ArticlePDF AvailableLiterature Review

Proton Pump Inhibitors and Radiofrequency Ablation for Treatment of Barrett's Esophagus

Authors:

Abstract and Figures

Gastroesophageal reflux disease (GERD) is characterized by acid and bile reflux in the distal oesophagus, and this may cause the development of reflux esophagitis and Barrett’s oesophagus (BE). The natural histological course of untreated BE is non-dysplastic or benign BE (ND) than low-grade (LGD), and high-grade dysplastic (HGD) BE, with the expected increase in malignancy transfer to oesophagal adenocarcinoma (EAC). The gold standard for BE diagnostics involves high-resolution white-light endoscopy, followed by uniform endoscopy findings description (Prague classification) with biopsy performance according to Seattle protocol. The medical treatment of GERD and BE includes the use of proton pump inhibitors (PPIs) regarding symptoms control. It is noteworthy that long-term use of PPIs increases gastrin level, which can contribute to transfer from BE to EAC, as a result of its effects on proliferation of BE epithelium. Endoscopy treatment includes a wide range of resection and ablative techniques, such as radio-frequency ablation (RFA), often concomitantly used in everyday endoscopy practice (multimodal therapy). RFA promotes mucosal necrosis of treated oesophagal region via high-frequency energy. Laparoscopic surgery, partial or total fundoplication, is reserved for PPIs and endoscopy indolent patients or in those with progressive disease. This review aims to explain distinct effects of PPIs and RFA modalities, illuminate certain aspects of molecular mechanisms involved, as well as the effects of their concomitant use regarding the treatment of BE and prevention of its transfer to EAC.
Content may be subject to copyright.
Send Orders for Reprints to reprints@benthamscience.net
Mini-Reviews in Medicinal Chemistry, 2020, 20, 000-000 1
REVIEW ARTICLE
1389-5575/20 $65.00+.00 © 2020 Bentham Science Publishers
Proton Pump Inhibitors and Radiofrequency Ablation for Treatment of
Barrett's Esophagus
Predrag Dugalic1,*, Srdjan Djuranovic2, Aleksandra Pavlovic-Markovic2, Vladimir Dugalic3,
Ratko Tomasevic4, Zoran Gluvic5, Milan Obradovic6, Vladan Bajic6 and Esma R. Isenovic6,*
1Department of Gastroenterology and Hepatology, University Clinical-Hospital Centre Zemun-Belgrade, Belgrade,
Serbia; 2Clinical Centre of Serbia, Clinic for Gastroenterology and Hepatology, Faculty of Medicine, University of Bel-
grade, Belgrade, Serbia; 3Clinical Centre of Serbia, Clinic for Surgery, Faculty of Medicine, University of Belgrade,
Belgrade, Serbia; 4Department of Gastroenterology and Hepatology, Faculty of Medicine, University of Belgrade, Uni-
versity Clinical-Hospital Centre Zemun-Belgrade, Belgrade, Serbia; 5Department of Endocrinology and Diabetes, Fac-
ulty of Medicine, University of Belgrade, University Clinical-Hospital Centre Zemun-Belgrade, Belgrade, Serbia;
6Department of Radiobiology and Molecular Genetics, Institute of Nuclear Sciences Vinca, University of Belgrade, Bel-
grade, Serbia
Abstract: Gastroesophageal reflux disease (GERD) is characterized by acid and bile reflux in the dis-
tal oesophagus, and this may cause the development of reflux esophagitis and Barrett’s oesophagus
(BE). The natural histological course of untreated BE is non-dysplastic or benign BE (ND), then low-
grade (LGD) and high-grade dysplastic (HGD) BE, with the expected increase in malignancy transfer
to oesophagal adenocarcinoma (EAC). The gold standard for BE diagnostics involves high-resolution
white-light endoscopy, followed by uniform endoscopy findings description (Prague classification)
with biopsy performance according to Seattle protocol. The medical treatment of GERD and BE in-
cludes the use of proton pump inhibitors (PPIs) regarding symptoms control. It is noteworthy that long-
term use of PPIs increases gastrin level, which can contribute to transfer from BE to EAC, as a result
of its effects on the proliferation of BE epithelium. Endoscopy treatment includes a wide range of re-
section and ablative techniques, such as radio-frequency ablation (RFA), often concomitantly used in
everyday endoscopy practice (multimodal therapy). RFA promotes mucosal necrosis of treated
oesophagal region via high-frequency energy. Laparoscopic surgery, partial or total fundoplication, is
reserved for PPIs and endoscopy indolent patients or in those with progressive disease. This review
aims to explain distinct effects of PPIs and RFA modalities, illuminate certain aspects of molecular
mechanisms involved, as well as the effects of their concomitant use regarding the treatment of BE and
prevention of its transfer to EAC.
A R T I C L E H I S T O R Y
Received: April 01, 2019
Revised: June 04, 2019
Accepted: June 25, 2019
DOI:
10.2174/1389557519666191015203636
Keywords: GERD, Barrett’s oesophagus, proton pump inhibitors, radio-frequency ablation, action of PPIs, oesophagus.
1. INTRODUCTION
Barrett’s oesophagus (BE) represents a premalignant
condition, and it is considered to be a precursor for the de-
velopment of oesophagal adenocarcinoma (EAC) [1]. Gas-
troesophageal reflux disease (GERD) is characterized by
acid and bile reflux in the distal oesophagus, which can lead
*Address correspondence to these authors at the Department of Gastroen-
terology and Hepatology, University Clinical-Hospital Centre Zemun-
Belgrade, Belgrade, Serbia; E-mail: predrag.dugalic@gmail.com; and De-
partment of Radiobiology and Molecular Genetics, Institute of Nuclear
Sciences Vinca, University of Belgrade, Belgrade, Serbia; Tel: +38111-
3408147; E-mail: isenovic@yahoo.com
to the development of reflux esophagitis and BE [2, 3]. The
prevalence of BE in Western populations is under 5%, and
the estimate is around 1.5% [2, 4-6], while in patients with
GERD, the prevalence of BE is estimated to be 15% [7]. The
gold standard for BE diagnostics involves high-resolution
white-light endoscopy, followed by uniform endoscopy find-
ings description (Prague classification) with biopsy perfor-
mance according to Seattle protocol [8, 9]. Treatment of BE
could be medical, endoscopic, and surgical [10, 11]. Medical
treatment is necessary to control GERD and its associated
symptoms [10]. Medical treatment is related to proton pump
inhibitors (PPIs) use since H2 blockers do not demonstrate
the decrease in risk progression of BE to oesophagal adeno-
2 Mini-Reviews in Medicinal Chemistry, 2020, Vol. 20, No. 0 Dugalic et al.
carcinoma (EAC) [12-14]. In general, current international
and national endoscopy societies recommend endoscopic
treatment in the cases of confirmed high-grade dysplasia
(HGD) and early EAC. Radiofrequency ablation (RFA) is
among the most frequently used ablation techniques of BE
[10, 15]. RFA aims to destroy damaged squamous epithelium
and to allow the growth of neosquamous epithelium [16]. The
EAC rate may significantly decrease by eradicating BE meta-
plasia and dysplasia [17]). Currently, RFA is established as an
endoscopic technique for BE eradication as well as for the
eradication of residual BE after endoscopic resection of early
EAC [18]. RFA should not be the appropriate first choice for
the treatment of nodular BE. The best approach is to treat
visible or nodular BE by resection technique followed by
RFA [17, 19]. Surgery is reserved for patients with persisted
GERD symptoms despite the long-term use of PPIs. Another
indication for surgical treatment is a progressive disease.
Current laparoscopic techniques (partial or total fundoplica-
tion) are the surgical procedures of choice [11, 20]. This
review aims to explain distinct effects of medical (PPIs) and
tissue-acquiring endoscopy technique (RFA) modalities,
illuminate certain aspects of molecular mechanisms in-
volved, as well as the effects of their concomitant use (mul-
timodal therapy) regarding the treatment of BE and preven-
tion of its transfer to EAC.
2. BARRETT’S OESOPHAGUS (BE)
BE is the pathological condition where the normal strati-
fied squamous epithelium of distal oesophagus replaced with
metaplastic columnar epithelium (with goblet cells). BE rep-
resents a premalignant condition, and it is considered to be a
precursor for the development of EAC. Although Norman
Barrett described this condition in the middle of the last cen-
tury [1], some facts regarding the origin, pathogenesis, diag-
nostics, and prevention of BE are not completely understood
[21].
2.1. Predisposing Factors
Several risk factors for development BE have been
identified, where GERD is considered as a predominant risk
factor [2, 3]. The results from the meta-analysis study showed
that symptoms of GERD are associated with a five-fold in-
creased risk of BE [3], while the results from one cross-
sectional study performed on 683 patients younger than 30
years, with weekly symptoms of GERD, indicate even 15-
fold increased risk of BE [22]. Other strong risk factors for
BE include age, male gender, central obesity, white race,
smoking, and also genetic predisposition [6, 23-26]. The
prevalence of BE increases with age, and it is twice more
frequent in men than in women [24, 27].
Furthermore, cigarette smoking considerable amplifies
risk for development BE [28, 29]. Abdominal obesity has been
recognized as a significant risk factor for BE, especially in
patients with a genetic predisposition to develop obesity [30,
31]. Also, there is a positive correlation with insulin re-
sistance, type 2 diabetes, high serum concentrations of leptin,
metabolic syndrome, and sleep apnea with an increased risk of
BE [32-36]. Individuals with a positive family history of BE,
especially first-degree relatives with BE, have a significantly
increased risk of BE [23]. Interestingly, evidence from the
literature indicates that infection with Helicobacter pylori
decreases the risk of BE [37, 38], most likely through a
mechanism that includes reduction of ghrelin and leptin
signaling [39, 40].
2.2. Epidemiology
The prevalence of BE in the general population is not
easy to clarify because BE is asymptomatic, and its diagnosis
is accurate only when endoscopic examination and histologi-
cal confirmation are performed [41]. One of the rare studies
with randomly recruited adults (Swedish study), performed
on 1000 residents who underwent upper gastrointestinal en-
doscopy. The histological results from the Swedish study
confirmed BE in 1.6% subjects, where 5 subjects have a long
segment, and 11 subjects have a short segment [5]. Similarly,
the Loiano-Monghidoro study noted a 3.6% prevalence of
BE in 1033 subjects from Italy, while histological analysis
confirms Barrett’s metaplasia in 1.3% of all respondents [4].
Also, the Chinese SILC study found (by endoscopy) 1.8%
cases of BE in a population of 1030 subjects [42]. Further-
more, systematic review and meta-analysis of the prevalence
of BE in Asian countries which include 51 studies and
453,147 patients, reported that the endoscopic prevalence of
BE was 7.8%, while histologically confirmed prevalence of
BE was 1.3% [43]. Some differences in the prevalence of BE
between populations may be explained by the difference in
the prevalence of risk factors, primarily GERD, obesity, diet,
and smoking [44].
2.3. Origin and Pathogenesis
Besides numerous investigations, the origin of BE remains
poorly understood. Some hypotheses have been proposed to
the defined origin of BE, such as reprogramming of
oesophagal-squamous stem cells, repopulation from submu-
cosal oesophagal glands, or proximal migration of the gastric
cardiac epithelium with subsequent intestinalization [45-50].
By using p63-deficient mice, with a lack of squamous epi-
thelia (an animal model for GERD), Wang et al. reported
that embryos of these mice rapidly develop intestine-like
columnar metaplasia which replaces squamous mucosa, sim-
ilar to Barrett's metaplasia [46]. The same authors also
reported these precursor cells of metaplasia at the squamoco-
lumnar junction (SCJ) in adult mice and humans [46]. The
progressive inflammation, metaplasia, and dysplasia at the
SCJ were also found in another animal model of BE such as
human IL-1β transgenic mice, overexpressing interleukin-1
(IL-1) [47]. The development of BE lesion and progression
to EAC was accelerated by exposure to bile acids and nitros-
amines [47]. It is proposed that BE and EAC arise from gas-
tric progenitors by a mechanism involving IL-1β and IL-6
signaling cascade and Dll1-dependent Notch signaling [47].
Since the transitional zone between different types of epithe-
lium is a critical point for metaplastic and malignant chang-
es, Jiang et al. described a transitional columnar epithelium
with distinct basal progenitor cells (p63+KRT5+KRT7+) at
the SCJ in a tg mice model [46]. The same authors show that
these transitional basal progenitors cells could differentiate
into intestinal-like epithelium with goblet cells, and thus
represent the cells of origin for multi-layered epithelium and
Treatment of Barrett’s Esophagus Mini-Reviews in Medicinal Chemistry, 2020, Vol. 20, No. 0 3
BE [48]. Also, literature data indicate that refluxed gastric
juice plays a role in the pathogenesis of BE probably due to
cytokine-mediated inflammation and damage the oesophagus
tissue, rather than caustic acid injury [51, 52]. Other studies
also found that the clonality of Barrett’s glands underlies the
development of EAC [53, 54].
3. DIAGNOSTICS OF BE
Although the annual rate of BE transfer to the EAC is
small, the high mortality rate of this malignancy increases
the need for an early and reliable diagnosis. However, identi-
fying subjects with BE is a challenge for the physician with
the current diagnostic approach. Also, the use of biomarkers
to identify subjects with BE and for evaluation of risk-
stratification still needs additional assessment [55, 56].
The gold standard for BE diagnostics is high-resolution
white-light endoscopy, followed by uniform endoscopy find-
ings description (Prague classification) with biopsy perfor-
mance according to Seattle protocol [8, 9]. Based on Prague
classification, BE is arbitrarily defined depending on the
length of the columnar epithelium as a long segment (meta-
plastic segments length is equal or more than 3 cm), and as a
short segment (the length of BE is less than 3 cm) [8].
Changes less than 1 cm in length are not considered as BE,
but according to the American College of Gastroenterology,
these changes are defined as specialized intestinal metaplasia
(IM) of the oesophagogastric junction (irregular z-line) or
ultra-short segment BE [11].
Based the most guidelines, a diagnosis of BE implies the
presence of salmon-pink coloured columnar epithelium 1 cm
proximal to the gastroesophageal (GE) junction with biopsies
consistent with IM and the presence of mucin-containing gob-
let cells [11, 57, 58]. To increase the probability of finding IM,
it is recommended to increase the number of random biopsy
samples [59]. Use the Seattle protocol required four-quadrant
biopsy for every 1-2 cm of BE segment length [9]. The histo-
logical examination of biopsy samples are categorised as fol-
lows: negative for dysplasia or benign BE (ND), indefinite for
dysplasia, low-grade dysplasia (LGD), HGD, oesophagal ade-
nocarcinoma (EAC) [60, 61].
4. TREATMENT OF BARRETT’S OESOPHAGUS (BE)
Treatment of BE can be medical, endoscopic, and surgi-
cal [10, 11].
4.1. Medical Treatment
Medical treatment is necessary to control GERD and its
associated symptoms [10], and it is related to PPIs use be-
cause H2 blockers do not demonstrate the decrease in risk
progression of BE to oesophagal adenocarcinoma (EAC)
[10, 12-14]. Besides its acid-suppressive activity (chemopre-
vention), PPIs promote anti-inflammatory effects [11, 62].
The use of PPIs is now considered even in BE patients with
no reflux symptoms [19].
4.1.1. Proton Pump Inhibitors (PPIs)
The aim of PPIs use in the treatment of BE is to disrupt
the chain of GERD pathogenesis progression from NERD to
long-segment BE, and as from LGD to HGD and their pro-
gression to the final stage- EAC [10, 63, 64]. Today, statins
and PPIs are defined as factors that influence the natural histo-
ry of BE [41, 65]. Results from one meta-analysis demonstrate
a 71% risk reduction of neoplastic transfer of HGD [66]. The
benefits of PPIs are reflux symptoms and esophagitis control,
prevention of post-endoscopy treatment strictures, prevention
of dysplasia progression, and favouring of its regression
[67]. Some studies suggest the concomitant use of PPI with
NSAIDs/aspirin/statin treatment, regarding their synergistic
anti-inflammatory effects [68-70]. However, the routine use
of NSAIDs/aspirin cannot be recommended, partly because
of their serious adverse effects profile and questionable pro-
tective effects [11, 65]. Disagreements in different guidelines
regarding PPIs dosage are still subject to debates. Some so-
cieties advise using PPIs just in the cases of symptomatic
BE, but another one suggesting PPIs in doses higher than
usual for the control of reflux symptoms [8, 71, 72]. The use
of PPIs can favour neoplastic transformation of BE induced
by the increase in gastrin levels or by direct effects of bile
salts on oesophagal epithelium [9, 73]. It is recommended to
use PPIs once-daily, while the use of twice-daily PPIs regi-
men is reserved in the cases of poor reflux symptoms control
or due to esophagitis presence [11].
A turning point for the treatment of acid-related disorders
was in 1989 when omeprazole, a PPIs, was introduced to
clinical practice. The new class of drugs shows to be more
efficient, have better safety profiles, and more tolerant than
previously synthesized agents, such as histamine receptor
agonist, anticholinergic drugs, and analogues of the prosta-
glandin family [74]. After omeprazole in 1989, several ben-
zimidazole derivates have been synthesized [75], such are
lansoprazole, pantoprazole [74, 76], rabeprazole [74, 77],
and the stereo-isomeric compounds dexlansoprazole [78] and
esomeprazole [79]. Table 1 summarizes some properties of
different PPIs.
The name proton pump inhibitor comes from their prop-
erties to irreversibly bind to hydrogen/potassium adenosine-
triphosphatase (H+/K+-ATPase) enzyme that is found in gas-
tric parietal cells [80]. For most patients, PPIs represent the
first choice for treatment of GERD [81], dyspepsia [82], pep-
tic ulcer, cough, laryngitis [13], Zollinger-Ellison syndrome
[83], BE [63], NERD [84], and prevention of NSAID associ-
ated ulcers. PPIs are also a part of a treatment for eradication
caused by Helicobacter pylori [85].
4.1.2. The Chemical Action of (PPIs)
PPIs are heterocyclic organic molecules that have a
pyridine and benzimidazole moiety linked by a methylsulfinyl
groups (Fig. 1). PPIs as acid-activated prodrugs inhibit the
H+/K+-ATPase by binding covalently to active H+/K+-
ATPase [80, 86]. Evolutionary, H+/K+-ATPase have occurred
in cells independently more than once. The H+/K+-ATPase are
an integral part of the membrane, with the main role to in-
crease the proton gradient across the same membrane [87-89].
H+/K+-ATPase are divided into major classes regarding their
ability for utilizing different sources of energy, different pol-
ypeptide compositions, thus 3D conformation structure sug-
gesting different evolutionary origins [89, 90].
Please replace
4.1.2. The Chemical Action of (PPIs),
with
4.1.2. The Chemical Action of PPIs
4 Mini-Reviews in Medicinal Chemistry, 2020, Vol. 20, No. 0 Dugalic et al.
Table 1. PPIs chemical, structural, pharmacological, toxicological and metabolic properties.
PPIs Name
Chemical structure
Pharmacology
Toxicity/Side Effects
Refs.
Omeprazol
(approved in 1988)
N
N
S
O
O
N
O
H
Selective and irreversible
H+/K+-ATPase inhibitor
Peak plasma concentra-
tions: 1h
Plasma half-life is 1h
Clostridium difficile colitis
pneumonia
Risk of bone fractures
Potential of masking stom-
ach cancer
Hypomagnesaemia
Can interact with
clopidogrel, and oxyco-
done
[74]
[75]
[76]
[91]
Lansoprazol
(approved in 1988)
H
N
N
S
O
N
O
FF
F
Selective and irreversible
H+/K+-ATPase inhibitor.
Peak plasma concentrations
attained about 1.7h
Plasma half-life is 1h
Can bind to the pathogenic
form of Tau protein,
Hypomagnesaemia
[74]
[76]
[85]
[91]
Pantoprazol
(approved in 1994)
N
N
OF
F
S
O
H
N
O O
Selective and irreversible
H+/K+-ATPase inhibitor
Peak plasma concentrations:
2.5h
Plasma half-life is 1-2h
Headache
Diarrhea
Vomiting
Abdominal pain
Joint pain
Hypomagnesaemia
[74]
[76]
[85]
[91]
Rabeprazol
(approved in 1997)
N
N
S
O
H
N
O
O
Selective and irreversible
H+/K+-ATPase inhibitor
Peak plasma concentra-
tions: 1h
Plasma half-life is 1-2h
Osteoporosis
Hypomagnesaemia
Clostridium difficile infec-
tion
Pneumonia
[77]
[91]
Esomeprazol
(approved in 2001)
N
N
O
S
O
H
N
O
:
Selective and irreversible
H+/K+-ATPase inhibitor
Peak plasma concentra-
tions: 1h
Plasma half-life is 1.2 -1.4
Angioedema
Clostridium difficile infec-
tion
Pneumonia
Hypomagnesaemia
[79]
[84]
[85]
[91]
Dexlansoprazol
(approved in 2009)
an R-isomer of
Lansoprazol
N
N
S
O
H
N
O
FF
F
:
Selective and irreversible
H+/K+-ATPase inhibitor
Peak plasma concentra-
tions: 1h
Plasma half-life is 1h
Osteoporosis
Hypomagnesaemia
Clostridium difficile infec-
tion
Anaphylaxis
Pneumonia
[78]
[85]
[91]
Revaprazan
(approved in 2005)
F
N
N
NN
H
H+/K+-ATPase antagonist,
or potassium-competitive
acid blockers
Peak plasma concentra-
tions: 1.3-2.5h
Plasma half-life is 9h
Indigestion
Diarrhea
Constipation
Headache
[91]
[93]
[94]
The proton pump of the stomach is the H+/K+-ATPase,
which is located in parietal cells, which are a specialized
type of cells, concentrated in the inner lining of the stomach
(gastric mucosa). The H+/K+-ATPase is the major protein
concentrated within the membranes of parietal cells [90].
The role of this specific pump is the exchange of the potassi-
um anion from the intestinal lumen with hydronium within
the cytoplasm [80, 90]. The H+/K+-ATPase enzyme is re-
sponsible for the pH level in the stomach (acidification),
thereby physiologically responsible for protein digestion by
activating the digestive enzyme, pepsin [91]. As the H+/K+-
ATPase is at the last terminal stage of gastric secretion, it
becomes the best target for PPIs to inhibit acid secretion.
PPIs act by irreversibly blocking the H+/K+-ATPase enzyme
system (Fig. 2). PPIs are more effective than histamine
antagonists and PPIs reduce 99% of acid secretion from the
parietal cells to the gastric lumen. Despite the beneficial
effects of PPIs to decrease acid, several should be evaluated
before using PPIs, since secretion is needed for protein
digestion, vitamin B12, calcium, iron absorption, and other
nutrients.
PPIs are in essence pro-drugs, meaning that for their ac-
tivity, an activation process is needed. Activation of PPIs
Treatment of Barrett’s Esophagus Mini-Reviews in Medicinal Chemistry, 2020, Vol. 20, No. 0 5
occurs in the acidic environment of the stomach, in the parie-
tal cells, crossing the cell membrane (lipophilic) where the
inactive drug is protonated and thus rearranges to its active
form. This active form consequently covalently binds to the
H+/K+-ATPase and irreversibly inhibits its action to produce
H+ [19] (Fig. 2).
Since the irreversible properties to covalently bond to the
H+/K+-ATPase is dampened by the short half-life of current-
ly known PPIs like omeprazole (90 minutes) [91], other ap-
proaches have been proposed. One is the synthesis of APIs
or H+/K+-ATPase antagonist. APAs are competitive inhibi-
tors act by blocking acid secretion through K+ sites [92].
Revaprazan is an APA, with a faster action than PPIs as it
does not require acid activation [93, 94]. Revaprazan is reg-
istered in Korea, for gastritis, but not in Europe and the US.
To best of our knowledge, there are no known trials in
GERD and BE [90] for Revaprazan in the EU and US. Even
though Revaprazan provides better acid control as its action
lasts longer than PPIs. For severe acid-related problems such
as BE, PPIs are the prescribed drugs for treatment and cure
[74, 95].
4.1.3. The Risk and Benefit Ratio of PPIs
After more than 20 years of PPIs usage, some concerns
have been addressed related to the possible side effects [74,
96]. Previously, mutations in the α-subunit of H+/K+-ATPase
in mice had led to hypochlorhydria resulting in problems of
iron absorption [97] and consequently to anaemia. In hu-
mans, results of PPIs trials showed that the H+/K+-ATPase
does not play a fundamental role in iron absorption, so the
use of PPIs have not been correlated to the risk of iron ab-
sorption [97], but still some concerns remain regarded to
vitamin B12 absorption [98]. The most concern has been
raised from research on Benzimidazole derivatives, Astemi-
zole (AST) and Lansoprazole (LNS). The use of these deriv-
atives has shown to propagate interaction with a pathological
feature of Alzheimer’s disease, phosphorylated TAU, and its
aggregates (neurofibrillary tangles) leading to an increased
pace to dementia [99-101].
When concerning Alzheimer’s disease, research suggests
that PPIs are active in a non-selective manner, i.e., they act
on sodium-potassium pumps of the brain [102]. This interac-
tion could be utilized solely or by interaction with other
drugs, such as digoxin, warfarin, etc. The other focal point of
action may be associated with the sodium pump in the brain
or the alpha three subunits in astrocytes, a group of cells
important for memory. The chronic use of PPIs is considered
to cause down-regulation of the α subunit, thus damaging
astrocytes [103]. For the treatment of BE, PPIs are approved
for long term use and symptom control [104]. Some con-
cerns of PPIs adverse effects have been addressed in the ob-
servational studies conducted by Nehra et al., 2018; this
study only revelled mild overall associations [98] of PPIs
and their adverse effects suggest that the overall risk remains
small. Still, some recommendations are in effect, blood
count every year, B12 levels every five years, etc. in patients
on long therapy regimes with PPIs [98].
4.1.4. PPI and BE
A basic concern of the chronic use of PPIs should be
addressed, since it is well known that BE could lead to
adenocarcinoma by a progression of pathological stages,
from oesophagus inflammation to IM, then dysplasia, and
EAC. The long list of adverse effects related to PPIs shows
that the quality of evidence is still low [104]. So, in a clinical
setting, the use of PPIs is recommended for GERD, GERD
with BE, and NSAID bleeding prophylaxis, and for these
patients, PPIs show a high benefit-risk ratio, as for the other
groups or categories the risk-benefit ratio is less clear [104].
PPIs reduce acid reflux, which is one of the drivers of
BE, but PPIs also downregulate cyclooxygenase-2 expres-
sion suggesting that it might protect against neoplastic pro-
gression [15]. Results from observational data confirm that
patients taking PPIs have a reduced rate of neoplastic pro-
gression [104]. New studies where PPIs are giving with aspi-
rin [104], (since aspirin reduces the rate of oesophagal carci-
noma [15]), show that the best results were in groups with
the highest dose of Esomeprazole (2x40mg) and aspirin (325
mg once a day). Interestingly, the group of 704 patients
receiving high dose Esomeprazole had 303 serious adverse
events in comparison to high dose Esomeprazole and aspirin
that showed 274 events from 571 patients, suggesting that
there is little difference between these two groups [104]. The
overall results suggest that the combination of PPIs and aspi-
rin in the patients that well-tolerated this combination im-
proves the overall survival rate (death, cancer, and dysplasia)
in Barrett's syndrome patients for a time of 9 years [104].
Pharmacogenomic differences in users and non-users
(one time users) when considering the reaction to PPIs or
their adverse effects make it very difficult to study PPIs in
retrospect despite a vast number of studies. We stand on the
firm line that the overall quality of evidence to PPIs adverse
potential remains poor or very low [104]. Today clinicians
upheld recommendations [104] to appropriately prescribe
PPIs as then the benefits outweigh the risks, while inappro-
priately prescribed, even modest risks can out potential value
benefits.
Fig. (1). Schematic structure of PPIs (updated from Wikipedia).
PPIs have a common structure. All PPIs share a common back-
bone, a pyridine linked to a benzimidazole, the asterisk indicates
stereo centre (methylsulfinyl group).
XN
H
N
R1
SH
O
N
R4
OR2R3
Benzimidazole
Methylsulfinyl
group
Pyridine
moiety
6 Mini-Reviews in Medicinal Chemistry, 2020, Vol. 20, No. 0 Dugalic et al.
Fig. (2). Mechanism of action of the PPIs. The parietal cell represents the cell in the stomach lining that contains enzymes that make H+ and
also is the foundation for the H+/K+-ATPase. The H+/K+-ATPase is an enzyme system that actively transports hydrogen into the stomach and
K+ into the parietal cells by the mechanism of hydrolysis of ATP. All PPIs, from omeprazole to the most recently synthesized Dexlansopra-
zole (in 2009), are prodrugs and require activation. Once activated, they inhibit the H+/K+-ATPase. Their metabolites are inactive and do not
inhibit the H+/K+-ATPase. The next generation of PPIs has been presented in schematic presentation, i.e. acid pump antagonists are under
development. These agents, in which we used Revaprazan as a model suppress gastric activity by blocking the action of the H+/K+-ATPase by
binding to or near the site of the K+ channel. These compounds potentially achieve faster inhibition and longer duration in comparison to
classical PPIs. We also presented a class of drugs, H2RA or H2 blockers that act on histamine at the histamine H2 receptors in the parietal
cell. These drugs have their place in gastrointestinal management but for BER the preferred treatment is with PPIs. Gastrin is a peptide hor-
mone that stimulates acid secretion in two ways, directly through the parietal cell and indirectly through CCK2 receptors and through releas-
ing histamine acts on cells to produce H+ ions. Cl- - chloride ions, H+ - hydrogen ions, HCl - hydrochloric acid, K+ - potassium ions, H+/K+-
ATPase - hydrogen/potassium adenosine-triphosphatase. (A higher resolution/colour version of this figure is available in the electronic copy
of the article).
4.2. Endoscopic Treatment
In general, current international and national endoscopy
societies recommend endoscopic treatment in the cases of
confirmed HGD and early EAC. There is some doubt regard-
ing the endoscopic treatment of BE with confirmed LGD and
patients with simple BE-confirmed IM. This conundrum is
surpassed elegantly with regular chemoprevention and endo-
scopic surveillance in the cases of BE with no dysplasia and
performance of endoscopic treatment in selected patients
with LGD involving patients and physician “shared decision
making” [11, 105, 106].
Regarding modalities of applied endoscopic treatment,
the combined use of tissue acquiring or resection techniques
(i.e. endoscopic mucosal resection-EMR or endoscopic sub-
mucosal dissection-ESD) and ablative techniques (i.e. radiof-
requency ablation-RFA, argon plasma coagulation-APC,
cryoablation-CRY, photodynamic therapy-PDT, and multi-
polar electrocoagulation-MPEC) are currently recommended
in the patients with HGD, early EAC, and selected ones with
LGD [107]. The reason for endoscopic treatment of selected
patients with LGD and patients with HGD is expected high
progression risk to EAC (<13.4% and 10-18% patients per
year) [10, 106]. The precise pre-treatment endoscopic stag-
ing is the most important criterion that determines the choice
of therapy. The usual endoscopic approach to visible lesions
is EMR/ESD of the lesion of interest in combination with the
ablative endoscopic technique of endoscopists’ choice or
experience after histology examination (“multimodal thera-
py”) [17]. The importance of trained pathologists in BE is
cumbersome to ascertain patient risk for disease progression
because 50-85% of patients initially presented as LGD was
downstaged to non-dysplastic BE after expert pathologist
review [108]. According to that, confirmation of LGD by BE
experienced pathologist is crucial in deciding to ablate LGD
[109, 110].
Treatment of Barrett’s Esophagus Mini-Reviews in Medicinal Chemistry, 2020, Vol. 20, No. 0 7
Tissue-acquiring techniques are reserved for the resection
of visible lesions <2cm (EMR) and >2cm (ESD) in diameter.
The achievement of complete remission of dysplasia and IM
is very high (>80% of patients). Both procedures are general-
ly safe with low-rate of potential complications, such as
bleeding, strictures formation, and perforation [10, 106,
107]. Additionally, the findings of endoscopic resection di-
rectly influence the future management plan of the patient. If
histology of the resected specimen reveals no dysplasia, the
patient is selected just for endoscopic surveillance. If histol-
ogy revels LGD, HGD, or EAC, thorough specimen exami-
nation (invasion depth and margins) determined future resec-
tion, ablative, or surgical management as well as surveillance
[11].
4.3. Radiofrequency Ablation (RFA)
RFA is among the most frequently used ablation tech-
niques of BE [10, 15]. RFA aims to destroy damaged squa-
mous epithelium and to allow the growth of neosquamous
epithelium [16]. The EAC rate may significantly decrease by
eradicating BE metaplasia and dysplasia [17]. Currently,
RFA is established as an endoscopic technique for BE eradi-
cation as well as for the eradication of residual BE after en-
doscopic resection of early EAC [18]. RFA should not be the
appropriate first choice for the treatment of nodular BE. The
best approach is to treat visible or nodular BE by resection
technique followed by RFA [17, 19].
High-frequency energy is delivered to the mucosa in
depth 500-1000 micm to induce electromagnetic field with
oscillation and collision of charged ions with thermal energy
(approximately 300W) released for a short period (<300ms)
[106, 111]. When subsequent coagulation necrosis of epithe-
lial and muscularis mucosa cells is achieved [10, 106], treat-
ed mucosa will be removed under endoscopic visualization
with the possibility to repeat RFA delivery during one en-
doscopy session. Depending on the tissue response or the
length of BE, RFA can be repeated, usually in 2-3 months
periods [112]. Regarding achieve complete eradication of
BE, approximately 2-4 repeated RFA sessions are necessary
[106]. Depending on the mode of energy delivery and the
length and type of BE, RFA can be circumferential and focal
[106]. Circumferential RFA is reserved for the management
of long circumferential segment BE (>3cm), and focal RFA
for the management of short-segment BE (<3cm), BE islands
and tongues. Both modes of RFA administration need specif-
ic devices- HALO 360 for circumferential and HALO 90,
60, Ultra Long for focal RFA [11, 106, 112]. Currently, there
are two protocols for RFA use: standard (2x12-15J/cm2-
cleaning-2x12-15J/cm2) and simplified protocol (3x12J/cm2,
no cleaning) [113-115]. The effects are similar, but the sim-
plified protocol is less time consuming than standard and is
preferred regimen for focal RFA of BE [106, 116]. Almost
complete eradication of dysplasia and IM are encountered in
RFA treated patients with LGD, HGD, and “simple” BE
(90%, 81%, and 77% patients, respectively) [117, 118].
RFA is a safe procedure, with low rates of occurred com-
plications, such as bleeding, postprocedure chest pain, stric-
ture formation, and perforation [119, 120]. After RFA ses-
sion, management with PPIs and analgesics and no antimi-
crobials are recommended, with new endoscopy after 3
months [106]. In every fifth treated patient who achieved
complete eradication of IM, BE recurred after 2.4 years of
follow-up [121]. Some factors can influence incomplete or
poor response to RFA regarding dysplasia eradication, such
as longer length and duration of BE, poor reflux control,
abnormalities of p16 locus and p53 and RFA treatment per-
formed in low-frequency centres [109, 110, 122, 123]. IM
under the new squamous epithelium (“buried metaplasia”)
could be endoscopically invisible and carries a significant
risk of malignancy [124]. Post-RFA EAC is rare but indi-
cates endoscopic surveillance after RFA, even after the
treatment goal is achieved [106]. RFA for BE should be car-
ried out by trained and experienced endoscopists if it is pos-
sible in reference centre [16].
4.4. Surgical Treatment
Surgery is reserved for patients with persisted GERD
symptoms despite the long-term use of PPIs. Another indica-
tion of surgical treatment is a progressive disease. Current
laparoscopic techniques (partial or total fundoplication) are
the surgical procedures of choice [11, 20]. Former recom-
mendations regarding mandatory surgical treatment of EAC
are now changed- early EAC can be treated by endoscopic
eradication treatment, especially in early stages [10].
However, in the cases of multifocal HGD, selected patients
can be considered for surgery because of the higher risk of
occult EAC [125, 126].
CONCLUSION
BE represents a premalignant condition for the
development of EAC, so it is mandatory to break BE's
natural history chain by the use of medical, endoscopic, and
surgical strategies [10, 11]. Medical treatment is related to
PPIs in regard to control GERD symptoms and to decrease
the risk in the progression of BE to EAC via anti-acid and
anti-inflammatory effects, opposite to H2 blockers EAC [10,
63, 64]. Some concerns of PPIs adverse effects have been
addressed, such as an increase in gastrin levels and the risk
of Alzheimer's disease, but observational studies suggested
that the overall risk remains small even in long term use [99-
101]. Endoscopic treatment is recommended in those with
confirmed HGD, early EAC, and in selected LGD patients
[10, 103]. RFA is among the most frequently used ablation
techniques of BE [10, 15]. Currently, RFA has established an
endoscopic technique for BE eradication as well as for the
eradication of residual BE after endoscopic resection of early
EAC [18]. The imperfection of RFA as the procedure is
based on possible BE recurrence, the presence of "buried
metaplasia" as well as post-RFA-EAC. So, after RFA
session, management with PPIs is recommended, with new
endoscopy after 3 months and then at regular intervals,
depending on the purpose of endoscopic surveillance. The
conditions for successful RFA are its performance by trained
and experienced endoscopists in reference center and the
biopsy specimens histology examined by a pathologist
experienced in BE and EAC. Despite the well-recognized
8 Mini-Reviews in Medicinal Chemistry, 2020, Vol. 20, No. 0 Dugalic et al.
treatment use for BE and EAC, the best treatment for BE and
EAC needs to be further explored. Currently, the combined
use of chemoprevention (PPIs) and endoscopy treatment
(both tissue acquiring and ablative techniques) are the
mainstays of BE and EAC treatment, but in treatment
indolent and progressive disease surgical procedures are the
preferable choice.
CONSENT FOR PUBLICATION
Not applicable.
FUNDING
This work has been supported by grant No. 173033
(E.R.I.) from the Ministry of Science, Education and Tech-
nological Development, Republic of Serbia.
CONFLICT OF INTEREST
The authors declare no conflict of interest, financial or
otherwise.
ACKNOWLEDGEMENTS
Author’s contribution: P.D. and E.R.I. designed and
wrote the paper, S.Dj., A.P.M., V.D., R.T., Z.G., M.O., and
V.B. wrote the paper.
REFERENCES
[1] Barrett, N.R. Chronic peptic ulcer of the oesophagus and ‘oesopha-
gitis’. Br. J. Surg., 1950, 38(150), 175-182.
[http://dx.doi.org/10.1002/bjs.18003815005] [PMID: 14791960]
[2] El-Serag, H.B.; Sweet, S.; Winchester, C.C.; Dent, J. Update on the
epidemiology of gastro-oesophageal reflux disease: a systematic
review. Gut, 2014, 63(6), 871-880.
[http://dx.doi.org/10.1136/gutjnl-2012-304269] [PMID: 23853213]
[3] Taylor, J.B.; Rubenstein, J.H. Meta-analyses of the effect of symp-
toms of gastroesophageal reflux on the risk of Barrett's esophagus.,
Am J Gastroenterol., 2010, 105(8), 1729-1730.
[4] Zagari, R.M.; Fuccio, L.; Wallander, M.A.; Johansson, S.; Fiocca,
R.; Casanova, S.; Farahmand, B.Y.; Winchester, C.C.; Roda, E.;
Bazzoli, F. Gastro-oesophageal reflux symptoms, oesophagitis and
Barrett’s oesophagus in the general population: the Loiano-
Monghidoro study. Gut, 2008, 57(10), 1354-1359.
[http://dx.doi.org/10.1136/gut.2007.145177] [PMID: 18424568]
[5] Ronkainen, J.; Aro, P.; Storskrubb, T.; Johansson, S.E.; Lind, T.;
Bolling-Sternevald, E.; Vieth, M.; Stolte, M.; Talley, N.J.; Agréus,
L. Prevalence of Barrett’s esophagus in the general population: an
endoscopic study. Gastroenterology, 2005, 129(6), 1825-1831.
[http://dx.doi.org/10.1053/j.gastro.2005.08.053] [PMID: 16344051]
[6] Whiteman, D.C.; Kendall, B.J. Barrett’s oesophagus: epidemiolo-
gy, diagnosis and clinical management. Med. J. Aust., 2016,
205(7), 317-324.
[http://dx.doi.org/10.5694/mja16.00796] [PMID: 27681974]
[7] Connor, M.J.; Weston, A.P.; Mayo, M.S.; Sharma, P. The preva-
lence of Barrett’s esophagus and erosive esophagitis in patients un-
dergoing upper endoscopy for dyspepsia in a VA population. Dig.
Dis. Sci., 2004, 49(6), 920-924.
[http://dx.doi.org/10.1023/B:DDAS.0000034549.55326.67]
[PMID: 15309878]
[8] Sharma, P.; Dent, J.; Armstrong, D.; Bergman, J.J.; Gossner, L.;
Hoshihara, Y.; Jankowski, J.A.; Junghard, O.; Lundell, L.; Tytgat,
G.N.; Vieth, M. The development and validation of an endoscopic
grading system for Barrett’s esophagus: the Prague C & M criteria.
Gastroenterology, 2006, 131(5), 1392-1399.
[http://dx.doi.org/10.1053/j.gastro.2006.08.032] [PMID: 17101315]
[9] Spechler, S.J.; Sharma, P.; Souza, R.F.; Inadomi, J.M.; Shaheen,
N.J. American Gastroenterological Association. American Gastro-
enterological Association technical review on the management of
Barrett’s esophagus. Gastroenterology, 2011, 140(3), e18-e52.
[http://dx.doi.org/10.1053/j.gastro.2011.01.031] [PMID: 21376939]
[10] Martinucci, I.; de Bortoli, N.; Russo, S.; Bertani, L.; Furnari, M.;
Mokrowiecka, A.; Malecka-Panas, E.; Savarino, V.; Savarino, E.;
Marchi, S. Barrett’s esophagus in 2016: From pathophysiology to
treatment. World J. Gastrointest. Pharmacol. Ther., 2016, 7(2),
190-206.
[http://dx.doi.org/10.4292/wjgpt.v7.i2.190] [PMID: 27158534]
[11] Shaheen, N.J.; Falk, G.W.; Iyer, P.G.; Gerson, L.B. American
College of Gastroenterology. ACG Clinical Guideline: Diagnosis
and Management of Barrett’s Esophagus. Am. J. Gastroenterol.,
2016, 111(1), 30-50.
[http://dx.doi.org/10.1038/ajg.2015.322] [PMID: 26526079]
[12] Brown, C.S.; Lapin, B.; Wang, C.; Goldstein, J.L.; Linn, J.G.;
Denham, W.; Haggerty, S.P.; Talamonti, M.S.; Howington, J.A.;
Carbray, J.; Ujiki, M.B. Reflux control is important in the man-
agement of Barrett’s Esophagus: results from a retrospective 1,830
patient cohort. Surg. Endosc., 2015, 29(12), 3528-3534.
[http://dx.doi.org/10.1007/s00464-015-4103-3] [PMID: 25676204]
[13] Hillman, L.C.; Chiragakis, L.; Shadbolt, B.; Kaye, G.L.; Clarke,
A.C. Proton-pump inhibitor therapy and the development of dys-
plasia in patients with Barrett’s oesophagus. Med. J. Aust., 2004,
180(8), 387-391.
[PMID: 15089728]
[14] Kastelein, F.; Spaander, M.C.; Steyerberg, E.W.; Biermann, K.;
Valkhoff, V.E.; Kuipers, E.J.; Bruno, M.J. Proton pump inhibitors
reduce the risk of neoplastic progression in patients with Barrett’s
esophagus. Clin. Gastroenterol. Hepatol., 2013, 11(4), 382-388.
[http://dx.doi.org/10.1016/j.cgh.2012.11.014] [PMID: 23200977]
[15] Corley, D.A.; Kerlikowske, K.; Verma, R.; Buffler, P. Protective
association of aspirin/NSAIDs and esophageal cancer: a systematic
review and meta-analysis. Gastroenterology, 2003, 124(1), 47-56.
[http://dx.doi.org/10.1053/gast.2003.50008] [PMID: 12512029]
[16] Epithelial radiofrequency ablation for Barrett's oesophagus. Inter-
ventional procedures guidance [IPG344]; National Institute for
Health and Clinical Excellence:, 2010.
[17] Sappati Biyyani, R.S.; Chak, A. Barrett’s esophagus: review of
diagnosis and treatment. Gastroenterol. Rep. (Oxf.), 2013, 1(1), 9-
18.
[http://dx.doi.org/10.1093/gastro/got015] [PMID: 24759662]
[18] Bennett, C.; Vakil, N.; Bergman, J.; Harrison, R.; Odze, R.; Vieth,
M.; Sanders, S.; Gay, L.; Pech, O.; Longcroft-Wheaton, G.;
Romero, Y.; Inadomi, J.; Tack, J.; Corley, D.A.; Manner, H.;
Green, S.; Al Dulaimi, D.; Ali, H.; Allum, B.; Anderson, M.; Cur-
tis, H.; Falk, G.; Fennerty, M.B.; Fullarton, G.; Krishnadath, K.;
Meltzer, S.J.; Armstrong, D.; Ganz, R.; Cengia, G.; Going, J.J.;
Goldblum, J.; Gordon, C.; Grabsch, H.; Haigh, C.; Hongo, M.;
Johnston, D.; Forbes-Young, R.; Kay, E.; Kaye, P.; Lerut, T.; Lov-
at, L.B.; Lundell, L.; Mairs, P.; Shimoda, T.; Spechler, S.; Sontag,
S.; Malfertheiner, P.; Murray, I.; Nanji, M.; Poller, D.; Ragunath,
K.; Regula, J.; Cestari, R.; Shepherd, N.; Singh, R.; Stein, H.J.;
Talley, N.J.; Galmiche, J.P.; Tham, T.C.; Watson, P.; Yerian, L.;
Rugge, M.; Rice, T.W.; Hart, J.; Gittens, S.; Hewin, D.;
Hochberger, J.; Kahrilas, P.; Preston, S.; Sampliner, R.; Sharma, P.;
Stuart, R.; Wang, K.; Waxman, I.; Abley, C.; Loft, D.; Penman, I.;
Shaheen, N.J.; Chak, A.; Davies, G.; Dunn, L.; Falck-Ytter, Y.;
Decaestecker, J.; Bhandari, P.; Ell, C.; Griffin, S.M.; Attwood, S.;
Barr, H.; Allen, J.; Ferguson, M.K.; Moayyedi, P.; Jankowski, J.A.
Consensus statements for management of Barrett’s dysplasia and
early-stage esophageal adenocarcinoma, based on a Delphi process.
Gastroenterology, 2012, 143(2), 336-346.
[http://dx.doi.org/10.1053/j.gastro.2012.04.032] [PMID: 22537613]
[19] Falk, G.W. Updated Guidelines for Diagnosing and Managing
Barrett Esophagus. Gastroenterol. Hepatol. (N. Y.), 2016, 12(7),
449-451.
[PMID: 27489529]
[20] Galmiche, J.P.; Hatlebakk, J.; Attwood, S.; Ell, C.; Fiocca, R.;
Eklund, S.; Långström, G.; Lind, T.; Lundell, L. LOTUS Trial Col-
laborators. Laparoscopic antireflux surgery vs esomeprazole treat-
ment for chronic GERD: the LOTUS randomized clinical trial.
JAMA, 2011, 305(19), 1969-1977.
[http://dx.doi.org/10.1001/jama.2011.626] [PMID: 21586712]
Treatment of Barrett’s Esophagus Mini-Reviews in Medicinal Chemistry, 2020, Vol. 20, No. 0 9
[21] Amadi, C.; Gatenby, P. Barrett’s oesophagus: Current controver-
sies. World J. Gastroenterol., 2017, 23(28), 5051-5067.
[http://dx.doi.org/10.3748/wjg.v23.i28.5051] [PMID: 28811703]
[22] Thrift, A.P.; Kramer, J.R.; Qureshi, Z.; Richardson, P.A.; El-Serag,
H.B. Age at onset of GERD symptoms predicts risk of Barrett’s
esophagus. Am. J. Gastroenterol., 2013, 108(6), 915-922.
[http://dx.doi.org/10.1038/ajg.2013.72] [PMID: 23567358]
[23] Chak, A.; Lee, T.; Kinnard, M.F.; Brock, W.; Faulx, A.; Willis, J.;
Cooper, G.S.; Sivak, M.V., Jr; Goddard, K.A. Familial aggregation
of Barrett’s oesophagus, oesophageal adenocarcinoma, and oe-
sophagogastric junctional adenocarcinoma in Caucasian adults.
Gut, 2002, 51(3), 323-328.
[http://dx.doi.org/10.1136/gut.51.3.323] [PMID: 12171951]
[24] van Blankenstein, M.; Looman, C.W.; Johnston, B.J.; Caygill, C.P.
Age and sex distribution of the prevalence of Barrett’s esophagus
found in a primary referral endoscopy center. Am. J. Gastroenter-
ol., 2005, 100(3), 568-576.
[http://dx.doi.org/10.1111/j.1572-0241.2005.40187.x]
[PMID: 15743353]
[25] Runge, T.M.; Abrams, J.A.; Shaheen, N.J. Epidemiology of Bar-
rett’s Esophagus and Esophageal Adenocarcinoma. Gastroenterol.
Clin. North Am., 2015, 44(2), 203-231.
[http://dx.doi.org/10.1016/j.gtc.2015.02.001] [PMID: 26021191]
[26] Corley, D.A.; Kubo, A.; Levin, T.R.; Block, G.; Habel, L.; Ru-
more, G.; Quesenberry, C.; Buffler, P. Race, ethnicity, sex and
temporal differences in Barrett’s oesophagus diagnosis: a large
community-based study, 1994-2006. Gut, 2009, 58(2), 182-188.
[http://dx.doi.org/10.1136/gut.2008.163360] [PMID: 18978173]
[27] Cook, M.B.; Wild, C.P.; Forman, D. A systematic review and me-
ta-analysis of the sex ratio for Barrett’s esophagus, erosive reflux
disease, and nonerosive reflux disease. Am. J. Epidemiol., 2005,
162(11), 1050-1061.
[http://dx.doi.org/10.1093/aje/kwi325] [PMID: 16221805]
[28] Andrici, J.; Cox, M.R.; Eslick, G.D. Cigarette smoking and the risk
of Barrett’s esophagus: a systematic review and meta-analysis. J.
Gastroenterol. Hepatol., 2013, 28(8), 1258-1273.
[http://dx.doi.org/10.1111/jgh.12230] [PMID: 23611750]
[29] Cook, M.B.; Shaheen, N.J.; Anderson, L.A.; Giffen, C.; Chow,
W.H.; Vaughan, T.L.; Whiteman, D.C.; Corley, D.A. Cigarette
smoking increases risk of Barrett’s esophagus: an analysis of the
Barrett’s and Esophageal Adenocarcinoma Consortium. Gastroen-
terology, 2012, 142(4), 744-753.
[http://dx.doi.org/10.1053/j.gastro.2011.12.049] [PMID: 22245667]
[30] Kendall, B.J.; Macdonald, G.A.; Hayward, N.K.; Prins, J.B.;
O’Brien, S.; Whiteman, D.C. Study of Digestive Health. The risk
of Barrett’s esophagus associated with abdominal obesity in males
and females. Int. J. Cancer, 2013, 132(9), 2192-2199.
[http://dx.doi.org/10.1002/ijc.27887] [PMID: 23034724]
[31] Thrift, A.P.; Shaheen, N.J.; Gammon, M.D.; Bernstein, L.; Reid,
B.J.; Onstad, L.; Risch, H.A.; Liu, G.; Bird, N.C.; Wu, A.H.; Cor-
ley, D.A.; Romero, Y.; Chanock, S.J.; Chow, W.H.; Casson, A.G.;
Levine, D.M.; Zhang, R.; Ek, W.E.; MacGregor, S.; Ye, W.; Har-
die, L.J.; Vaughan, T.L.; Whiteman, D.C. Obesity and risk of
esophageal adenocarcinoma and Barrett’s esophagus: a Mendelian
randomization study. J. Natl. Cancer Inst., 2014, 106(11)dju252
[http://dx.doi.org/10.1093/jnci/dju252] [PMID: 25269698]
[32] Leggett, C.L.; Nelsen, E.M.; Tian, J.; Schleck, C.B.; Zinsmeister,
A.R.; Dunagan, K.T.; Locke, G.R., III; Wang, K.K.; Talley, N.J.;
Iyer, P.G. Metabolic syndrome as a risk factor for Barrett esopha-
gus: a population-based case-control study. Mayo Clin. Proc.,
2013, 88(2), 157-165.
[http://dx.doi.org/10.1016/j.mayocp.2012.09.017]
[PMID: 23374619]
[33] Iyer, P.G.; Borah, B.J.; Heien, H.C.; Das, A.; Cooper, G.S.; Chak,
A. Association of Barrett’s esophagus with type II Diabetes Melli-
tus: results from a large population-based case-control study. Clin.
Gastroenterol. Hepatol., 2013, 11(9), 1108-1114.e5.
[http://dx.doi.org/10.1016/j.cgh.2013.03.024] [PMID: 23591277]
[34] Leggett, C.L.; Gorospe, E.C.; Calvin, A.D.; Harmsen, W.S.;
Zinsmeister, A.R.; Caples, S.; Somers, V.K.; Dunagan, K.; Lutzke,
L.; Wang, K.K.; Iyer, P.G. Obstructive sleep apnea is a risk factor
for Barrett’s esophagus. Clin. Gastroenterol. Hepatol., 2014, 12(4),
583-8.e1.
[http://dx.doi.org/10.1016/j.cgh.2013.08.043] [PMID: 24035775]
[35] Greer, K.B.; Thompson, C.L.; Brenner, L.; Bednarchik, B.; Daw-
son, D.; Willis, J.; Grady, W.M.; Falk, G.W.; Cooper, G.S.; Li, L.;
Chak, A. Association of insulin and insulin-like growth factors
with Barrett’s oesophagus. Gut, 2012, 61(5), 665-672.
[http://dx.doi.org/10.1136/gutjnl-2011-300641] [PMID: 21930730]
[36] Kendall, B.J.; Macdonald, G.A.; Hayward, N.K.; Prins, J.B.;
Brown, I.; Walker, N.; Pandeya, N.; Green, A.C.; Webb, P.M.;
Whiteman, D.C. Study of Digestive Health. Leptin and the risk of
Barrett’s oesophagus. Gut, 2008, 57(4), 448-454.
[http://dx.doi.org/10.1136/gut.2007.131243] [PMID: 18178609]
[37] Corley, D.A.; Kubo, A.; Levin, T.R.; Block, G.; Habel, L.; Zhao,
W.; Leighton, P.; Rumore, G.; Quesenberry, C.; Buffler, P.; Par-
sonnet, J. Helicobacter pylori infection and the risk of Barrett’s oe-
sophagus: a community-based study. Gut, 2008, 57(6), 727-733.
[http://dx.doi.org/10.1136/gut.2007.132068] [PMID: 17895354]
[38] Thrift, A.P.; Pandeya, N.; Smith, K.J.; Green, A.C.; Hayward,
N.K.; Webb, P.M.; Whiteman, D.C. Helicobacter pylori infection
and the risks of Barrett’s oesophagus: a population-based case-
control study. Int. J. Cancer, 2012, 130(10), 2407-2416.
[http://dx.doi.org/10.1002/ijc.26242] [PMID: 21681741]
[39] Francois, F.; Roper, J.; Goodman, A.J.; Pei, Z.; Ghumman, M.;
Mourad, M.; de Perez, A.Z.; Perez-Perez, G.I.; Tseng, C.H.; Blaser,
M.J. The association of gastric leptin with oesophageal inflamma-
tion and metaplasia. Gut, 2008, 57(1), 16-24.
[http://dx.doi.org/10.1136/gut.2007.131672] [PMID: 17761783]
[40] Nwokolo, C.U.; Freshwater, D.A.; O’Hare, P.; Randeva, H.S.
Plasma ghrelin following cure of Helicobacter pylori. Gut, 2003,
52(5), 637-640.
[http://dx.doi.org/10.1136/gut.52.5.637] [PMID: 12692045]
[41] Kuipers, E.J.; Spaander, M.C. Natural History of Barrett’s Esopha-
gus. Dig. Dis. Sci., 2018, 63(8), 1997-2004.
[http://dx.doi.org/10.1007/s10620-018-5161-x] [PMID: 29905908]
[42] Zou, D.; He, J.; Ma, X.; Chen, J.; Gong, Y.; Man, X.; Gao, L.;
Wang, R.; Zhao, Y.; Yan, X.; Liu, W.; Wernersson, B.; Johansson,
S.; Dent, J.; Sung, J.J.; Li, Z. Epidemiology of symptom-defined
gastroesophageal reflux disease and reflux esophagitis: the system-
atic investigation of gastrointestinal diseases in China (SILC).
Scand. J. Gastroenterol., 2011, 46(2), 133-141.
[http://dx.doi.org/10.3109/00365521.2010.521888]
[PMID: 20955088]
[43] Shiota, S.; Singh, S.; Anshasi, A.; El-Serag, H.B. Prevalence of
Barrett’s Esophagus in Asian Countries: A Systematic Review and
Meta-analysis. Clin. Gastroenterol. Hepatol., 2015, 13(11), 1907-
1918.
[http://dx.doi.org/10.1016/j.cgh.2015.07.050] [PMID: 26260107]
[44] Fock, K.M.; Ang, T.L. Global epidemiology of Barrett’s esopha-
gus. Expert Rev. Gastroenterol. Hepatol., 2011, 5(1), 123-130.
[http://dx.doi.org/10.1586/egh.10.82] [PMID: 21309677]
[45] McDonald, S.A.; Lavery, D.; Wright, N.A.; Jansen, M. Barrett
oesophagus: lessons on its origins from the lesion itself. Nat. Rev.
Gastroenterol. Hepatol., 2015, 12(1), 50-60.
[http://dx.doi.org/10.1038/nrgastro.2014.181] [PMID: 25365976]
[46] Wang, X.; Ouyang, H.; Yamamoto, Y.; Kumar, P.A.; Wei, T.S.;
Dagher, R.; Vincent, M.; Lu, X.; Bellizzi, A.M.; Ho, K.Y.; Crum,
C.P.; Xian, W.; McKeon, F. Residual embryonic cells as precursors
of a Barrett’s-like metaplasia. Cell, 2011, 145(7), 1023-1035.
[http://dx.doi.org/10.1016/j.cell.2011.05.026] [PMID: 21703447]
[47] Quante, M.; Bhagat, G.; Abrams, J.A.; Marache, F.; Good, P.; Lee,
M.D.; Lee, Y.; Friedman, R.; Asfaha, S.; Dubeykovskaya, Z.;
Mahmood, U.; Figueiredo, J.L.; Kitajewski, J.; Shawber, C.; Light-
dale, C.J.; Rustgi, A.K.; Wang, T.C. Bile acid and inflammation ac-
tivate gastric cardia stem cells in a mouse model of Barrett-like
metaplasia. Cancer Cell, 2012, 21(1), 36-51.
[http://dx.doi.org/10.1016/j.ccr.2011.12.004] [PMID: 22264787]
[48] Jiang, M.; Li, H.; Zhang, Y.; Yang, Y.; Lu, R.; Liu, K.; Lin, S.;
Lan, X.; Wang, H.; Wu, H.; Zhu, J.; Zhou, Z.; Xu, J.; Lee, D.K.;
Zhang, L.; Lee, Y.C.; Yuan, J.; Abrams, J.A.; Wang, T.C.; Sepul-
veda, A.R.; Wu, Q.; Chen, H.; Sun, X.; She, J.; Chen, X.; Que, J.
Transitional basal cells at the squamous-columnar junction gener-
ate Barrett’s oesophagus. Nature, 2017, 550(7677), 529-533.
[http://dx.doi.org/10.1038/nature24269] [PMID: 29019984]
[49] Inadomi, J.; Alastal, H.; Bonavina, L.; Gross, S.; Hunt, R.H.;
Mashimo, H.; di Pietro, M.; Rhee, H.; Shah, M.; Tolone, S.; Wang,
D.H.; Xie, S.H. Recent advances in Barrett’s esophagus. Ann. N. Y.
Acad. Sci., 2018, 1434(1), 227-238.
10 Mini-Reviews in Medicinal Chemistry, 2020, Vol. 20, No. 0 Dugalic et al.
[http://dx.doi.org/10.1111/nyas.13909] [PMID: 29974975]
[50] Yamamoto, Y.; Wang, X.; Bertrand, D.; Kern, F.; Zhang, T.; Dule-
ba, M.; Srivastava, S.; Khor, C.C.; Hu, Y.; Wilson, L.H.; Blaszyk,
H.; Rolshud, D.; Teh, M.; Liu, J.; Howitt, B.E.; Vincent, M.; Crum,
C.P.; Nagarajan, N.; Ho, K.Y.; McKeon, F.; Xian, W. Mutational
spectrum of Barrett’s stem cells suggests paths to initiation of a
precancerous lesion. Nat. Commun., 2016, 7, 10380.
[http://dx.doi.org/10.1038/ncomms10380] [PMID: 26783136]
[51] Souza, R.F.; Huo, X.; Mittal, V.; Schuler, C.M.; Carmack, S.W.;
Zhang, H.Y.; Zhang, X.; Yu, C.; Hormi-Carver, K.; Genta, R.M.;
Spechler, S.J. Gastroesophageal reflux might cause esophagitis
through a cytokine-mediated mechanism rather than caustic acid in-
jury. Gastroenterology, 2009, 137(5), 1776-1784.
[http://dx.doi.org/10.1053/j.gastro.2009.07.055] [PMID: 19660463]
[52] Dunbar, K.B.; Agoston, A.T.; Odze, R.D.; Huo, X.; Pham, T.H.;
Cipher, D.J.; Castell, D.O.; Genta, R.M.; Souza, R.F.; Spechler,
S.J. Association of Acute Gastroesophageal Reflux Disease With
Esophageal Histologic Changes. JAMA, 2016, 315(19), 2104-2112.
[http://dx.doi.org/10.1001/jama.2016.5657] [PMID: 27187303]
[53] Lavery, D.L.; Nicholson, A.M.; Poulsom, R. The stem cell organi-
sation, and the proliferative and gene expression profile of Bar-
rett's epithelium, replicates pyloric-type gastric glands; , 2014, 63,
pp. (12)1854-1863.
[http://dx.doi.org/10.1136/gutjnl-2013-306508]
[54] Ross-Innes, C.S.; Becq, J.; Warren, A.; Cheetham, R.K.; Northen,
H.; O’Donovan, M.; Malhotra, S.; di Pietro, M.; Ivakhno, S.; He,
M.; Weaver, J.M.J.; Lynch, A.G.; Kingsbury, Z.; Ross, M.;
Humphray, S.; Bentley, D.; Fitzgerald, R.C. Whole-genome se-
quencing provides new insights into the clonal architecture of Bar-
rett’s esophagus and esophageal adenocarcinoma. Nat. Genet.,
2015, 47(9), 1038-1046.
[http://dx.doi.org/10.1038/ng.3357] [PMID: 26192915]
[55] Timmer, M.R.; Sun, G.; Gorospe, E.C.; Leggett, C.L.; Lutzke, L.;
Krishnadath, K.K.; Wang, K.K. Predictive biomarkers for Barrett’s
esophagus: so near and yet so far. Dis. Esophagus, 2013, 26(6),
574-581.
[http://dx.doi.org/10.1111/dote.12015] [PMID: 23316980]
[56] Konda, V.J.A.; Souza, R.F. Biomarkers of Barrett’s Esophagus:
From the Laboratory to Clinical Practice. Dig. Dis. Sci., 2018,
63(8), 2070-2080.
[http://dx.doi.org/10.1007/s10620-018-5088-2] [PMID: 29713984]
[57] Whiteman, D.C.; Appleyard, M.; Bahin, F.F.; Bobryshev, Y.V.;
Bourke, M.J.; Brown, I.; Chung, A.; Clouston, A.; Dickins, E.;
Emery, J.; Eslick, G.D.; Gordon, L.G.; Grimpen, F.; Hebbard, G.;
Holliday, L.; Hourigan, L.F.; Kendall, B.J.; Lee, E.Y.; Levert-
Mignon, A.; Lord, R.V.; Lord, S.J.; Maule, D.; Moss, A.; Norton,
I.; Olver, I.; Pavey, D.; Raftopoulos, S.; Rajendra, S.; Schoeman,
M.; Singh, R.; Sitas, F.; Smithers, B.M.; Taylor, A.C.; Thomas,
M.L.; Thomson, I.; To, H.; von Dincklage, J.; Vuletich, C.; Wat-
son, D.I.; Yusoff, I.F. Australian clinical practice guidelines for the
diagnosis and management of Barrett’s esophagus and early esoph-
ageal adenocarcinoma. J. Gastroenterol. Hepatol., 2015, 30(5),
804-820.
[http://dx.doi.org/10.1111/jgh.12913] [PMID: 25612140]
[58] Spechler, S.J.; Sharma, P.; Souza, R.F.; Inadomi, J.M.; Shaheen,
N.J. American Gastroenterological Association. American Gastro-
enterological Association medical position statement on the man-
agement of Barrett’s esophagus. Gastroenterology, 2011, 140(3),
1084-1091.
[http://dx.doi.org/10.1053/j.gastro.2011.01.031] [PMID: 21376940]
[59] Harrison, R.; Perry, I.; Haddadin, W.; McDonald, S.; Bryan, R.;
Abrams, K.; Sampliner, R.; Talley, N.J.; Moayyedi, P.; Jankowski,
J.A. Detection of intestinal metaplasia in Barrett’s esophagus: an
observational comparator study suggests the need for a minimum
of eight biopsies. Am. J. Gastroenterol., 2007, 102(6), 1154-1161.
[http://dx.doi.org/10.1111/j.1572-0241.2007.01230.x]
[PMID: 17433019]
[60] Reid, B.J.; Haggitt, R.C.; Rubin, C.E.; Roth, G.; Surawicz, C.M.;
Van Belle, G.; Lewin, K.; Weinstein, W.M.; Antonioli, D.A.;
Goldman, H. Observer variation in the diagnosis of dysplasia in
Barrett’s esophagus. Hum. Pathol., 1988, 19(2), 166-178.
[http://dx.doi.org/10.1016/S0046-8177(88)80344-7]
[PMID: 3343032]
[61] Montgomery, E.; Arnold, C.A.; Lam-Himlin, D.; Salimian, K.;
Waters, K. Some observations on Barrett esophagus and associated
dysplasia. Ann. Diagn. Pathol., 2018, 37, 75-82.
[http://dx.doi.org/10.1016/j.anndiagpath.2018.09.013]
[PMID: 30312881]
[62] Kedika, R.R.; Souza, R.F.; Spechler, S.J. Potential anti-
inflammatory effects of proton pump inhibitors: a review and dis-
cussion of the clinical implications. Dig. Dis. Sci., 2009, 54(11),
2312-2317.
[http://dx.doi.org/10.1007/s10620-009-0951-9] [PMID: 19714466]
[63] Bennett, C.; Moayyedi, P.; Corley, D.A.; DeCaestecker, J.; Falck-
Ytter, Y.; Falk, G.; Vakil, N.; Sanders, S.; Vieth, M.; Inadomi, J.;
Aldulaimi, D.; Ho, K.Y.; Odze, R.; Meltzer, S.J.; Quigley, E.; Git-
tens, S.; Watson, P.; Zaninotto, G.; Iyer, P.G.; Alexandre, L.; Ang,
Y.; Callaghan, J.; Harrison, R.; Singh, R.; Bhandari, P.; Bisschops,
R.; Geramizadeh, B.; Kaye, P.; Krishnadath, S.; Fennerty, M.B.;
Manner, H.; Nason, K.S.; Pech, O.; Konda, V.; Ragunath, K.;
Rahman, I.; Romero, Y.; Sampliner, R.; Siersema, P.D.; Tack, J.;
Tham, T.C.; Trudgill, N.; Weinberg, D.S.; Wang, J.; Wang, K.;
Wong, J.Y.; Attwood, S.; Malfertheiner, P.; MacDonald, D.; Barr,
H.; Ferguson, M.K.; Jankowski, J. BOB CAT Consortium. BOB
CAT: A Large-Scale Review and Delphi Consensus for Manage-
ment of Barrett’s Esophagus With No Dysplasia, Indefinite for, or
Low-Grade Dysplasia. Am. J. Gastroenterol., 2015, 110(5), 662-
682.
[http://dx.doi.org/10.1038/ajg.2015.55] [PMID: 25869390]
[64] Evans, J.A.; Early, D.S.; Fukami, N.; Ben-Menachem, T.; Chandra-
sekhara, V.; Chathadi, K.V.; Decker, G.A.; Fanelli, R.D.; Fisher,
D.A.; Foley, K.Q.; Hwang, J.H.; Jain, R.; Jue, T.L.; Khan, K.M.;
Lightdale, J.; Malpas, P.M.; Maple, J.T.; Pasha, S.F.; Saltzman,
J.R.; Sharaf, R.N.; Shergill, A.; Dominitz, J.A.; Cash, B.D. ASGE
Standards of Practice Committee; Standards of Practice Committee
of the American Society for Gastrointestinal Endoscopy. The role
of endoscopy in Barrett’s esophagus and other premalignant condi-
tions of the esophagus. Gastrointest. Endosc., 2012, 76(6), 1087-
1094.
[http://dx.doi.org/10.1016/j.gie.2012.08.004] [PMID: 23164510]
[65] Krishnamoorthi, R.; Singh, S.; Ragunathan, K.; Visrodia, K.;
Wang, K.K.; Katzka, D.A.; Iyer, P.G. Factors Associated With
Progression of Barrett’s Esophagus: A Systematic Review and Me-
ta-analysis. Clin. Gastroenterol. Hepatol., 2018, 16(7), 1046-
1055.e8.
[http://dx.doi.org/10.1016/j.cgh.2017.11.044] [PMID: 29199147]
[66] Nguyen, T.; Duan, Z.; Naik, A.D.; Kramer, J.R.; El-Serag, H.B.
Statin use reduces risk of esophageal adenocarcinoma in US veter-
ans with Barrett’s esophagus: a nested case-control study. Gastro-
enterology, 2015, 149(6), 1392-1398.
[http://dx.doi.org/10.1053/j.gastro.2015.07.009] [PMID: 26208896]
[67] Triadafilopoulos, G.; Friedland, S. Precision care for Barrett’s
esophagus. Transl. Gastroenterol. Hepatol., 2018, 3, 67.
[http://dx.doi.org/10.21037/tgh.2018.09.10] [PMID: 30363757]
[68] Das, D.; Chilton, A.P.; Jankowski, J.A. Chemoprevention of oe-
sophageal cancer and the AspECT trial. Recent Results Cancer
Res., 2009, 181, 161-169.
[http://dx.doi.org/10.1007/978-3-540-69297-3_15]
[PMID: 19213566]
[69] Nguyen, D.M.; Richardson, P.; El-Serag, H.B. Medications
(NSAIDs, statins, proton pump inhibitors) and the risk of esopha-
geal adenocarcinoma in patients with Barrett’s esophagus. Gastro-
enterology, 2010, 138(7), 2260-2266.
[http://dx.doi.org/10.1053/j.gastro.2010.02.045] [PMID: 20188100]
[70] Richter, J.E.; Penagini, R.; Tenca, A.; Pohl, D.; Dvorak, K.; Gold-
man, A.; Savarino, E.; Zentilin, P.; Savarino, V.; Watson, J.T.;
Wong, R.K.; Pace, F.; Casini, V.; Peura, D.A.; Herzig, S.J.; Kami-
ya, T.; Pelosini, I.; Scarpignato, C.; Armstrong, D.; DeVault, K.R.;
Bechi, P.; Taddei, A.; Freschi, G.; Ringressi, M.N.; De-
gli’Innocenti, D.R.; Castiglione, F.; Masini, E.; Hunt, R.H. Bar-
rett’s esophagus: proton pump inhibitors and chemoprevention II.
Ann. N. Y. Acad. Sci., 2011, 1232, 114-139.
[http://dx.doi.org/10.1111/j.1749-6632.2011.06048.x]
[PMID: 21950810]
[71] Fitzgerald, R.C.; di Pietro, M.; Ragunath, K.; Ang, Y.; Kang, J.Y.;
Watson, P.; Trudgill, N.; Patel, P.; Kaye, P.V.; Sanders, S.;
O’Donovan, M.; Bird-Lieberman, E.; Bhandari, P.; Jankowski,
J.A.; Attwood, S.; Parsons, S.L.; Loft, D.; Lagergren, J.; Moayyedi,
Treatment of Barrett’s Esophagus Mini-Reviews in Medicinal Chemistry, 2020, Vol. 20, No. 0 11
P.; Lyratzopoulos, G.; de Caestecker, J. British Society of Gastro-
enterology. British Society of Gastroenterology guidelines on the
diagnosis and management of Barrett’s oesophagus. Gut, 2014,
63(1), 7-42.
[http://dx.doi.org/10.1136/gutjnl-2013-305372] [PMID: 24165758]
[72] Gashi, Z.; Bahtiri, E.; Gashi, A.; Sherifi, F. Proton Pump Inhibitors
Diminish Barrett’s Esophagus Length: Our Experience. Open Ac-
cess Maced. J. Med. Sci., 2018, 6(6), 1041-1045.
[http://dx.doi.org/10.3889/oamjms.2018.232] [PMID: 29983798]
[73] Alsalahi, O.; Dobrian, A.D. Proton Pump Inhibitors: The Culprit
for Barrett’s Esophagus? Front. Oncol., 2015, 4, 373.
[http://dx.doi.org/10.3389/fonc.2014.00373] [PMID: 25621278]
[74] Strand, D.S.; Kim, D.; Peura, D.A. 25 Years of Proton Pump Inhib-
itors: A Comprehensive Review. Gut Liver, 2017, 11(1), 27-37.
[http://dx.doi.org/10.5009/gnl15502] [PMID: 27840364]
[75] Shirasaka, Y.; Sager, J.E.; Lutz, J.D.; Davis, C.; Isoherranen, N.
Inhibition of CYP2C19 and CYP3A4 by omeprazole metabolites
and their contribution to drug-drug interactions. Drug Metab. Dis-
pos., 2013, 41(7), 1414-1424.
[http://dx.doi.org/10.1124/dmd.113.051722] [PMID: 23620487]
[76] Sachs, G.; Shin, J.M.; Howden, C.W. Review article: the clinical
pharmacology of proton pump inhibitors. Aliment. Pharmacol.
Ther., 2006, 23(Suppl. 2), 2-8.
[http://dx.doi.org/10.1111/j.1365-2036.2006.02943.x]
[PMID: 16700898]
[77] Marelli, S.; Pace, F. Rabeprazole for the treatment of acid-related
disorders. Expert Rev. Gastroenterol. Hepatol., 2012, 6(4), 423-
435.
[http://dx.doi.org/10.1586/egh.12.18] [PMID: 22928894]
[78] Fass, R.; Inadomi, J.; Han, C.; Mody, R.; O’Neil, J.; Perez, M.C.
Maintenance of heartburn relief after step-down from twice-daily
proton pump inhibitor to once-daily dexlansoprazole modified re-
lease. Clin. Gastroenterol. Hepatol., 2012, 10(3), 247-253.
[http://dx.doi.org/10.1016/j.cgh.2011.11.021] [PMID: 22155561]
[79] Röhss, K.; Hasselgren, G.; Hedenström, H. Effect of esomeprazole
40 mg vs omeprazole 40 mg on 24-hour intragastric pH in patients
with symptoms of gastroesophageal reflux disease. Dig. Dis. Sci.,
2002, 47(5), 954-958.
[http://dx.doi.org/10.1023/A:1015009300955] [PMID: 12018920]
[80] Shin, J.M.; Sachs, G. Long lasting inhibitors of the gastric H,K-
ATPase. Expert Rev. Clin. Pharmacol., 2009, 2(5), 461-468.
[http://dx.doi.org/10.1586/ecp.09.33] [PMID: 21132072]
[81] Inadomi, J.M.; Jamal, R.; Murata, G.H.; Hoffman, R.M.; Lavezo,
L.A.; Vigil, J.M.; Swanson, K.M.; Sonnenberg, A. Step-down
management of gastroesophageal reflux disease. Gastroenterology,
2001, 121(5), 1095-1100.
[http://dx.doi.org/10.1053/gast.2001.28649] [PMID: 11677201]
[82] Mainie, I.; Tutuian, R.; Shay, S.; Vela, M.; Zhang, X.; Sifrim, D.;
Castell, D.O. Acid and non-acid reflux in patients with persistent
symptoms despite acid suppressive therapy: a multicentre study us-
ing combined ambulatory impedance-pH monitoring. Gut, 2006,
55(10), 1398-1402.
[http://dx.doi.org/10.1136/gut.2005.087668] [PMID: 16556669]
[83] Meko, J.B.; Norton, J.A. Management of patients with Zollinger-
Ellison syndrome. Annu. Rev. Med., 1995, 46, 395-411.
[http://dx.doi.org/10.1146/annurev.med.46.1.395]
[PMID: 7598474]
[84] Sjöstedt, S.; Befrits, R.; Sylvan, A.; Harthon, C.; Jörgensen, L.;
Carling, L.; Modin, S.; Stubberöd, A.; Toth, E.; Lind, T. Daily
treatment with esomeprazole is superior to that taken on-demand
for maintenance of healed erosive oesophagitis. Aliment. Pharma-
col. Ther., 2005, 22(3), 183-191.
[http://dx.doi.org/10.1111/j.1365-2036.2005.02553.x]
[PMID: 16091055]
[85] Chiba, T.; Malfertheiner, P.; Satoh, H. Proton Pump Inhibitors: A
Balanced View., Frontiers of Gastrointestinal Research; Frontiers
of Gastrointestinal Research: Karger, 2013.
[http://dx.doi.org/10.1159/isbn.978-3-318-02416-6]
[86] Sachs, G.; Shin, J.M.; Vagin, O.; Lambrecht, N.; Yakubov, I.;
Munson, K. The gastric H,K ATPase as a drug target: past, present,
and future. J. Clin. Gastroenterol., 2007, 41(Suppl. 2), S226-S242.
[http://dx.doi.org/10.1097/MCG.0b013e31803233b7]
[PMID: 17575528]
[87] Dunbar, L.A.; Caplan, M.J. Ion pumps in polarized cells: sorting
and regulation of the Na+, K+- and H+, K+-ATPases. J. Biol.
Chem., 2001, 276(32), 29617-29620.
[http://dx.doi.org/10.1074/jbc.R100023200] [PMID: 11404365]
[88] Poulsen, H.; Morth, P.; Egebjerg, J.; Nissen, P. Phosphorylation of
the Na+,K+-ATPase and the H+,K+-ATPase. FEBS Lett., 2010,
584(12), 2589-2595.
[http://dx.doi.org/10.1016/j.febslet.2010.04.035].
[PMID: 20412804]
[89] Sakai, H.; Fujii, T.; Takeguchi, N. Proton-Potassium (H(+)/K(+))
ATPases: Properties and Roles in Health and Diseases. Met. Ions
Life Sci., 2016, 16, 459-483.
[http://dx.doi.org/10.1007/978-3-319-21756-7_13]
[PMID: 26860309]
[90] Shin, J.M.; Munson, K.; Vagin, O.; Sachs, G. The gastric HK-
ATPase: structure, function, and inhibition. Pflugers Arch., 2009,
457(3), 609-622.
[http://dx.doi.org/10.1007/s00424-008-0495-4] [PMID: 18536934]
[91] Shi, S.; Klotz, U. Proton pump inhibitors: an update of their clinical
use and pharmacokinetics. Eur. J. Clin. Pharmacol., 2008, 64(10),
935-951.
[http://dx.doi.org/10.1007/s00228-008-0538-y] [PMID: 18679668]
[92] Kim, J.S.; Park, J.H.; Jeong, S.C.; Kim, D.S.; Yousaf, A.M.; Din,
F.U.; Kim, J.O.; Yong, C.S.; Youn, Y.S.; Oh, K.T.; Jin, S.G.; Choi,
H.G. Novel revaprazan-loaded gelatin microsphere with enhanced
drug solubility and oral bioavailability. J. Microencapsul., 2018,
35(5), 421-427.
[http://dx.doi.org/10.1080/02652048.2018.1515997]
[PMID: 30136606]
[93] Yu, K.S.; Bae, K.S.; Shon, J.H.; Cho, J.Y.; Yi, S.Y.; Chung, J.Y.;
Lim, H.S.; Jang, I.J.; Shin, S.G.; Song, K.S.; Moon, B.S. Pharma-
cokinetic and pharmacodynamic evaluation of a novel proton pump
inhibitor, YH1885, in healthy volunteers. J. Clin. Pharmacol.,
2004, 44(1), 73-82.
[http://dx.doi.org/10.1177/0091270003261321] [PMID: 14681344]
[94] Inatomi, N.; Matsukawa, J.; Sakurai, Y.; Otake, K. Potassium-
competitive acid blockers: Advanced therapeutic option for acid-
related diseases. Pharmacol. Ther., 2016, 168, 12-22.
[http://dx.doi.org/10.1016/j.pharmthera.2016.08.001]
[PMID: 27514776]
[95] Das, D.; Ishaq, S.; Harrison, R.; Kosuri, K.; Harper, E.; Decaes-
tecker, J.; Sampliner, R.; Attwood, S.; Barr, H.; Watson, P.; Mo-
ayyedi, P.; Jankowski, J. Management of Barrett’s esophagus in the
UK: overtreated and underbiopsied but improved by the introduc-
tion of a national randomized trial. Am. J. Gastroenterol., 2008,
103(5), 1079-1089.
[http://dx.doi.org/10.1111/j.1572-0241.2008.01790.x]
[PMID: 18445097]
[96] Trifirò, G.; Corrao, S.; Alacqua, M.; Moretti, S.; Tari, M.; Caputi,
A.P.; Arcoraci, V. UVEC Group. Interaction risk with proton pump
inhibitors in general practice: significant disagreement between dif-
ferent drug-related information sources. Br. J. Clin. Pharmacol.,
2006, 62(5), 582-590.
[http://dx.doi.org/10.1111/j.1365-2125.2006.02687.x]
[PMID: 16822281]
[97] Krieg, L.; Milstein, O.; Krebs, P.; Xia, Y.; Beutler, B.; Du, X.
Mutation of the gastric hydrogen-potassium ATPase alpha subunit
causes iron-deficiency anemia in mice. Blood, 2011, 118(24),
6418-6425.
[http://dx.doi.org/10.1182/blood-2011-04-350082]
[PMID: 21976678]
[98] Nehra, A.K.; Alexander, J.A.; Loftus, C.G.; Nehra, V. Proton Pump
Inhibitors: Review of Emerging Concerns. Mayo Clin. Proc., 2018,
93(2), 240-246.
[http://dx.doi.org/10.1016/j.mayocp.2017.10.022]
[PMID: 29406201]
[99] Fawaz, M.V.; Brooks, A.F.; Rodnick, M.E.; Carpenter, G.M.;
Shao, X.; Desmond, T.J.; Sherman, P.; Quesada, C.A.; Hockley,
B.G.; Kilbourn, M.R.; Albin, R.L.; Frey, K.A.; Scott, P.J. High af-
finity radiopharmaceuticals based upon lansoprazole for PET imag-
ing of aggregated tau in Alzheimer’s disease and progressive su-
pranuclear palsy: synthesis, preclinical evaluation, and lead selec-
tion. ACS Chem. Neurosci., 2014, 5(8), 718-730.
[http://dx.doi.org/10.1021/cn500103u] [PMID: 24896980]
12 Mini-Reviews in Medicinal Chemistry, 2020, Vol. 20, No. 0 Dugalic et al.
[100] Gomm, W.; von Holt, K.; Thomé, F.; Broich, K.; Maier, W.; Fink,
A.; Doblhammer, G.; Haenisch, B. Association of Proton Pump In-
hibitors With Risk of Dementia: A Pharmacoepidemiological
Claims Data Analysis. JAMA Neurol., 2016, 73(4), 410-416.
[http://dx.doi.org/10.1001/jamaneurol.2015.4791]
[PMID: 26882076]
[101] Rojo, L.E.; Alzate-Morales, J.; Saavedra, I.N.; Davies, P.; Maccio-
ni, R.B. Selective interaction of lansoprazole and astemizole with
tau polymers: potential new clinical use in diagnosis of Alz-
heimer’s disease. J. Alzheimers Dis., 2010, 19(2), 573-589.
[http://dx.doi.org/10.3233/JAD-2010-1262] [PMID: 20110603]
[102] Carmona, M.A.; Murai, K.K.; Wang, L.; Roberts, A.J.; Pasquale,
E.B. Glial ephrin-A3 regulates hippocampal dendritic spine mor-
phology and glutamate transport. Proc. Natl. Acad. Sci. USA, 2009,
106(30), 12524-12529.
[http://dx.doi.org/10.1073/pnas.0903328106] [PMID: 19592509]
[103] Ben Haim, L.; Carrillo-de Sauvage, M.A.; Ceyzériat, K.; Escartin,
C. Elusive roles for reactive astrocytes in neurodegenerative dis-
eases. Front. Cell. Neurosci., 2015, 9, 278.
[http://dx.doi.org/10.3389/fncel.2015.00278] [PMID: 26283915]
[104] Jankowski, J.A.Z.; de Caestecker, J.; Love, S.B.; Reilly, G.; Wat-
son, P.; Sanders, S.; Ang, Y.; Morris, D.; Bhandari, P.; Brooks, C.;
Attwood, S.; Harrison, R.; Barr, H.; Moayyedi, P. AspECT Trial
Team. Esomeprazole and aspirin in Barrett’s oesophagus (As-
pECT): a randomised factorial trial. Lancet, 2018, 392(10145),
400-408.
[http://dx.doi.org/10.1016/S0140-6736(18)31388-6]
[PMID: 30057104]
[105] Harrison, M.; Allen, J.E.; Gorrepati, V.S.; López-Jamar, J.M.E.;
Sharma, P. Management of Barrett’s esophagus with low-grade
dysplasia. Dis. Esophagus, 2018, 31(4)
[http://dx.doi.org/10.1093/dote/doy004] [PMID: 29506235]
[106] Krajciova, J.; Vackova, Z.; Spicak, J.; Martinek, J. Radiofrequency
ablation for Barrett’s esophagus-related neoplasia. Minerva Chir.,
2018, 73(4), 366-377.
[PMID: 29806757]
[107] Blevins, C.H.; Iyer, P.G. Endoscopic therapy for Barrett’s oesoph-
agus. Best Pract. Res. Clin. Gastroenterol., 2015, 29(1), 167-177.
[http://dx.doi.org/10.1016/j.bpg.2014.11.007] [PMID: 25743464]
[108] Pech, O.; Vieth, M.; Schmitz, D.; Gossner, L.; May, A.; Seitz, G.;
Stolte, M.; Ell, C. Conclusions from the histological diagnosis of
low-grade intraepithelial neoplasia in Barrett’s oesophagus. Scand.
J. Gastroenterol., 2007, 42(6), 682-688.
[http://dx.doi.org/10.1080/00365520601075803]
[PMID: 17505989]
[109] Small, A.J.; Araujo, J.L.; Leggett, C.L.; Mendelson, A.H.;
Agarwalla, A.; Abrams, J.A.; Lightdale, C.J.; Wang, T.C.; Iyer,
P.G.; Wang, K.K.; Rustgi, A.K.; Ginsberg, G.G.; Forde, K.A.;
Gimotty, P.A.; Lewis, J.D.; Falk, G.W.; Bewtra, M. Radiofrequen-
cy Ablation Is Associated With Decreased Neoplastic Progression
in Patients With Barrett’s Esophagus and Confirmed Low-Grade
Dysplasia. Gastroenterology, 2015, 149(3), 567-76.e3.
[http://dx.doi.org/10.1053/j.gastro.2015.04.013] [PMID: 25917785]
[110] Weusten, B.; Bisschops, R.; Coron, E.; Dinis-Ribeiro, M.; Dumon-
ceau, J.M.; Esteban, J.M.; Hassan, C.; Pech, O.; Repici, A.; Berg-
man, J.; di Pietro, M. Endoscopic management of Barrett’s esopha-
gus: European Society of Gastrointestinal Endoscopy (ESGE) Posi-
tion Statement. Endoscopy, 2017, 49(2), 191-198.
[http://dx.doi.org/10.1055/s-0042-122140] [PMID: 28122386]
[111] Pouw, R.E.; Gondrie, J.J.; Rygiel, A.M.; Sondermeijer, C.M.; ten
Kate, F.J.; Odze, R.D.; Vieth, M.; Krishnadath, K.K.; Bergman, J.J.
Properties of the neosquamous epithelium after radiofrequency ab-
lation of Barrett’s esophagus containing neoplasia. Am. J. Gastro-
enterol., 2009, 104(6), 1366-1373.
[http://dx.doi.org/10.1038/ajg.2009.88] [PMID: 19491850]
[112] Davila, M.L.; Hofstetter, W.L. Endoscopic management of Bar-
rett’s esophagus with high-grade dysplasia and early-stage esopha-
geal adenocarcinoma. Thorac. Surg. Clin., 2013, 23(4), 479-489.
[http://dx.doi.org/10.1016/j.thorsurg.2013.07.010]
[PMID: 24199698]
[113] Künzli, H.T.; Schölvinck, D.W.; Phoa, K.N.; Schoon, E.J.; Hou-
ben, M.H.; Bergman, J.J.; Weusten, B.L. Simplified protocol for
focal radiofrequency ablation using the HALO90 device: short-
term efficacy and safety in patients with dysplastic Barrett’s esoph-
agus. Endoscopy, 2015, 47(7), 592-597.
[http://dx.doi.org/10.1055/s-0034-1391436] [PMID: 25675174]
[114] Pouw, R.E.; Wirths, K.; Eisendrath, P.; Sondermeijer, C.M.; Ten
Kate, F.J.; Fockens, P.; Devière, J.; Neuhaus, H.; Bergman, J.J. Ef-
ficacy of radiofrequency ablation combined with endoscopic resec-
tion for barrett’s esophagus with early neoplasia. Clin. Gastroen-
terol. Hepatol., 2010, 8(1), 23-29.
[http://dx.doi.org/10.1016/j.cgh.2009.07.003] [PMID: 19602454]
[115] van Vilsteren, F.G.; Phoa, K.N.; Alvarez Herrero, L.; Pouw, R.E.;
Sondermeijer, C.M.; Visser, M.; Ten Kate, F.J.; van Berge He-
negouwen, M.I.; Weusten, B.L.; Schoon, E.J.; Bergman, J.J. A
simplified regimen for focal radiofrequency ablation of Barrett’s
mucosa: a randomized multicenter trial comparing two ablation
regimens. Gastrointest. Endosc., 2013, 78(1), 30-38.
[http://dx.doi.org/10.1016/j.gie.2013.02.002] [PMID: 23528655]
[116] Pouw, R.E.; Künzli, H.T.; Bisschops, R.; Sondermeijer, C.M.;
Koch, A.D.; Didden, P.; Gotink, A.W.; Schoon, E.J.; Curvers,
W.L.; Bergman, J.J.G.H.M.; Weusten, B.L.A.M. Simplified versus
standard regimen for focal radiofrequency ablation of dysplastic
Barrett’s oesophagus: a multicentre randomised controlled trial.
Lancet Gastroenterol. Hepatol., 2018, 3(8), 566-574.
[http://dx.doi.org/10.1016/S2468-1253(18)30157-2]
[PMID: 29934224]
[117] Phoa, K.N.; van Vilsteren, F.G.; Weusten, B.L.; Bisschops, R.;
Schoon, E.J.; Ragunath, K.; Fullarton, G.; Di Pietro, M.; Ravi, N.;
Visser, M.; Offerhaus, G.J.; Seldenrijk, C.A.; Meijer, S.L.; ten
Kate, F.J.; Tijssen, J.G.; Bergman, J.J. Radiofrequency ablation vs
endoscopic surveillance for patients with Barrett esophagus and
low-grade dysplasia: a randomized clinical trial. JAMA, 2014,
311(12), 1209-1217.
[http://dx.doi.org/10.1001/jama.2014.2511] [PMID: 24668102]
[118] Shaheen, N.J.; Sharma, P.; Overholt, B.F.; Wolfsen, H.C.; Sam-
pliner, R.E.; Wang, K.K.; Galanko, J.A.; Bronner, M.P.; Goldblum,
J.R.; Bennett, A.E.; Jobe, B.A.; Eisen, G.M.; Fennerty, M.B.;
Hunter, J.G.; Fleischer, D.E.; Sharma, V.K.; Hawes, R.H.; Hoff-
man, B.J.; Rothstein, R.I.; Gordon, S.R.; Mashimo, H.; Chang,
K.J.; Muthusamy, V.R.; Edmundowicz, S.A.; Spechler, S.J.; Sid-
diqui, A.A.; Souza, R.F.; Infantolino, A.; Falk, G.W.; Kimmey,
M.B.; Madanick, R.D.; Chak, A.; Lightdale, C.J. Radiofrequency
ablation in Barrett’s esophagus with dysplasia. N. Engl. J. Med.,
2009, 360(22), 2277-2288.
[http://dx.doi.org/10.1056/NEJMoa0808145] [PMID: 19474425]
[119] Bulsiewicz, W.J.; Kim, H.P.; Dellon, E.S.; Cotton, C.C.; Pasricha,
S.; Madanick, R.D.; Spacek, M.B.; Bream, S.E.; Chen, X.; Orlan-
do, R.C.; Shaheen, N.J. Safety and efficacy of endoscopic mucosal
therapy with radiofrequency ablation for patients with neoplastic
Barrett’s esophagus. Clin. Gastroenterol. Hepatol., 2013, 11(6),
636-642.
[http://dx.doi.org/10.1016/j.cgh.2012.10.028] [PMID: 23103824]
[120] Kumar, S.; Iyer, P.G. Endoscopic eradication therapy for Barrett’s
esophagus: Adverse outcomes, patient values, and cost-
effectiveness. Tech. Gastrointest. Endosc., 2018, 20(2), 75-81.
[http://dx.doi.org/10.1016/j.tgie.2018.02.002]
[121] Pasricha, S.; Bulsiewicz, W.J.; Hathorn, K.E.; Komanduri, S.;
Muthusamy, V.R.; Rothstein, R.I.; Wolfsen, H.C.; Lightdale, C.J.;
Overholt, B.F.; Camara, D.S.; Dellon, E.S.; Lyday, W.D.; Ertan,
A.; Chmielewski, G.W.; Shaheen, N.J. Durability and predictors of
successful radiofrequency ablation for Barrett's esophagus., Clin
Gastroenterol Hepatol., 2014, 12(11), 1840-1847.
[http://dx.doi.org/10.1016/j.cgh.2014.04.034]
[122] Tan, M.C.; Kanthasamy, K.A.; Yeh, A.G.; Kil, D.; Pompeii, L.;
Yu, X.; El-Serag, H.B.; Thrift, A.P. Factors Associated With Re-
currence of Barrett’s Esophagus After Radiofrequency Ablation.
Clin. Gastroenterol. Hepatol., 2019, 17(1), 65-72.e5.
[http://dx.doi.org/10.1016/j.cgh.2018.05.042] [PMID: 29902646]
[123] Timmer, M.R.; Brankley, S.M.; Gorospe, E.C.; Sun, G.; Lutzke,
L.S.; Iyer, P.G.; Halling, K.C.; Krishnadath, K.K.; Wang, K.K.
Prediction of response to endoscopic therapy of Barrett’s dysplasia
by using genetic biomarkers. Gastrointest. Endosc., 2014, 80(6),
984-991.
[http://dx.doi.org/10.1016/j.gie.2014.06.023] [PMID: 25085335]
[124] Gupta, N.; Mathur, S.C.; Dumot, J.A.; Singh, V.; Gaddam, S.;
Wani, S.B.; Bansal, A.; Rastogi, A.; Goldblum, J.R.; Sharma, P.
Adequacy of esophageal squamous mucosa specimens obtained
during endoscopy: are standard biopsies sufficient for postablation
Treatment of Barrett’s Esophagus Mini-Reviews in Medicinal Chemistry, 2020, Vol. 20, No. 0 13
surveillance in Barrett’s esophagus? Gastrointest. Endosc., 2012,
75(1), 11-18.
[http://dx.doi.org/10.1016/j.gie.2011.06.040] [PMID: 21907985]
[125] Dunbar, K.B.; Spechler, S.J. The risk of lymph-node metastases in
patients with high-grade dysplasia or intramucosal carcinoma in
Barrett’s esophagus: a systematic review. Am. J. Gastroenterol.,
2012, 107(6), 850-862.
[http://dx.doi.org/10.1038/ajg.2012.78] [PMID: 22488081]
[126] van Sandick, J.W.; van Lanschot, J.J.; ten Kate, F.J.; Offerhaus,
G.J.; Fockens, P.; Tytgat, G.N.; Obertop, H. Pathology of early in-
vasive adenocarcinoma of the esophagus or esophagogastric junc-
tion: implications for therapeutic decision making. Cancer, 2000,
88(11), 2429-2437.
[http://dx.doi.org/10.1002/1097-0142(20000601)88:11<2429::AID-
CNCR1>3.0.CO;2-H] [PMID: 10861416]
DISCLAIMER: The above article has been published in Epub (ahead of print) on the basis of the materials provided by the author. The Edito-
rial Department reserves the right to make minor modifications for further improvement of the manuscript.
... Although surgery remains the mainstay for solid pancreatic neoplasms, EUS-guided RFA offers a safe and effective treatment option in patients deemed unfit for, or who do not wish to undergo, surgery. [1][2][3][4] Here, we present a case of an 82-year-old woman with osteoarthritis who presented with progressively worsening upperabdominal pain over the past month and accompanying weight loss. CT of the abdomen and pelvis at an outside hospital revealed a 2.6-cm hypoattenuating mass in the pancreatic head, with pancreatic ductal dilation and atrophic changes in the body and tail (Fig. 1). ...
Preprint
Full-text available
Low energy/frequency radiofrequency (LRF) combined with the transdermal delivery of liposome (L) encapsulated antiaging peptides technology is a remarkable, newly developed physical noninvasive transdermal penetration technique; it is considered a highly efficient, comprehensive and safe technology. In this study, our objective was to evaluate the physical and chemical mechanisms underlying the efficacy of this innovative technique involving a combination of LRF and L, termed LLRF, that exerts a synergistic anti-aging effect on human skin, via an animal experiment. Physical and chemical analyses indicated that a relatively stable liposome with a uniform nano-size, which was formed, possessed good transdermal permeability that was 2.74 folds higher than that of the free peptide (F). LLRF exhibited a higher transdermal permeation performance that was of 3.65 folds higher than that of the free one, which was substantiated via confocal laser scanning fluorescence microscopy. The mouse UVB photoaging model trial confirmed that the LLRF technology exerted a significant synergistic effect compared to liposome technology, or free peptide, by downregulating inflammatory factors (IL-6, TNF-α), inhibiting the mRNA and protein expression of matrix metalloproteinases (MMP1, MMP3), promoting the mRNA and protein expression of related collagens (Procollagen, Col1α1 and Col3α1), and repairing the stratum corneum barrier function, as evidenced by trans-epidermal water loss (TEWL), skin cuticle hydration (SCH), and decreased expression of β-gal, an aging marker. These findings indicated that photoaging skin can be effectively and comprehensively rejuvenated, and that even photodamage can be reversed, thereby restoring the original physiological characteristics of healthy skin. Clinical tests have confirmed that although liposome technology is an effective antiaging method which helps exert tightening and anti-wrinkle effects on human skin, LLRF is an even more effective anti-aging technique. This study reveals a highly effective technique involving a combination physical and chemical therapy that may be utilized for antiaging purposes as well as repairing lightly damaged skin, and can be made readily available in the future.
Article
Full-text available
Background: Oesophageal adenocarcinoma is the sixth most common cause of cancer death worldwide and Barrett's oesophagus is the biggest risk factor. We aimed to evaluate the efficacy of high-dose esomeprazole proton-pump inhibitor (PPI) and aspirin for improving outcomes in patients with Barrett's oesophagus. Methods: The Aspirin and Esomeprazole Chemoprevention in Barrett's metaplasia Trial had a 2 × 2 factorial design and was done at 84 centres in the UK and one in Canada. Patients with Barrett's oesophagus of 1 cm or more were randomised 1:1:1:1 using a computer-generated schedule held in a central trials unit to receive high-dose (40 mg twice-daily) or low-dose (20 mg once-daily) PPI, with or without aspirin (300 mg per day in the UK, 325 mg per day in Canada) for at least 8 years, in an unblinded manner. Reporting pathologists were masked to treatment allocation. The primary composite endpoint was time to all-cause mortality, oesophageal adenocarcinoma, or high-grade dysplasia, which was analysed with accelerated failure time modelling adjusted for minimisation factors (age, Barrett's oesophagus length, intestinal metaplasia) in all patients in the intention-to-treat population. This trial is registered with EudraCT, number 2004-003836-77. Findings: Between March 10, 2005, and March 1, 2009, 2557 patients were recruited. 705 patients were assigned to low-dose PPI and no aspirin, 704 to high-dose PPI and no aspirin, 571 to low-dose PPI and aspirin, and 577 to high-dose PPI and aspirin. Median follow-up and treatment duration was 8·9 years (IQR 8·2-9·8), and we collected 20 095 follow-up years and 99·9% of planned data. 313 primary events occurred. High-dose PPI (139 events in 1270 patients) was superior to low-dose PPI (174 events in 1265 patients; time ratio [TR] 1·27, 95% CI 1·01-1·58, p=0·038). Aspirin (127 events in 1138 patients) was not significantly better than no aspirin (154 events in 1142 patients; TR 1·24, 0·98-1·57, p=0·068). If patients using non-steroidal anti-inflammatory drugs were censored at the time of first use, aspirin was significantly better than no aspirin (TR 1·29, 1·01-1·66, p=0·043; n=2236). Combining high-dose PPI with aspirin had the strongest effect compared with low-dose PPI without aspirin (TR 1·59, 1·14-2·23, p=0·0068). The numbers needed to treat were 34 for PPI and 43 for aspirin. Only 28 (1%) participants reported study-treatment-related serious adverse events. Interpretation: High-dose PPI and aspirin chemoprevention therapy, especially in combination, significantly and safely improved outcomes in patients with Barrett's oesophagus. Funding: Cancer Research UK, AstraZeneca, Wellcome Trust, and Health Technology Assessment.
Article
Full-text available
AIM: Our main objectives were to evaluate the influence of two-year proton pump inhibitors (PPI) therapy in patients with Barrett's oesophagus on its length, in both types, short and long segment. METHODS: In this single-centre, prospective interventional controlled study were analysed data collected prospectively over two years from patients with Barrett's oesophagus diagnosed by endoscopy. Patients who received continuous proton pump inhibitors (PPI) for 2 years. At each patient visit symptoms were recorded, and at each endoscopy, the length of Barrett's oesophagus (BE) was measured. Biopsies were taken along the length of the oesophagus at intervals of 1 cm. In total, 50 patients with Barrett's oesophagus were included in the study: 10 of whom had long-segment Barrett's oesophagus, and 40 patients had short-segment Barrett's oesophagus. The mean number of endoscopies performed was 3 per patient. RESULTS: The length of Barrett's esophagus (BE) was influenced by PPI therapy: Circumferential extension in BE patients short-segment Barrett's esophagus (SSBE) (before treatment was 1.5 cm and after treatment was 0.8 cm Maximum proximal extension in SSBE group before treatment was 2.3 cm (SD ± 1.1 cm), and 1.1 cm (SD ± 0.9 cm), respectively. Squamous islands were detected in 25% of patients examined after 2 years on PPIs. CONCLUSIONS: PPIs achieve a reduction to the length of Barrett's oesophagus, in both types, and the development of squamous islands is commonly associated with their use.
Article
Full-text available
Barrett’s esophagus (BE) is a very common condition. We have obtained fairly profound knowledge of the natural history of this condition. This results from many cross-sectional and cohort studies, many describing patients undergoing long-term surveillance. Their consent to use their clinical data has improved our knowledge to the benefit of these same and other patients. The prevalence of BE increases with age both in men and in women. This increase starts at a younger age in men than in women. The incidence of high-grade dysplasia and cancer in BE depends on segment length, gender, and age. The latter two likely indicate the duration of the presence of BE in an individual patient. Other factors that influence the incidence of dysplasia and cancer are smoking behavior and use of certain medications such as PPIs, statins, and NSAIDs. Surveillance of BE and treatment of dysplasia can impact the incidence of and mortality due to esophageal adenocarcinoma. This is of major benefit to a subgroup of BE patients. The epidemiology and burden of disease ask for further efforts to develop targeted screening, surveillance, and intervention techniques in coming years.
Article
Biopsy samples from esophageal columnar metaplasia and dysplasia are commonly encountered in Western pathology practice and knowing a few pitfalls can save both pathologists and patients a great deal of anxiety. Herein we discuss criteria for Barrett esophagus, evaluation of dysplasia, and some pitfalls in reviewing endoscopic mucosal resections. Also included is a summary of suggested follow-up for patients with Barrett esophagus.
Article
Modern recognition and management of Barrett’s esophagus, a precursor to esophageal adenocarcinoma depends on diagnostic accuracy, risk assessment, technical expertise and consideration of many options to best tailor therapy for every patient. Concomitant management of acid reflux is essential, frequently with proton pump inhibitors. Ablation and resection favorably affect the evolution towards cancer. Using precision medicine tools, such as imaging, molecular diagnostics and analytics may lead to cost- and comparatively-effective therapies ultimately aiming at cancer prevention. Knowledge of the risk factors for Barrett’s esophagus and progression to dysplasia and cancer can help tailor a precision medicine approach with more aggressive screening and surveillance targeted at patients that are most likely to benefit. © Translational Gastroenterology and Hepatology. All rights reserved.
Article
To develop a novel revaprazan-loaded gelatin microsphere with enhanced solubility and oral bioavailability, numerous gelatin microspheres were prepared using a spray-drying technique. The impact of gelatin amount on drug solubility in the gelatin microspheres was investigated. The physicochemical properties of the selected gelatin microsphere, such as shape, particle size and crystallinity, were evaluated. Moreover, its dissolution and pharmacokinetics in rats were assessed in comparison with revaprazan powder. Amongst the gelatin microspheres tested, the gelatin microsphere consisting of revaprazan and gelatin (1:2, w/w), which gave about 150-fold increased solubility, had the most enhanced drug solubility. It provided a spherical shape, amorphous drug and reduced particle size. Furthermore, it gave a higher dissolution rate and plasma concentration than did revaprazan powder. Particularly, it gave about 2.3-fold improved oral bioavailability in comparison with revaprazan powder. Therefore, this novel gelatin microsphere system is recommended as an oral pharmaceutical product of poorly water-soluble revaprazan.
Article
Barrett's esophagus (BE) is the only known precursor of esophageal adenocarcinoma, one of the few cancers with increasing incidence in developed countries. The pathogenesis of BE is unclear with regard to either the cellular origin of this metaplastic epithelium or the manner in which malignant transformation occurs, although recent data indicate a possible junctional origin of stem cells for BE. Treatment of BE may be achieved using endoscopic eradication therapy; however, there is a lack of discriminatory tools to identify individuals at sufficient risk for cancer development in whom intervention is warranted. Reduction in gastroesophageal reflux of gastric contents including acid is mandatory to achieve remission from BE after endoscopic ablation, and can be achieved using medical or nonmedical interventions. Research topics of greatest interest include the mechanism of BE development and transformation to cancer, risk stratification methods to identify individuals who may benefit from ablation of BE, optimization of eradication therapy, and surveillance methods to ensure that remission is maintained after eradication is achieved. Barrett's esophagus (BE) is defined as the replacement of the normal squamous lining of the esophagus with columnar epithelium (intestinal metaplasia) and is the only known precursor for the development of esophageal adenocarcinoma (EAC). Our review stems from the discussion of topics in BE at the 14th OESO World Conference: Global perspectives in Esophageal disease, and examines emerging data about the pathogenesis, screening and surveillance strategies, and treatment options for individuals with BE.
Article
Background: For focal radiofrequency ablation of Barrett's oesophagus, a simplified regimen (3 × 15 J/cm2, without cleaning) has proven to be as effective as the standard regimen (2 × 15 J/cm2, followed by cleaning, followed by 2 × 15 J/cm2). However, this simplified regimen seemed to be associated with a higher stenosis rate. Therefore, we lowered the radiofrequency energy and hypothesised that this new simplified regimen would be as effective and safe as the standard regimen. Methods: This randomised non-inferiority trial included patients with dysplastic Barrett's oesophagus or residual Barrett's oesophagus after endoscopic resection or circumferential radiofrequency ablation, in five European tertiary referral centres. Patients were randomly assigned (1:1) to the new simplified regimen (3 × 12 J/cm2, without cleaning) or the standard regimen, with variable block sizes of four, six, and eight patients, stratified by participating hospital. Focal radiofrequency ablation was done every 3 months, up to a maximum of three treatments, until all Barrett's oesophagus was eradicated. The primary outcome was complete endoscopic and histological regression of dysplasia and intestinal metaplasia after two focal radiofrequency ablation treatments, assessed in the intention-to-treat population. Non-inferiority was assessed on the basis of the difference between groups in the median percentage of Barrett's oesophagus surface regression, with a non-inferiority margin of -15%. This study is registered with www.trialregister.nl, number NTR4994, and is completed. Findings: Between March 25, 2015, and July 25, 2016, 84 patients were randomly assigned to treatment: 44 to receive the simplified regimen and 40 to receive the standard regimen. One patient assigned to the simplified regimen and four assigned to the standard regimen were excluded because they weree found not to be eligible; therefore the final intention-to-treat population consisted of 43 patients in the simplified ablation group and 36 in the standard ablation group. Complete endoscopic and histological regression of dysplasia and intestinal metaplasia after two focal radiofrequency ablation treatments was achieved in 32 (74%, 95% CI 59-87) patients treated with the simplified protocol, versus 30 (83%, 95% CI 67-94) patients treated with the standard protocol (p=0·34). Median Barrett's oesophagus surface regression after two focal radiofrequency ablation sessions was 98% (IQR 95-100) in the simplified regimen group and 100% (97-100) in the standard regimen group. The difference between medians was 2% (95% CI -0·562 to 3·162); thus the simplified regimen was deemed non-inferior to the standard regimen. Stenoses requiring dilatation were observed in four (9%) of 43 patients in the simplified regimen group and four (11%) of 36 in the standard regimen group. Post-procedural bleeding requiring repeat endoscopy occurred in one (2%) patient in the simplified ablation group and three (8%) patients in the standard ablation group. One patient (2%) in the simplified treatment group died 36 days after the second radiofrequency ablation procedure, due to an unknown cause. Interpretation: Based on the results of this study, we conclude that the simplified regimen is the preferred regimen for focal radiofrequency ablation of Barrett's oesophagus. Funding: None.
Article
Background & aims: Radiofrequency ablation (RFA) is effective treatment for Barrett's esophagus (BE). However, some patients have recurrence after complete eradication of intestinal metaplasia (CEIM). We investigated the incidence of and factors associated with BE recurrence, with or without neoplasia, after RFA and CEIM using data from the national Veterans Affairs (VA) healthcare system. Methods: We performed a retrospective cohort study of Veterans with BE treated by RFA from 2005 through 2016 with follow-up endoscopy. Subjects were followed until BE recurrence, neoplasia, death until October 2016. CEIM, BE recurrence, and factors associated with recurrence were identified by review of medical records. We calculated incidence rates of BE recurrence, with and without neoplasia, after CEIM and identified predictors using Cox proportional hazards models. Results: We identified 430 Veterans with BE who were treated with RFA; of these 337 achieved CEIM (78.4%). Most were men (98.6%), White (83.7%), and 66.0% had baseline dysplasia. Of those with CEIM, 98 patients (29.1%) had recurrence of BE during a total 906.0 patient-years of follow-up (median 1.9 years) after CEIM (incidence, 10.8%/patient-year). Dysplasia developed in 20 patients (2.2%/patient-year) and cancer in 3 patients (0.3%/patient-year). Baseline dysplasia (hazard ratio [HR], 1.71; 95% CI, 1.03-2.84) and long-segment BE (HR, 1.61; 95% CI, 1.03-2.54) increased risk of BE recurrence whereas treatment at high-volume RFA facilities reduced risk of BE recurrence (for quartile 4 vs quartile 1: HR, 0.16; 95% CI, 0.05-0.60). Conclusion: In a nationwide VA system study of outcomes of RFA for BE, we associated baseline dysplasia, long-segment BE, and treatment at low-volume RFA centers with recurrence of BE after CEIM. The findings call for performing these procedures in high-volume centers.
Article
Barrett's esophagus (BE) is a premalignant condition associated with increased risk of developing esophageal adenocarcinoma. In the past, BE patients with high-grade intraepithelial neoplasia (IEN) or early adenocarcinoma (EAC) were indicated for esophagectomy. With the recent advance in endoscopy, endoscopic techniques have surpassed esophagectomy in the treatment of Barrett's esophagus related neoplasia (BORN) and minimized the treatment-related morbidity. Patients with IEN are candidates for endoscopic treatment - endoscopic mucosal resection (ER) of visible lesions and/or ablation therapy of flat Barrett's mucosa. ER combined with radiofrequency ablation (RFA) is now considered as a gold standard for treatment of patients with early Barrett's cancer. RFA is currently the most effective method of ablation used in the treatment of LGIN/HGIN without visible lesions and for ablation of residual Barrett's mucosa following ER/ESD of EAC or HGIN aiming to achieve complete eradication of Barrett's surface and thus, decreasing the risk of recurrent dysplasia or cancer. The rates of complete remission of neoplasia (CR-N) and metaplasia (CR-M) after completion of endoscopic treatment are 81-92.6% and 75- 88.2%, respectively. The aim of this article is to review the principles, techniques, indications, efficacy and safety of this ablative method and surveillance of patients after successful treatment with RFA.