current known limitations of PPIs are P-CABs, but development was suspended in all
summarized in the following four points [24]: cases due to their insufficient efficacy,
(1) since they are acid labile, there is a need for hepatotoxicity, and human Ether-a-go-go
preparation-related modifications such as related gene (hERG) channel blockage [27–30].
enteric-coated formulations; (2) there are
interindividual variations in pharmacokinetics DISCOVERY OF VONOPRAZAN
as they are mainly metabolized by cytochrome FUMARATE
P450 2C19 (CYP2C19) of the liver
drug-metabolizing enzyme that exhibits At Takeda Pharmaceutical Company Limited,
genetic polymorphism; (3) show a slow onset we initiated exploratory research with the
of action, usually taking 3–5 days for the objective of discovering ‘‘the ultimate acid
manifestation of optimal effects; (4) often a secretion inhibitor’’ to overcome the
stable inhibitory effect on acid secretion limitations of PPIs and eliminate issues of
cannot be sustained for 24 h, and hence PPIs insufficient efficacy and hepatotoxicity shown
fail to adequately suppress nocturnal acid by previously developed P-CABs.
secretion. We conducted high-throughput random
Development of drugs in order to overcome screening of our in-house chemical library,
the limitations of PPIs has long been attempted. and identified a pyrrole derivative with weak
A new class of acid suppressants, H?,K?-ATPase inhibitory activity, which was
potassium-competitive acid blockers (P-CABs), stable in acidic conditions (Fig. 3, Hit
inhibit the K?-stimulated ATPase activity by Compound 1) [31]. This pyrrole derivative is
competing with the binding of K? to characterized by a structure having relatively
H?,K?-ATPase and prevent K?-stimulated high basicity wherein a sulfonyl group is at the
dephosphorylation [25]. The compound that 1-position, an aromatic ring is at the
was first discovered to block the H?,K?-ATPase 5-position, and an ethyl-amino-methyl group
by this mechanism was SCH28080, an is at the 3-position of the pyrrole ring. In
imidazo[1,2a]pyridine derivative [26]. addition to having the advantages of a low
Subsequently, screening was conducted for molecular weight and a large scope for
? ?
compounds that block H ,K -ATPase by structural conversion, its chemical structure
competitive inhibition of K? binding similar was unique and unprecedented for an acid
to SCH28080, resulting in the discovery of suppressant.
compounds such as pyrimidine derivatives, In order to evaluate the potential of this hit
imidazonaphthyridine derivatives, and compound, we synthesized various pyrrole
pyrrolopyridine derivatives. These compounds derivatives, and studied the structure–activity
Adv Ther

Fig. 3 Discovery of vonoprazan. Reproduced with mode of action—physicochemical properties, non-clinical


permission from K. Otake, Y. Sakurai, H. Nishida data, and a proposed mode of action. Prog Med
et al. Vonoprazan fumarate (TAK-438), a new 2014;34:2183–2194
potassium-competitive acid blocker (P-CAB) with a novel

relationship in detail. As a result, we succeeded In preclinical testing and chemistry,


in discovering a novel pyrrole derivative (Fig. 3, manufacturing, and control testing, vonoprazan
Lead Compound 2) with a more powerful and exhibited a gastric acid secretion inhibitory effect
sustained action than conventional PPIs [32]. that was more potent and sustained than
Subsequently, based on this lead compound, we conventional PPIs and had satisfactory
conducted lead optimization focusing on physicochemical properties. In addition,
factors such as lipophilicity and the effects of vonoprazan had no observable issues of
substituents. This process led to the discovery of hepatotoxicity or hERG channel blockage [34].
vonoprazan fumarate (development code: Phase III clinical trials conducted in Japan for
TAK-438, hereinafter ‘‘vonoprazan’’), which indications including reflux esophagitis, gastric
had a potent and sustained acid secretion ulcers, duodenal ulcers, and H. pylori eradication,
inhibitory effect combined with an excellent demonstrated potent efficacy and favorable safety
pharmacokinetic (absorption, distribution, and tolerability [35–38]. An application for
metabolism, excretion) and toxicological approval of manufacture and sale of vonoprazan
profiles, leading to the start of its development was filed with the Japanese Ministry of Health,
(Fig. 3) [33]. Labor, and Welfare on February 28, 2014.
Adv Ther

CHARACTERISTICS Vonoprazan also exhibits satisfactory solubility


OF VONOPRAZAN over a wide pH range (Fig. 4b) [34] and has been
confirmed to rapidly dissolve from tablets
Excellent Physicochemical Properties (Fig. 4c) [34]. These results have suggested that
vonoprazan has a rapid onset of action and can
Vonoprazan has the following superior exhibit a sustained gastric acid secretion
physicochemical properties, thus, overcoming inhibition under the acidic environment of
the aforementioned limitations of PPIs. secretory canaliculi.
Since the stability of vonoprazan under Furthermore, vonoprazan is a basic
acidic conditions is superior to that of compound; the pKa (acid dissociation
conventional PPIs, it has the major advantage constant) value of the side chain amino group
that it does not require an enteric coating. In an portion is 9.3. It exhibits highly satisfactory
acidic environment (pH 1*2) within the membrane permeability in an environment of
stomach, vonoprazan is stable for at least 8 h pH 7.4, which is similar to the pH of blood. In
(Fig. 4a, internal company data), and exhibits contrast, a clear decline in membrane
satisfactory stability even at a neutral pH. permeability has been confirmed in lower pH

A B
Time (h) Survival rate (%) pH Solubility (mg/mL)a
(pH 1.2*, 37ºC)
1.2 b 19.0
0 100.0 c
4.5 4.54
2 99.2
6.8 d 5.48
4 98.3 a
37ºC
6 97.2 b
First liquid of Japanese Pharmacopoeia elution
test liquids
8 96.0 c
0.05M acetic acid buffer liquid
d
* First liquid of Japanese Pharmacopoeia elution test Second liquid of Japanese Pharmacopoeia
liquids elution test liquids

C D Membrane
permeability*
Solvent pH (nm/sec)
First liquid of Japanese
Pharmacopoeia elution test 1.2 0
Elution rate (%)

liquids

PrismaTM universal buffer 3.0 0


TM
Prisma universal buffer 5.0 118

PrismaTM universal buffer 7.0 315


* Parallel Artificial Membrane Permeability Assay (PAMPA)

Time (minutes)

Fig. 4 Physicochemical properties of vonoprazan. H. Nishida et al. Vonoprazan fumarate (TAK-438), a new
a Stability in acidic conditions (pH 1.2). b Solubility at potassium-competitive acid blocker (P-CAB) with a novel
different pH values. c Elution test at different pH values. mode of action—physicochemical properties, non-clinical
d Membrane permeability at different pH values. data, and a proposed mode of action. Prog Med
Reproduced with permission from K. Otake, Y. Sakurai, 2014;34:2183–2194
Adv Ther

Fig. 5 Inhibitory activity of vonoprazan and LPZ on from K. Otake, Y. Sakurai, H. Nishida et al. Vonoprazan
H?, K?-ATPase. Enzyme inhibitory activity was measured fumarate (TAK-438), a new potassium-competitive acid blocker
after 40-min incubation of porcine H?, K?-ATPase with (P-CAB) with a novel mode of action—physicochemical
different concentrations of a vonoprazan and b LPZ at pH 6.5 properties, non-clinical data, and a proposed mode of action.
(closed circle) or pH 7.5 (open triangle). Adapted with permission Prog Med 2014;34:2183–2194

environments due to a further drop in the CYP2C19 isoenzymes cause interindividual


non-ionic form of the compound (Fig. 4d, variations in the efficacy of PPIs. The genetic
internal company data). Based on these polymorphism of CYP2C19 can be determined
chemical and structural properties, it may be by a single nucleotide polymorphism of exon 4
inferred that vonoprazan can rapidly move into and exon 5, and is classified into homozygous
the acidic secretory canaliculi and remain there extensive metabolizer (homoEM: *1/*1),
over a long period [39]. heterozygous extensive metabolizer (hetEM:
The in vitro enzyme inhibitory activity of *1/*2 or *1/*3), and poor metabolizer (PM: *2/
the typical PPI, lansoprazole compared with *2 or *2/*3 or *3*3) in the combination of
vonoprazan on isolated and purified porcine CYP2C19*2 (*2) and CYP2C19*3 (*3) [41–43]. It
H?,K?-ATPase is shown in Fig. 5. The IC50 of is known that variation in CYP2C19 activity is
vonoprazan at pH 6.5 and pH 7.5 were 0.019 greater in Japanese people than in Europeans
and 0.028 lM, respectively. In contrast, the IC50 and North Americans. The rate of PM
of lansoprazole at pH 6.5 and pH 7.5 were 6.8 prevalence is 1–2% in Europeans and North
and 66 lM, respectively. These results Americans, and approximately 20% in Asians
demonstrate that vonoprazan has an [44]. In a study which analyzed 300 Japanese
? ?
H ,K -ATPase inhibitory activity that is people, the prevalence of homoEM was 35%,
358–2357 times stronger than that of hetEM was 49%, and PM was 16% [45]. In
lansoprazole in the weakly acidic to neutral people with homoEM, PPIs are rapidly
range pH, and is not significantly affected by pH metabolized, leading to a low blood
[40]. concentration, whereas in people with PM, the
blood concentration of PPIs is high due to their
Minimal Interindividual Variation low metabolic activity. Interindividual
in Pharmacokinetics variation in the pharmacokinetics of PPIs also
affects therapeutic efficacy. Administration of
Cytochrome P450 2C19 (CYP2C19) and 3A4 lansoprazole 30 mg daily over an 8-week period
(CYP3A4) are the main liver enzymes that to Japanese patients with gastroesophageal
metabolize PPIs. Genetic polymorphisms in reflux disease resulted in cure rates of 45.8,
Adv Ther

67.9, and 84.6% in patients with homoEM, PPIs after oral administration is short (1–2 h),
hetEM, and PM, respectively. These results and the concentration of the active form of PPIs
suggest a significant correlation between the existing in the secretory canaliculus decreases
therapeutic effects of lansoprazole and over a short time [51]. On the other hand, the
CYP2C19 activity in Japanese patients with half-life of H?,K?-ATPase is approximately 50 h
gastroesophageal reflux disease [46]. and 25% of H?,K?-ATPase is newly
Vonoprazan is metabolized by multiple biosynthesized each day [52]. PPIs are unable
enzymes including CYP3A4 and a to sustain gastric acid secretion inhibitory
non-oxidative enzyme (sulfotransferase). action against H?,K?-ATPase that is constantly
CYP2C19 partially contributes to the being produced and supplemented.
metabolism of vonoprazan [47], and its Thus, in order to demonstrate their
contribution is considered to be small. maximum effect, 3–5 days of repeated
Single-dose or repetitive oral administration of administration of PPIs is required to achieve a
vonoprazan to healthy Japanese adults did not steady-state concentration and consistent
show any significant correlation of the clinical effects [53–56].
CYP2C19 genotype with total drug exposure In contrast, as already described, vonoprazan
(area under the curve from time 0 to infinity; is stable in acid, and non-covalently binds with
AUC0-inf) and maximum serum concentration H?,K?-ATPase in competition with K? [40].
(Cmax) [48, 49]. In vitro studies in HEK293 that express rabbit
H?,K?-ATPase cells showed that non-labeled
Optimal Efficacy can be Expected vonoprazan (2.5 lg/mL) was able to substitute
from the First Dose half of the [14C] vonoprazan (50 nM) within
4.7 h. The binding was extremely stable and the
It is known that PPIs require several days to dissociation was gradual [57]. This slow
have a maximum effect. Even when PPIs dissociation rate can also be explained from
migrate into parietal cells, they are not the structural and biological findings relating to
converted to the active form in the cytoplasm P-CABs. An electron diffraction analysis of
or in the tubulovesicles, and are thus unable to porcine H?,K?-ATPase and SCH28080, a
inhibit H?,K?-ATPase within the tubulovesicles. prototype of P-CABs, using cryo-electron
The site where PPIs are activated is the secretory microscopy, revealed that a major structural
canaliculus of the parietal cell. They migrate change is induced over the entirety of the
into the secretory canaliculus, where they are molecule by inhibitor binding. As a result of
then converted into the active form inhibitor binding, the transmembrane helix
(sulfenamide form) in a strongly acidic constitutes widely open luminal-open
environment. The active form then inhibits conformations, and the linker that connects
acid production by forming disulfide bonds (S–S the M2 helix of the transmembrane domain
bonds) with the cysteine residue of and the A domain inside the cytoplasm
H?,K?-ATPase on the secretory canaliculus [50]. subsequently forms an a-helix. This structural
However, as active forms of PPIs are easily change triggers a reconfiguration of the entire
decomposed in acid, they are unable to inhibit cytoplasm domain, closely resembling the
? ?
all of the H ,K -ATPase of the secretory E2P ground state homologous to sarcoplasmic
canaliculus. Moreover, the plasma half-life of reticulum Ca2?-ATPase and other P2-type
Adv Ther

Fig. 6 Accumulation of vonoprazan and LPZ in cultured et al. Vonoprazan fumarate (TAK-438), a new
rabbit fundic glands. Primary cultured rabbit fundic glands potassium-competitive acid blocker (P-CAB) with a novel
were incubated with different concentrations of a [14C] mode of action—physicochemical properties, non-clinical
vonoprazan and b [14C] LPZ for 15, 30, 60, and 120 min. data, and a proposed mode of action. Prog Med
Accumulation was measured after washing. Reproduced 2014;34:2183–2194
with permission from K. Otake, Y. Sakurai, H. Nishida

ATPase families [58]. For this reason, inhibitory effect of vonoprazan was sustained
dissociation of the inhibitor requires a major even at 8 h after washout [59].
conformational change with an orientation that
is the reverse of binding, and with dissociation Maximum Effect is Sustained over a Long
rate that is extremely slow. Period
Vonoprazan can accumulate over a longer
period in the gastric fundic glands than PPIs. A study was performed to demonstrate the
Results of accumulation experiments conducted duration of effect of vonoprazan in rats. When
with respect to vonoprazan and lansoprazole drug concentrations in plasma and the stomach
using isolated primary cultured rabbit gastric wall were measured up to 24 h after oral
fundic glands are shown in Fig. 6. Remaining administration of 2 mg/kg of [14C] vonoprazan
concentration measurements after conducting to rats, vonoprazan concentrations in plasma
washout following incubation of [14C] and the stomach wall at 0.25, 1, 2, and 24 h
14
vonoprazan and [ C] lansoprazole at various after administration were 23, 11, 5, and 0 ng/
concentrations (0.01, 0.03, 0.1, 0.3 lM) with ml, respectively, in plasma and 2412, 957, 941,
primary cultured rabbit gastric fundic glands for and 20 ng/g, respectively, in the stomach wall
15, 30, 60, and 120 min, showed that (Fig. 7). These results show that following oral
vonoprazan was accumulated at higher administration to rats, vonoprazan rapidly
concentrations than lansoprazole [59]. A study disappears from plasma, but remains in the
of the acid secretion inhibitory effect after stomach even after 24 h [39].
incubating rabbit cultured gastric fundic A study of the inhibitory effect on
glands with vonoprazan and lansoprazole for histamine-stimulated gastric acid secretion up
2 h showed that the acid secretion inhibitory to 48 h after oral administration of vonoprazan
effect of lansoprazole disappeared immediately and lansoprazole to Heidenhain pouch dogs
after washout, while the acid secretion showed that when 1 mg/kg of vonoprazan was
Adv Ther

inhibitory effect of vonoprazan is more potent


and longer-lasting than that of lansoprazole,
and that its effect is not influenced by gastric
acid secretory conditions.
These results are also supported by clinical
pharmacology testing. In a pH monitoring test
of healthy adults, vonoprazan exhibited a
dose-dependent acid secretion inhibitory
effect. In the vonoprazan 40 mg
administration group, the acid secretion
inhibitory effect (pH[6) was sustained for
Fig. 7 Concentration of vonoprazan in the plasma and almost 24 h [48, 49]. A study of the food-effect
gastric wall of rats after oral administration. Plasma
concentration (ng/mL) and gastric wall concentration on the pharmacokinetics of vonoprazan showed
(ng/g) were measured at 0.25, 1, 2, and 24 h after oral that the total drug exposure (area under the
administration of 2 mg/kg of [14C] vonoprazan to rats. curve from time 0 to 48 h; AUC0–48) and Cmax of
Reproduced with permission from K. Otake, Y. Sakurai, vonoprazan were similar in both fasting and fed
H. Nishida et al. Vonoprazan fumarate (TAK-438), a new
potassium-competitive acid blocker (P-CAB) with a novel conditions [60]. These results show that the
mode of action—physicochemical properties, non-clinical maximum effect of vonoprazan is sustained for
data, and a proposed mode of action. Prog Med 24 h, and is unaffected by gastric pH or meals.
2014;34:2183–2194 It is well known that the efficacy of PPIs is
affected by food intake, and it is recommended
administered, an inhibitory effect of that they are prescribed to be taken 30–60 min
approximately 100, 80, and 60% was sustained before food intake for optimal efficacy [61]. On
from 1 to 6, 24, and 48 h, respectively. On the the other hand, the efficacy of vonoprazan is
other hand, lansoprazole exhibited an unaffected by food intake, is highly acid stable,
inhibitory effect of approximately 100% at 1 h and has an acid secretion inhibitory effect that
after administration of 3 mg/kg, but the effect is not influenced by gastric acid secretory
was not sustained, and had almost completely conditions [39]. Therefore, vonoprazan is also
disappeared at 48 h after administration (Fig. 8) superior to PPIs in terms of administration
[39]. Another study showed that inhibitory irrespective of food intake.
effects on histamine-stimulated gastric acid
secretion from 4h after intraperitoneal THE MECHANISM OF ACTION
administration of vonoprazan (0.7 mg/kg) or OF VONOPRAZAN ESTIMATED
lansoprazole (1 mg/kg) to rats whose gastric pH FROM THE DATA
had been temporarily raised to near neutral by
intravenous administration of cimetidine The aforementioned characteristics of
(30 mg/kg), an H2RA, the acid secretion vonoprazan can be explained by the following
inhibitory effect of vonoprazan was sustained mechanism of action in comparison with the
and unaffected by pH, while lansoprazole mechanism of action of conventional PPIs.
exhibited an acid secretion inhibitory effect When PPIs are orally administered, the
only under acidic conditions (Fig. 9) [39]. These enteric-coated formulation is not dissolved in
results reveal that the gastric acid secretion the stomach. However, since movement inside
Adv Ther

Fig. 8 Inhibitory effect of vonoprazan and LPZ on Otake, Y. Sakurai, H. Nishida et al. Vonoprazan fumarate
histamine-stimulated gastric acid secretion in Heidenhain (TAK-438), a new potassium-competitive acid blocker
pouch dogs. Vonoprazan and LPZ were administered orally. (P-CAB) with a novel mode of action—physicochemical
Histamine 2HCl (30 lg/kg) was injected subcutaneously properties, non-clinical data, and a proposed mode of
1 day before and 1, 3, 6, 24 and 48 h after drug and vehicle action. Prog Med 2014;34:2183–2194
administration. Reproduced with permission from K.

Fig. 9 Inhibitory effect of vonoprazan and LPZ on collected 3 h after histamine administration. Reproduced
histamine-stimulated gastric acid secretion in rats after with permission from K. Otake, Y. Sakurai, H. Nishida
cimetidine pretreatment. Cimetidine (30 mg/kg) or a et al. Vonoprazan fumarate (TAK-438), a new
vehicle was injected intravenously, and after 15 min potassium-competitive acid blocker (P-CAB) with a novel
vonoprazan at 0.7 mg/kg, LPZ at 1 mg/kg, or a vehicle mode of action—physicochemical properties, non-clinical
was administered intraperitoneally. The pylorus was ligated data, and a proposed mode of action. Prog Med
and histamine (30 mg/kg s.c.) was administered 4 h after 2014;34:2183–2194
drug or vehicle administration. Gastric contents were
Adv Ther

Fig. 10 Comparison of mechanism of action of vonoprazan (TAK-438), a new potassium-competitive acid blocker
and PPIs (hypothetical). a Suppression of gastric acid (P-CAB) with a novel mode of action—physicochemical
secretion by vonoprazan. b Suppression of gastric acid properties, non-clinical data, and a proposed mode of action.
secretion by PPIs. Adapted with permission from K. Otake, Prog Med 2014;34:2183–2194
Y. Sakurai, H. Nishida et al. Vonoprazan fumarate

the digestive tract is influenced by gastric secretion inhibitory effects do not differ
peristalsis and gastric emptying, variations significantly among individuals. Vonoprazan,
occur in the time required to reach the small which is basic, diffuses by passive transport
intestine [62]. Furthermore, PPIs, which are from the parietal cell cytoplasm into the
absorbed in the small intestine undergo secretory canaliculi in high concentrations.
hepatic metabolism to differing degrees by the The vonoprazan molecule which has now
genetically polymorphic CYP2C19, resulting in migrated into acidic space, and which has
interindividual variations in drug exposure [63]. undergone protonation in a strongly acidic
Vonoprazan, which does not need to be an environment, non-covalently binds with
enteric-coated formulation, readily dissolves in H?,K?-ATPase in a K?-competitive manner,
the stomach when orally administered, and thereby blocking H? secretion. In a strongly
moves to the small intestine where it is acidic environment, when the ratio of the
absorbed by the mucosa of the small intestine. non-ionic type of vonoprazan decreases, and
Unlike PPIs, vonoprazan is not primarily membrane permeability declines, passive
metabolized by CYP2C19, and thus, the acid transport from the acidic space in the parietal
Adv Ther

cell to the cytoplasm is inhibited, and it is for treatment of acid-related diseases, including
retained for a long period inside the parietal GERD, and for H. pylori eradication.
cell. Consequently, vonoprazan is also capable The characteristics of vonoprazan are
of exhibiting inhibitory action against summarized as follows.
H?,K?-ATPase that is activated by further • It is stable in acid.
stimulation of acid secretion that occurs after • It has satisfactory solubility under acidic to
the decline in blood concentration. neutral conditions.
On the other hand, when PPIs accumulate in • It has satisfactory membrane permeability
the secretory canaliculus of the parietal cells, under neutral conditions.
they are converted to the active form in the • It has stronger H?,K?-ATPase inhibitory
presence of acid. H?,K?-ATPase pumps are activity than PPIs under acidic to neutral
activated primarily by food and the active conditions, and its activity is minimally
form of the PPI blocks secretion of H? by affected by pH.
covalently binding with H?,K?-ATPase. • CYP2C19 has little effect on its metabolism.
?
The dissociation rate of vonoprazan from H , • There is no need for conversion to an active
?
K -ATPase is slow, and it is not decomposed by form.
acid. Therefore, binding of vonoprazan to • Its rate of dissociation from H?,K?-ATPase is
H?,K?-ATPase is maintained even after secretion very slow.
stimulus ceases. H?, K?-ATPase on the apical • Its retention time in the gastric mucosa is
membrane of the secretory canaliculus is again long (24 h or more).
taken into the tubulovesicles and migrates into Vonoprazan non-covalently binds
? ? ?
the parietal cell. H ,K -ATPase in a K -competitive manner,
The plasma half-life of PPIs is relatively thereby inhibiting its activity. Based on the
short—approximately 1–2 h. Consequently, aforementioned characteristics of vonoprazan,
after PPIs have been eliminated from the it can be expected to not only demonstrate an
blood, they no longer exhibit inhibitory action adequate clinical effect from the first dose, but
? ?
against newly activated H ,K -ATPase. The also sustain its maximum effect over a long
potent acid inhibitory effect is not period. In that vonoprazan functionally inhibits
demonstrated after the first dose, and the H?,K?-ATPase as a result of competing with the
amount of inactivated H?, K?-ATPase increases binding of K? ions, it is different from
over repeated administration. Therefore, it is conventional PPIs that structurally inhibit
necessary to repetitively administer PPIs over H?,K?-ATPase. We believe that it is important
3–5 days to reach steady-state inhibition of acid to thoroughly understand the similarities and
secretion [64]. differences between vonoprazan and
The mechanism described above is conventional PPIs in order to effectively and
schematically represented in Fig. 10. safely use vonoprazan, and we hope that this
paper is helpful in this regard.
CONCLUSION Phase III clinical trials for vonoprazan in
Japan have been completed for indications
Vonoprazan is an acid secretion inhibitor, including reflux esophagitis, gastric ulcers,
which has overcome the limitations of PPIs duodenal ulcers, and H. pylori eradication.
that are currently the first choice among drugs Vonoprazan has demonstrated efficacy and
Adv Ther

favorable safety and tolerability profiles in English was granted by the publisher of
[35–38]. PPIs with a similar inhibitory effect Progress in Medicine. Kazuyoshi Otake, Yuichi
on acid secretion to vonoprazan are also Sakurai, Haruyuki Nishida, Hideo Fukui,
reported to increase risk of fracture [65] and Yoshihiko Tagawa, Hitomi Yamasaki,
increase risk of gastrointestinal infection with Masatoshi Karashima, Keiichi Otsuka,
Clostridium difficile [66]. Given that Nobuhiro Inatomi. Vonoprazan Fumarate
vonoprazan blocks gastric acid production by (TAK-438). A New Potassium-Competitive Acid
? ?
inhibiting the same H ,K -ATPase enzyme as a Blocker (P-CAB) with a Novel Mode of Action—
PPI, it could be hypothesized that vonoprazan physicochemical Properties, Non-Clinical Data,
might result in similar risks and may also and a Proposed Mode of Action. Progress in
increase the degree of elevation in gastrin Medicine 2014:34(12):2183–2194. Translation of
levels. Details of these clinical trial results and the article from Japanese to English was funded
safety of vonoprazan will be presented in by Takeda Pharmaceuticals USA, Inc. and
future papers. performed by a native Japanese-speaking
translator specialized in the life sciences. The
translation was conducted according to rigid
ACKNOWLEDGMENTS ISO 9001:2008 and EN 15038:2006 certified
quality assurance processes. This article is a
Sponsorship and article processing charges for
translation of the original Japanese article to
this study were funded by Takeda
English with updates.
Pharmaceutical Company Limited.
All named authors meet the International
We express our thanks to Mr. Masayuki Aboshi
Committee of Medical Journal Editors (ICMJE)
and Mr. Shigeki Okita of the Medical Science
criteria for authorship for this manuscript, take
Liaison Group, Medical Affairs Department,
responsibility for the integrity of the work as a
Japan Research Center, Takeda Pharmaceutical
whole, and have given final approval for the
Company Limited, and Mr. Francois Jones of
version to be published.
Quintiles Transnational Japan Co., Ltd. for their
support in drafting this article. We also thank
Disclosures. Kazuyoshi Otake, Yuuichi
the Medical Science Liaison Group, Medical
Sakurai, Haruyuki Nishida, Hideo Fukui,
Affairs Department, Japan Research Center,
Yoshihiko Tagawa, Hitomi Yamasaki,
Takeda Pharmaceutical Company Limited for
Masatoshi Karashima, Keiichi Otsuka and
providing scientific advice. Dr. Keisuke Ishida of
Nobuhiro Inatomi are employees of Takeda
SunFlare Co., Ltd. provided medical writing
Pharmaceutical Company Limited, and there
support, which was funded by Takeda
are no other applicable matters regarding
Pharmaceutical Company Limited. Mr.
conflicts of interest.
Yoshitaka Sato of Saikou, provided medical
illustration service, which was funded by Compliance with Ethics Guidelines. This
Takeda Pharmaceutical Company Limited. article is based on previously conducted
This article was originally published in studies and does not involve any new studies
Japanese in the journal Progress in Medicine. of human or animal subjects performed by any
Permission to translate and republish the article of the authors.
Adv Ther

REFERENCES 14. Sachs G, Meyer-Rosberg K, Scott DR, Melchers K.


Acid, protons and Helicobacter pylori. Yale J Biol
Med. 1996;69:301–16.
1. Huang JQ, Hunt RH. pH, healing rate and
symptomatic relief in acid-related diseases. Yale J 15. Asaka M, Kato M, Takahashi S, et al. Guidelines for
Biol Med. 1996;69:159–74. the management of H. pylori infections: 2009
revised edition. Helicobacter. 2010;15:1–20.
2. Hirschowitz BI, Keeling D, Lewin M, et al.
Pharmacological aspects of acid secretion. Dig Dis 16. Yuan Y, Wang CC, Yuan YH, et al. The proportion
Sci. 1995;40(2 Suppl):3S–23S. of patients who are free of reflux symptoms during
the initial days of treatment with proton pump
3. Urushidani T, Nagao T. Calyculin A, a inhibitors (PPIs) in GERD trials: a meta-analysis.
phosphoprotein phosphatase inhibitor, stimulates Gastroenterology. 2008;134(Suppl 1):A174.
acid secretion in isolated gastric glands. Am J
Physiol. 1996;270(1 Pt 1):G103–12. 17. Hunt RH, Scarpignato C. Potassium-competitive acid
blockers (P-CABs): are they finally ready for prime
4. Forte JG, Ganser A, Beesley R, Forte TM. Unique time in acid-related disease? Clin Trans Gastroenterol.
enzymes of purified microsomes from pig fundic 2015;6:e119. doi:10.1038/ctg.2015.39.
mucosa. K?-stimulated adenosine triphosphatase
and K?-stimulated pNPPase. Gastroenterology. 18. Scarpignato C, Hunt RH. Proton pump inhibitors:
1975;69:175–89. the beginning of the end or the end of the beginning?
Curr Opin Pharmacol. 2008;8(6):677–84.
5. Chow DC, Forte JG. Functional significance of the
beta-subunit for heterodimeric P-type ATPases. 19. Tytgat GN. Are there unmet needs in acid
J Exp Biol. 1995;198:1–17. suppression? Best Pract Res Clin Gastroenterol.
2004;18(Suppl):67–72.
6. Rabon E, Cuppoletti J, Malinowska D, et al. Proton
secretion by the gastric parietal cell. J Exp Biol. 20. Fass R, Shapiro M, Dekel R, Sewell J. Systematic
1983;106:119–33. review: proton-pump inhibitor failure in
gastro-oesophageal reflux disease - where next?
7. Herrmann M, Selige J, Raffael S, Sachs G, Brambilla Aliment Pharmacol Ther. 2005;22(2):79–94.
A, Klein T. Systematic expression profiling of the
gastric H?/K? ATPase in human tissue. Scand J 21. Japanese Society for Gastroenterology. Guidelines
Gastroenterol. 2007;42:1275–88. for the management of GERD (2009). http://
minds4.jcqhc.or.jp/minds/GERD/gerd_gl.pdf.
8. Kraut JA, Helander KG, Helander HF, Iroezi ND,
Marcus EA, Sachs G. Detection and localization of 22. Mégraud F. H pylori antibiotic resistance:
H?-K?-ATPase isoforms in human kidney. Am J prevalence, importance, and advances in testing.
Physiol Renal Physiol. 2001;281(4):F763–8. Gut. 2004;53:1374–84.

9. Forte JG, Hanzel DK, Okamoto C, Chow D, 23. Graham DY, Fischbach L. Helicobacter pylori
Urushidani T. Membrane and protein recycling treatment in the era of increasing antibiotic
associated with gastric HCl secretion. J Intern Med resistance. Gut. 2010;59:1143–53.
Suppl. 1990;732:17–26.
24. Ashida K. Notes on features and uses of medications
10. Howden CW, Burget DW, Hunt RH. Appropriate used for PPI-resistant GERD, drugs under
acid suppression for optimal healing of duodenal development and future prospects. Jpn J Med
ulcer and gastro-oesophageal reflux disease. Scand J Pharm Sci. 2014;71:591–6.
Gastroenterol Suppl. 1994;201:79–82.
25. Stewart B, Wallmark B, Sachs G. The interaction of
11. Bell NJ, Burget D, Howden CW, Wilkinson J, Hunt H? and K? with the partial reactions of gastric
RH. Appropriate acid suppression for the (H??K?)-ATPase. J Biol Chem. 1981;256:2682–90.
management of gastro-oesophageal reflux disease.
Digestion. 1992;51(Suppl 1):59–67. 26. Mendlein J, Sachs G. Interaction of a K?
-competitive inhibitor, a substituted
12. Hunt RH. Importance of pH control in the imidazo[1,2a]pyridine, with the phospho- and
management of GERD. Arch Intern Med. dephosphoenzyme forms of H?, K?-ATPase. J Biol
1999;159:649–57. Chem. 1990;265:5030–6.

13. Mössner J, Caca K. Developments in the inhibition 27. Parsons ME, Keeling DJ. Novel approaches to the
of gastric acid secretion. Eur J Clin Invest. pharmacological blockade of gastric acid secretion.
2005;35:469–75. Expert Opin Investig Drugs. 2005;14:411–21.
Adv Ther

28. Kahrilas PJ, Dent J, Lauritsen K, et al. A randomized, second-line triple therapy for Helicobacter pylori
comparative study of three doses of AZD0865 and eradication: a phase III, randomised, double-blind
esomeprazole for healing of reflux esophagitis. Clin study. Gut. 2016. doi:10.1136/gutjnl-2015-311304
Gastroenterol Hepatol. 2007;5:1385–91. (Epub ahead of print).

29. Berg AL, Böttcher G, Andersson K, et al. Early 39. Hori Y, Matsukawa J, Takeuchi T, et al. A study
stellate cell activation and veno-occlusive-disease comparing the antisecretory effect of TAK-438, a
(VOD)-like hepatotoxicity in dogs treated with novel potassium-competitive acid blocker, with
AR-H047108, an imidazopyridine proton pump lansoprazole in animals. J Pharmacol Exp Ther.
inhibitor. Toxicol Pathol. 2008;36:727–37. 2011;337:797–804.

30. Dent J, Kahrilas PJ, Hatlebakk J, et al. A randomized, 40. Hori Y, Imanishi A, Matsukawa J, et al. 1-[5-(2-
comparative trial of a potassium-competitive acid Fluorophenyl)-1-(pyridin-3-ylsulfonyl)-1H-pyrrol-3-
blocker (AZD0865) and esomeprazole for the yl]-N-methylmethanamine monofumarate (TAK-
treatment of patients with nonerosive reflux 438), a novel and potent potassium-competitive
disease. Am J Gastroenterol. 2008;103:20–6. acid blocker for the treatment of acid-related
diseases. J Pharmacol Exp Ther. 2010;335:231–8.
31. Kondo M, Kawamoto M, Hasuoka A, et al.
High-throughput screening of potassium- 41. Adachi K, Katsube T, Kawamura A, et al. CYP2C19
competitive acid blockers. J Biomol Screen. genotype status and intragastric pH during dosing
2012;17:177–82. with lansoprazole or rabeprazole. Aliment
Pharmacol Ther. 2000;14(10):1259–66.
32. Nishida H, Hasuoka A, Arikawa Y, et al. Discovery,
synthesis, and biological evaluation of novel 42. Furuta T, Ohashi K, Kosuge K, et al. CYP2C19
pyrrole derivatives as highly selective genotype status and effect of omeprazole on
potassium-competitive acid blockers. Bioorg Med intragastric pH in humans. Clin Pharmacol Ther.
Chem. 2012;20:3925–38. 1999;65:552–61.

33. Arikawa Y, Nishida H, Kurasawa O, et al. Discovery 43. Shirai N, Furuta T, Xiao F, et al. Comparison of
of a novel pyrrole derivative 1-[5-(2- lansoprazole and famotidine for gastric acid
fluorophenyl)-1-(pyridin-3-ylsulfonyl)-1H-pyrrol-3- inhibition during the daytime and night-time in
yl]-N-methylmethanaminefumarate (TAK-438) as a different CYP2C19 genotype groups. Aliment
potassium-competitive acid blocker (P-CAB). J Med Pharmacol Ther. 2002;16(4):837–46.
Chem. 2012;55:4446–56.
44. Yamada S, Onda M, Kato S, et al. Genetic
34. Takeda Takecab (vonoprazan tablets). Japanese differences in CYP2C19 single nucleotide
Common Technical Document. Available at: polymorphisms among four Asian populations.
http://www.pmda.go.jp/drugs/2014/P201400173/ J Gastroenterol. 2001;36:669–72.
index.html. Accessed 20 Apr 2016.
45. Furuta T, Shirai N, Kodaira M, et al.
35. Ashida K, Sakurai Y, Hori T, et al. Randomised clinical Pharmacogenomics-based tailored versus standard
trial: vonoprazan, a novel potassium-competitive therapeutic regimen for eradication of H. pylori.
acid blocker, vs. lansoprazole for the healing of Clin Pharmacol Ther. 2007;81:521–28.
erosive oesophagitis. Aliment Pharmacol Ther.
2016;43(2):240–51. 46. Furuta T, Shirai N, Watanabe F, et al. Effect of
cytochrome P4502C19 genotypic differences on
36. Mizokami Y, Ashida K, Soen S, et al. TAK-438 versus cure rates for gastroesophageal reflux disease by
lansoprazole 15 mg for secondary prevention of lansoprazole. Clin Pharmacol Ther.
peptic ulcers associated with non-steroidal 2002;72:453–60.
anti-inflammatory drug (NSAID) therapy: results
of a phase 3 trial. Gastroenterology. 47. Kagami T, Sahara S, Ichikawa H, et al. Potent acid
2014;146(5,Suppl 1):S-739. inhibition by vonoprazan in comparison with
esomeprasole, with reference to CYP2C19
37. Kawai T, Ashida K, Mizokami Y, et al. TAK-438 genotype. Aliment Pharmacol Ther.
versus lansoprazole 15 mg for secondary prevention 2016;43(10):1048–59.
of peptic ulcers associated with low-dose aspirin
therapy: results of a phase 3 trial. Gastroenterology. 48. Sakurai Y, Nishimura A, Kennedy G, et al. Safety,
2014;146(5,Suppl 1):S-739. tolerability, pharmacokinetics, and
pharmacodynamics of single rising TAK-438
38. Murakami K, Sakurai Y, Shiino M, et al. (vonoprazan) doses in healthy male Japanese/
Vonoprazan, a novel potassium-competitive non-Japanese subjects. Clin Transl Gastroenterol.
acid blocker, as a component of first-line and 2015;25(6):e94.
Adv Ther

49. Jenkins H, Sakurai Y, Nishimura A, et al. Randomised 58. Abe K, Tani K, Fujiyoshi Y. Conformational
clinical trial: safety, tolerability, pharmacokinetics rearrangement of gastric H?, K?-ATPase induced
and pharmacodynamics of repeated doses of TAK- by an acid suppressant. Nat Commun.
438 (vonoprazan), a novel potassium-competitive 2011;2:155–61.
acid blocker, in healthy male subjects. Aliment
Pharmacol Ther. 2015;41(7):636–48. 59. Matsukawa J, Hori Y, Nishida H, kajino M, Inatomi
N. A comparative study on the modes of action of
50. Nagaya H, Satoh H, Kubo K, Maki Y. Possible TAK-438, a novel potassium-competitive acid
mechanism for the inhibition of gastric blocker, and lansoprazole in primary cultured
(H??K?)-adenosine triphosphatase by the proton rabbit gastric glands. Biochem Pharmacol.
pump inhibitor AG-1749. J Pharmacol Exp Ther. 2011;81:1145–51.
1989;248:799–805.
60. Takeda Takecab (vonoprazan tablets) package
51. Fock KM, Ang TL, Bee LC, Lee EJ. Proton pump insert. Available at: http://www.pmda.go.jp/
inhibitors: do differences in pharmacokinetics PmdaSearch/iyakuDetail/ResultDataSetPDF/400256_
translate into differences in clinical outcomes? 2329030F1020_1_04. Accessed 20 Apr 2016.
Clin Pharmacokinet. 2008;47:1–6.
61. Hatlebakk JG, Katz PO, Camacho-Lobato L, Castell
52. Gedda K, Scott D, Besancon M, Lorentzon P, Sachs DO. Proton pump inhibitors: better acid
G. Turnover of the gastric H?, K?-adenosine suppression when taken before a meal than
triphosphatase a subunit and its effect on without a meal. Aliment Pharmacol Ther. 2000;
inhibition of rat gastric acid secretion. 14(10):1267–72.
Gastroenterology. 1995;109:1134–41.
62. Wada F, Murase K, Isomoto H, et al. Polymorphism
53. Sachs G. Improving on PPI-based therapy of GORD. of CYP2C19 and gastric emptying in patients with
Eur J Gastroenterol Hepatol. 2001;13(Suppl 1): proton pump inhibitor-resistant gastric ulcers. J Int
S35–41. Med Res. 2002;30:413–21.

54. Bytzer P, Blum AL. Personal view: rationale and 63. Klotz U. Clinical impact of CYP2C19
proposed algorithms for symptom-based proton polymorphism on the action of proton pump
pump inhibitor therapy for gastro-oesophageal inhibitors: a review of a special problem. Int J Clin
reflux disease. Aliment Pharmacol Ther. Pharmacol Ther. 2006;44:297–302.
2004;20(4):389–98.
64. Andersson K, Carlsson E. Potassium-competitive
55. Sachs G, Shin JM, Howden CW. Review article: the acid blockade: a new therapeutic strategy in
clinical pharmacology of proton pump inhibitors. acid-related diseases. Pharmacol Ther. 2005;108:
Aliment Pharmacol Ther. 2006;23(Suppl 2):2–8. 294–307.

56. Tytgat GN. Shortcomings of the first-generation 65. Zhou B, Huang Y, Li H, et al. Proton-pump
proton pump inhibitors. Eur J Gastroenterol inhibitors and risk of fractures: an update
Hepatol. 2001;13(Suppl 1):S29–33. meta-analysis. Osteoporos Int. 2016;27:339–47.

57. Shin JM, Inatomi N, Munson K, et al. 66. Cunningham R, Dial S. Is over-use of proton pump
Characterization of a novel potassium-competitive inhibitors fuelling the current epidemic of
acid blocker of the gastric H, K-ATPase, Clostridium difficle-associated diarrhea? J Hosp
1-[5-(2-fluorophenyl)-1-(pyridin-3-ylsulfonyl)-1H- Infect. 2008;70:1–6.
pyrrol-3-yl]-N-methylmethanamine monofumarate
(TAK-438). J Pharmacol Exp Ther. 2011;339:412–20.

You might also like