You are on page 1of 14

REVIEWS

Proton-pump inhibitors: understanding


the complications and risks
Peter Malfertheiner, Arne Kandulski and Marino Venerito
Abstract | Proton-pump inhibitors (PPIs) are the most effective therapy for the full spectrum
of gastric-acid-related diseases. However, in the past decade, a steadily increasing list of
complications following long-term use of PPIs has been reported. Their potent acid-suppressive
action induces several structural and functional changes within the gastric mucosa, including
fundic gland polyps, enterochromaffin-like cell hyperplasia and hypergastrinaemia, which can
be exaggerated in the presence of Helicobacter pylori infection. As discussed in this Review, most
associations of PPIs with severe adverse events are not based on sufficient evidence because
of confounding factors and a lack of plausible mechanisms. Thus, a causal relationship remains
unproven in most associations, and further studies are needed. Awareness of PPI-associated risks
should not lead to anxiety in patients but rather should induce the physician to consider the
appropriate dosing and duration of PPI therapy, including long-term monitoring strategies
in selected groups of patients because of their individual comorbidities and risk factors.

Proton-pump inhibitors (PPIs) became available in of acid-inhibiting agents (that is, histamine H2 receptor


clinical practice 25 years ago and have since added a antagonists) ­battling for the sales market at the time.
new successful therapeutic option to the management In our opinion, PPI-related adverse effects are, to a minor
of gastric-­acid-related diseases. These agents are among extent, substance-­related, but most are due to the induc-
the most commonly used drugs in the world, and their tion of gastric hypo­chlorhydria. In this Review, we will
wide use is best exemplified by sales of PPIs — one address the most rele­vant adverse events claimed with
­single PPI, esomeprazole, was reported to have more PPI use, reported from patient cohorts and population-­
than US$5 ­billion in sales worldwide in 2015 (REFS 1,2). based and community-­based studies, and indicate how
PPI therapy addressed previously unmet needs, such as they might be r­ elevant to clinical practice (TABLE 1).
healing and preventing peptic ulcerations, and helped
optimize the management of GERD. The clinical use Stomach
of PPIs has been extended to all diseases in which PPIs and structural or functional abnormalities.
gastric acid has either a primary or contributory role PPIs taken over a prolonged period of time lead to
(BOX 1). Depending on the nature of the disease, PPIs substantial changes in the structure and function of
are prescribed for short-term (2–8 weeks) or long-term the stomach (BOX 2). The most consistent structural
(>8 weeks) periods, with either continuous, intermittent changes in the gastric mucosa are the development of
or on‑demand intake. With the increasing use of PPIs, parietal cell hyperplasia and hypertrophy 4,5. Parietal cell
a series of important therapeutic challenges, adverse protrusion and swelling are reported after 3 months
effects and complications have emerged. In the past on omeprazole6.
Department of
decade, concerns have increasingly been raised about Experienced pathologists, therefore, might identify a
Gastroenterology, the long-term use of PPIs. Many of the adverse events patient on long-term PPI therapy solely on the finding
Hepatology and Infectious and complications of long-term PPI use have led to of parietal cell morphology in the oxyntic mucosa, even
Diseases, Otto von Guericke controversial results and have greatly contributed to in the absence of any clinical information. Parietal cell
University Hospital, Leipziger
the insecurity of prescribing physicians and the anxiety hyperplasia is related to the trophic, cell-­proliferation-
Strasse 44, 39120
Magdeburg, Germany. of patients. When PPIs were brought to market, initial inducing effect of hypergastrinaemia, which is most
Correspondence to P.M. 
concerns were related to PPI toxicity. Claims such as the often moderately increased in patients on long-term
peter.malfertheiner@ induction of vasculitis and anterior optical neuropathy PPI therapy 7,8. Hypergastrinaemia is paralleled by a
med.ovgu.de due to omeprazole were abandoned because of lack of decrease in somatostatin levels consequent to the potent
doi:10.1038/nrgastro.2017.117 causality 3. Indeed, some of the concerns raised in regard PPI-induced acid suppression (hypochlorhydria) 9,
Published online 20 Sep 2017 to PPI safety were a result of the highly competitive field regu­lated by an acid-driven negative feedback system.

NATURE REVIEWS | GASTROENTEROLOGY & HEPATOLOGY ADVANCE ONLINE PUBLICATION | 1


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Key points acid-suppression therapy (PPIs and/or H2 receptor


antago­nists) have been reported17. However, careful
• Proton-pump inhibitors (PPIs) induce structural and functional changes in the gastric documentation and discussion of the underlying mech­
mucosa related to potent acid suppression, which are exaggerated during anism have been provided for only two of these cases
Helicobacter pylori infection; PPIs alone are unlikely to be related to gastric of gastric carcinoids on long-term PPIs18,19. In one of
and gastrointestinal malignancies
these two patients, the gastric carcinoid was endo­
• The list of adverse events associated with PPI intake is increasing; few of these scopically removed, whereas it regressed spontaneously
associations are plausible or proven to have a causal relationship
in the other patient 16. In both patients, ECL cell hyper­
• The risk of bacterial enteric infections with Clostridium difficile, Salmonella and plasia regressed after cessation of PPI therapy. A modest
Campylobacter is increased in patients on PPI therapy — this risk is low to modest
increase in levels of gastrin and chromogranin is induced
• PPI use can rarely cause acute kidney injury and other morbid conditions related by PPIs, starting from day 5, and the levels return to
to idiosyncratic effects
­normal within a few days after PPI w ­ ithdrawal20 (FIG. 2).
• Long-term PPI intake interferes with magnesium and calcium homeostasis in small The diagnosis of gastrinoma can be delayed or pre-
subsets of patients with chronic kidney disease on diuretic therapy; the prevalence
vented by PPIs. The assumption is that PPIs can lead
of bone fractures attributable to PPIs in older patients is low
to complete symptom control and, therefore, mask the
• The debate on whether PPIs increase the risk of coronary events in patients on
endocrine neoplasia21. However, long-term PPI use is
clopidogrel seems to be resolved; the FDA recommends avoiding omeprazole
the appropriate therapy for most patients with this endo-
in patients taking clopidogrel
crine tumour. An increased risk of gastric cancer with
PPI use is often discussed but has never been proven.
Claims have even been made, on the basis of experi­
An important clinical effect of parietal cell hyperplasia mental studies, that PPIs might exert several poten-
is acid rebound, which occurs with a sudden stop of PPI tial anti-carcinogenic actions, including the induction
use after 8 weeks of continuous intake10. This abrupt of apoptosis and inhibition of inflammatory signals
withdrawal of a PPI after prolonged intake can lead to and of angiogenic activities by blocking gastrin recep-
acid-related symptoms or their relapse, and therefore, tor binding 22. Nevertheless, the clinical suspicion of a
patients should be instructed on how to taper down carcino­genic effect of PPIs in humans has been fuelled by
PPI intake if PPIs were taken for a prolonged period of a population-based cohort study. The incidence of gas-
time. No guidelines are available for how to proceed with tric cancer was increased among PPI users with the most
tapering, but in individual case management, PPI paus- prescriptions and longest follow‑up23. Furthermore, the
ing can be started with a 1‑day intermission with gradual gastric cancer incidence was more frequent in those on
prolongation of the intervals, and an attempt to stop PPIs PPIs that had received Helicobacter pylori eradication,
can be made after 2–4 weeks. In a subset of patients, but information on whether H. pylori eradication had
PPIs can lead to the development of so‑called cystic been successful in these patients was missing. Whether
glands, defined as fundic gland polyps (FGPs) because preneoplastic or early neoplastic changes had already
of their polyp-like endoscopic appearance (FIG. 1). FGPs, existed in patients developing gastric cancer during
although detected in ~2% of the general population, PPI intake cannot be extrapolated from the report.
are 2–4 times more frequent in patients with long-term H. pylori as a major confounding factor, in addition
PPI use, and there is a positive correlation between the to others (such as dietary habits or family history of
development of FGPs and the length of PPI exposure11,12. gastric cancer), was not taken into account in three
A systematic review with meta-analysis concluded from other observational studies, and therefore, the estim­
a subanalysis that the use of PPIs for at least 12 months ated risk of PPI-associated and drug-related gastric
is necessary for FGP development 13.
According to current literature, PPI-associated
FGPs have not been shown to develop into gastric Box 1 | Clinical use of PPIs
cancer. Extremely rare sporadic FGPs, independent of
• Peptic ulcer disease
PPI use, presenting with a mutation in β‑catenin with
a genetically predetermined risk of malignant pro- • Zollinger–Ellison syndrome
gression have been identified14,15. The benign nature • Stress ulcer prophylaxis
of FGPs and the exceptional reports of endoscopically • Prevention of NSAID-induced or ASA-induced
non-­distinguishable FGPs at risk of malignancy do not gastroduodenopathy
justify including examination for FGPs in regular gastric • Helicobacter pylori eradication
biopsy protocols. • Upper gastrointestinal bleeding
Development of enterochromaffin-like (ECL) cell • GERD (symptomatic)
hyperplasia in the oxyntic mucosa is found in 10–20% • Oesophagitis, Barrett oesophagus and peptic stricture
of patients on long-term PPIs5.The underlying mech- • Uninvestigated dyspepsia and functional dyspepsia
anism is a direct effect of gastrin (hypergastrinaemia),
• Oesophageal eosinophilia and suspected eosinophilic
and the dynamics of ECL cell proliferation are reflected oesophagitis
by increased serum levels of chromogranin A16. ECL
• Exocrine pancreatic insufficiency*
cell hyperplasia is of small clinical relevance and rarely
progresses to linear or even micronodular hyper­plasia. *Combined with enzyme replacement. ASA, acetylsalicylic
acid; PPI, proton-pump inhibitor.
To date, 11 cases of carcinoid tumours in patients on

2 | ADVANCE ONLINE PUBLICATION www.nature.com/nrgastro


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Table 1 | Evidence levels for the association between PPIs and various complications
Complications associated Level of Key findings
with PPI use evidence*
Structural or functional 2a Increased risk of fundic gland polyps
abnormalities
2a Increase in parietal cell mass
2b Hypergastrinaemia, hyperchromograninaemia
1c Enterochromaffin-like cell hyperplasia
Effects on Helicobacter 1c Shift from antrum-predominant to corpus-predominant gastritis,
pylori gastritis exaggerated increase in gastrin
Gastrointestinal infection 2a Increased risk of Clostridium difficile infection (OR 1.5–1.8)
2a/b For other pathogenic bacteria, >2–4‑fold increased risk
Pneumonia 2a/2b/2c Conflicting data, all from retrospective studies
2a/2b In large retrospective RCT and cohort studies, increased risk not confirmed
AIN 2b Considerable (fivefold increased) risk of AIN, re‑exposure to be avoided
CKD 2b Low risk of development of CKD
Hypomagnesaemia 3b Consider PPIs as a cause of hypomagnesaemia in patients with CKD
on diuretic therapy
Hypocalcaemia 4 No population-based or controlled studies
Fractures and osteoporosis 2a • PPI dose and duration dependency in older patients (aged >65 years)
• Low risk in patients without any other risk factors
Myopathies 4 • Possibly idiosyncratic reaction
• The underlying mechanisms leading to rhabdomyolysis with PPI use
are unclear but are possibly related to interactions with concomitantly
administered drugs such as HMG-CoA reductase inhibitors (statins).
Reduced clopidogrel 1c No increase of risk in randomized clinical trials
activation with increased
cardiovascular risks 2a Slightly increased risk in retrospective cohort studies.

Dementia 3b Increased risk (OR 1.6) with high bias


*Interpretation: 1a, systematic review of a randomized controlled trial (RCT) of good methodological quality and with
homogeneity; 1b, individual RCT with narrow 95% CI; 1c, individual RCT with risk of bias; 2a, systematic review of cohort studies
(with homogeneity); 2b, individual cohort study (including low-quality RCT, for example, <80% follow‑up); 2c, non-controlled
cohort studies or ecological studies; 3a, systematic review of case–control studies (with homogeneity); 3b, individual case–control
study; 4, case series or poor-quality cohort or case–control studies. AIN, acute interstitial nephritis; CKD, chronic kidney disease;
HMG-CoA, 3‑hydroxy‑3‑methylglutaryl-coenzyme A; OR, odds ratio; PPI, proton-pump inhibitor.

cancer (relative risk (RR) 1.43, 95% CI 1.23–1.66) 13 reverse causality and can be explained by the assump-
should not, in our opinion, cause concern at this time. tion that patients most likely started PPIs shortly after,
Further investigations of this important relationship or at the time of, pancreatic cancer diagnosis.
should be conducted before drawing a final conclusion.
Ideally, study design should be prospective and include PPIs and H. pylori gastritis. The interaction between
a large cohort of patients newly starting on PPIs with an PPIs and H. pylori is complex and depends on the pheno­
anticipated requirement for long-term PPI therapy for type of gastritis. In patients requiring PPIs, H. pylori gas-
follow-up. Most importantly, patients should be free or tritis is mostly antrum predominant27–29. With long-term
cured from H. pylori gastritis and should have no pre- PPI intake, levels of inflammation and bacterial coloniza­
neoplastic gastric mucosal lesions and no familial his- tion in the antrum decline, whereas the inflammation in
tory of gastric cancer at inclusion; a control group with the corpus and fundus was found to increase5,30,31 (FIG. 3).
no PPI use should also be included. Patient follow‑up Several studies have demonstrated that use of PPIs in
will require a decade or more. There is no indication H. pylori-infected patients is associated with the develop­
of increased risk of structural abnormalities in the mid ment of corpus-predominant gastritis32–34. Over the long
and lower gastrointestinal tract, including colorectal term, this process can lead to an accelerated loss of o
­ xyntic
adenoma and colorectal cancer, in patients on long-term glands and development of atrophic gastritis, impaired
PPI therapy 24,25. A statistically significant positive associ­ acid output, colonization of the stomach by other bacteria
ation with pancreatic cancer was reported for active (mainly those from the oral flora) and gastric hormone
PPI users (short-term and long-term): in a cohort of dysregulation26–28. The European Maastricht V/Florence
4,113 cases with pancreatic cancer and 16,012 matched consensus report recommended that H. pylori infection
controls, the prevalence of PPI use was 53% and 26%, be cured in patients requiring long-term PPI therapy to
respectively 26. The increased mortality in short-term prevent the development of gastric mucosa abnormalities
users is addressed by the study authors as an example of and progression to atrophic gastritis35.

NATURE REVIEWS | GASTROENTEROLOGY & HEPATOLOGY ADVANCE ONLINE PUBLICATION | 3


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Gastrointestinal tract has been reported42, but another study did not confirm
Gastric and intestinal microbiota. The effect of PPIs on this association43. Nearly all studies on the relation­
the gastric microbiota is primarily related to H. pylori. ship of PPIs with SIBO were based on glucose-H2
Microorganisms other than H. pylori are usually not breath tests, and most of them reported no statistically
detectable on the gastric mucosa but have been cul- signifi­cant association, as reported in a meta-analysis44.
tured from gastric biopsy samples and aspirates H2-based breath tests are less accurate than duodenal
obtained from patients on PPIs36. The gastric micro­ aspirate sampling. A study based on duodenal aspir­
biota p
­ attern on PPIs is similar to that seen in patients ates demonstrated a significantly increased preva-
with hypochlor­hydria associated with severe atrophic lence of bacterial and fungal overgrowth in patients on
gastritis37. The microbial colonization in the small and PPIs (P <0.0063), and that PPI use was an independ-
large bowel is also subject to variations in gastric acidity. ent risk factor for this overgrowth45. A study with the
Gut microbiota composition is altered by PPI intake, same methodology conducted in 713 patients on PPIs
according to a series of studies. In a crossover trial of reported an association between SIBO and IBS, but this
12 healthy individ­uals off and on PPIs, taxa (that is association was not affected by PPI intake46.
Enterococcaceae and Streptococcaceae) associated The assessment of SIBO and its clinical relevance
with Clostridium difficile became prominent ­colonizers must be readdressed by studies that will make use
of the lower gut, whereas Clostridiales decreased in of next-generation sequencing and of the analysis of
abundance38. Microbial diversity was reported to be meta­bolic products of the microorganisms colonizing
lower in patients on PPIs than in those not taking PPIs, the small bowel. Symptomatic SIBO developing during
according to a large twin cohort study 39. In a study of a PPI intake is, according to all available evidence, best
large cohort from the Netherlands, the gut microbiota handled on an individual basis.
assemblage was predisposing to C. difficile infection40.
A decrease in Bacteroides and an increase in Firmicutes Enteric infections. A threefold increased risk of enteric
abundance and the detection of Holdemania filiformis infections on PPI use, including by Salmonella and
were reported in a small cohort of 32 patients on long- Campylobacter spp., has been summarized in sys­tematic
term PPI use compared with 22 healthy individuals41. reviews of mostly observational and retrospective stud-
The clinical importance of these findings needs to be ies (six studies, 11,280 patients), highlighting substan-
further explored. Changes in gut microbiota compo- tial heterogeneity 47. A case–control study reported a
sition because of long-term PPI use might predispose fourfold increase in Salmonella enterica subsp. enter-
patients to dysbiosis and enteric infections. ica Enteritidis infection and an eightfold increase in
Salmonella enterica subsp. enterica Typhimurium
Small intestinal bacterial overgrowth. An increase infection with recent, even short-term, use of PPIs48.
in the number (colony forming units >1 × 105/ml) of Campylobacter jejuni diarrhoea has been reported with
endogenous symbiotic bacterial flora normally coloniz- an odds ratio of 2.9–11.7 from four studies49. Although
ing the colon defines the diagnosis of small intestinal the increased risk of enteric infection has been estab-
bacterial overgrowth (SIBO). The increase in gastric pH lished, the magnitude is heterogeneous and is estimated
by PPIs is the postulated mechanism of SIBO. At high to be on the order of 2–3‑fold. In a prospective Dutch
pH, the amount of bacteria normally transiting the population-based cohort study with 14,926 individ­
stomach and colonizing the small bowel is probably uals aged >45 years and a 24‑year follow‑up, PPI intake
increased. An increased risk of SIBO with PPI intake was associated with an increased risk (OR 1.94, 95% CI
1.15–3.25) of bacterial gastroenteritis50. Strong acid
inhibition might also facilitate traveller’s diarrhoea
Box 2 | Stomach-related PPI complications induced by common enteropathogens (such as toxigenic
Escherichia coli or Shigella spp.)49, but properly designed
• Hypochlorhydria prospective studies are not available. In clinical practice,
• Hypergastrinaemia patients are advised to first observe hygienic recom­
• Parietal cell hyperplasia mendations, but, in addition, they should reduce PPI
• Fundic gland polyps (FGPs) intake to a minimum effective required dose when visit­
• Enterochromaffin-like (ECL) cell hyperplasia along with ing subtropical or tropical regions with well-known risk
hyperchromograninaemia of enteric infections.
• Gastric carcinoids (rare) The risk of developing C.  difficile infection is
Proton-pump inhibitor (PPI) intake results in various
increased by 1.5–2‑fold even after recent PPI use. Two
degrees of acid suppression depending on dose, duration meta-analyses based on 42 (30 case–control, 12 cohort)
and individual responses. This process leads to a series of and 51 (37 case–control, 14 cohort) studies showed that
functional and structural consequences. The initial the risk of developing C. difficile infection in patients on
response to impaired acid production is an increase of PPIs is modest (OR 1.74, 95% CI 1.47–2.85 and OR 1.65,
gastrin, which is intended as a compensating mechanism 95% CI 1.47–1.85, respectively)51,52. These meta-­analyses
to stimulate the oxyntic gland’s components to produce noted several shortcomings related to the nature of the
acid and results in cellular increase (parietal cells, ECL studies — mostly observational, monocentric and retro­
cells). FGPs are four times more frequently detected. spective, with no adjustment for comorbidity and no
Gastric carcinoids are rare.
report on the duration of PPI exposure or antibiotic

4 | ADVANCE ONLINE PUBLICATION www.nature.com/nrgastro


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

a b Extraintestinal complications
Pneumonia. In critically ill patients, stress ulcer prophy­
laxis with PPIs was reported to be associated with an
increased risk of pneumonia (OR 1.3–2.7)62. However,
the putative mechanism of ascending bacterial coloniza­
tion from the hypoacidic stomach with transfer to the
* lung because of high intragastric pH is unproven and
lacks plausibility, except in aspiration pneumonia.
In a systematic review and meta-analysis published
in 2016, pneumonia and mortality were not found
to be s­ ignificantly increased in critically ill patients
|
Figure 1 Endoscopic appearance of fundic Naturegland polyps.
Reviews Characteristic&endoscopic
| Gastroenterology Hepatology taking PPIs63.
images of multiple fundic gland polyps of the oxyntic mucosa. a | Multiple fundic gland
polyps (arrow). b | Histomorphological characteristics of a cystic gland dilatation After the first report of community-acquired pneu­
(asterisk). These polyps are located in the fundic or corpus mucosa between gastric rugal monia by use of PPIs in an outpatient setting 64, a meta-
folds. They can also appear in a single polypoid structure. The size varies from a few analysis of five observational studies corrobor­ated this
millimetres up to 10 mm. The underlying lesion is not a mucosal neoformation, but a early observation65. However, later studies did not con-
cystic gland dilatation (histomorphology). Part b courtesy of D. Jechorek, University of firm an increased risk of community-acquired pneu-
Magdeburg, Germany, in collaboration with P. M. monia in patients on PPIs. A study involving eight
Canadian centres with >4 million patients newly pre-
scribed NSAIDs found that those exposed to PPIs were
intake — and concluded that a cause–effect relation- not more frequently affected by pneumonia than those
ship cannot be supported based on these findings. not exposed to PPIs66. No association between treatment
A meta-analysis published in 2016 based on 23 obser- with a PPI (esomeprazole) and community-­acquired
vational studies including 186,033 individuals con- respiratory tract infections (including pneumonia)
firmed the modest magnitude of the PPI-related risk of was reported in a retrospective analysis from 24 ran­
hospital-acquired C. difficile infection but emphasized domized double-blind clinical ­trials of patients receiving
the notable health and economic burden53. Notably, the esomepra­zole67. An overview of 33 sys­tematic reviews
coadministration of a PPI with antibiotics has an addi- on PPI-related adverse events underlines the finding
tive increased risk, and therefore, prescription of PPIs that the strongest association of community-­acquired
should be checked for ­appropriateness to minimize the pneumonia was most strongly associated with PPI
risk of C. difficile infection54. intake in the first 7–30 days68. This finding can be taken
as a hint that PPIs are more likely prov­ided to patients at
Microscopic colitis. An association between PPIs and the onset of pneumonia rather than as an adjunct med­
microscopic colitis has been reported in a series of ication. These latest studies seem to largely outweigh the
case reports55,56. In a retrospective analysis of 95 well-­ claim that PPI use increases the risk of pneumonia.
documented cases of microscopic colitis, exposure to
PPIs was identified as a risk factor 57. The magnitude of PPIs and dementia. The risk of incidental dementia
the risk associated with PPIs was low, but PPI intake as from PPI therapy was reported in a large p ­ rospective
a rare cause of diarrhoea should be remembered. The observational longitudinal study from Germany
risk of microscopic colitis is statistically significant for (OR 1.44, 95% CI 1.36–1.52)69. Notably, at inclusion,
concomitant PPI and NSAID therapy 58, although the patients had no dementia and a minimum age of
underlying mechanisms are unknown. 75 years. In this study, PPI intake was found to have
a similar degree of association with cognitive impair-
Spontaneous bacterial peritonitis and the liver. The ment as with age, depression and polypharmacy. Even
association between the risk of spontaneous bacterial after exclusion of these factors, the association of regu­
peritonitis (SBP) and PPI use is controversial. In a sys- lar PPI intake with dementia in this elderly population
tematic review including 12 articles and five confer- remained statistically significant. However, in a large
ence abstracts, the odds ratio for SBP was 2.17 (95% CI case–control study of primary care in patients with
1.46–3.23)59. In a prospective multicentre study, there dementia (mean age of 80 years), PPIs were associ-
was no evidence of an increased incidence of SBP60. In a ated with a reduced risk of dementia (OR 0.93, 95% CI
further study of a post-hoc analysis of different thera- 0.90–0.97). A similar effect was shown with statins and
peutic ­trials in an international patient cohort, the odds antihypertensive drugs70.
ratio for SBP was 1.72 (95% CI 1.10–2.69), and the risk Data on the association of PPIs and dementia are
of hepatic encephalopathy was also increased61. In our controversial, with either a modest or even reduced risk.
opinion, the appropriate use of PPIs in patients with Confounding factors such as patient characteristics,
liver cirrhosis is of no concern and should follow the comorbidities and polypharmacy are the most likely
usual established indications for PPIs. Inappropriate explanation for this discrepancy. In our opinion, stud-
use includes long-term PPI therapy in patients with ies that have so far been conducted only in patients with
oesophageal varices in the absence of GERD, as there is dementia in advanced age should not cause any worries
no evidence for protection from oesophageal variceal concerning the use of PPIs but rather should remind us
bleeding with long-term PPI intake. about their responsible use for proper indications.

NATURE REVIEWS | GASTROENTEROLOGY & HEPATOLOGY ADVANCE ONLINE PUBLICATION | 5


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

and series have documented this association72. The


PPIs most reason­able pathophysiological mechanism of PPI-
induced AIN seems to be an idiosyncratic reaction73. In a
Parietal cell case series of 133 patients with biopsy-proven AIN, 12%
of the cases were PPI-induced74. Thus, the widespread
prescription of PPIs requires awareness of this possible
H+ PPI complication75. A population-based nested case–­
control study from New Zealand including 572,661
+ patients without any underlying kidney disease showed
a fivefold increased risk of AIN on PPI therapy, especially
↑ Gastrin in older patients (aged >60 years of age)76. In another
population-­based study from the USA, >290,000 patients
who took PPI medication within 120 days before hos-
Blood pital admission due to acute kidney injury (AKI) were
vessel
↑ Chromogranin matched to an equal number of sex-matched and age-
↓ H+ matched controls who did not receive PPI medica-
↑ ECL cell tion77. Propensity score matching was used to establish
↑ pH
a highly comparable reference group of control patients.
PPI intake was associated with an increased risk of AKI
Hypergastrinaemia (HR 2.52, 95% CI 2.27–2.79) and AIN (HR 3.0, 95% CI
↑ Gastrin 1.47–6.14), again with higher ­frequency in older patients.
Early detection of AIN prompts drug withdrawal with
+ + Parietal cell recovery of renal function. Progression of PPI-induced
hyperplasia AIN to chronic kidney disease (CKD) after an episode
↓ D cell
• 2-4-fold of AIN is possible78. In a prospective survey of a large
increased
risk of fundic cohort of patients with 10,482 participants, self-reported
↑ G cell gland polyps PPI intake was associated with an increased risk of
CKD development (HR 1.50, 95% CI 1.14–1.96), with a
Figure 2 | Effect of PPIs on gastric physiology. The pathway of acid impairment by
Nature Reviews | Gastroenterology & Hepatology ­number needed to harm of 30 patients. The risk of CKD
proton-pump inhibitors (PPIs) is illustrated (dotted lines indicate putative pathways).
Hypochlorhydria interferes with antral D and G cells, resulting in increased gastrin
was higher among patients taking PPIs twice daily than
and chromogranin that activates a hyperplastic reaction in enterochromaffin-like (ECL) those taking them once daily 79. In a study published in
cells and parietal cells. 2016 including 172,321 patients, the risk attributed to
PPI intake for developing AKI was moderately increased
(HR 2.15, 95% CI 2.00–2.32); the incidence of CKD was
Myopathies. According to a review of the WHO adverse not of clinical concern80.
drug reaction database (VigiBase), PPI use might, All studies on the relationship of PPI intake and
although rarely, be associated with the occurrence of myo- increased risk of CKD have been performed in large
pathies, including rhabdomyolysis71. In total, the search cohorts of patients with robust statistical methods, but
identified 292 reports of various myopathies in association with relevant limitations. A substantial bias exists owing
with PPIs. In one-third of the cases, the PPI was the only to patient selection, concomitant drugs and cardio­
administered drug, whereas in 57% of the reports, the PPI vascular comorbidity. The association of CKD with PPIs
was the only suspected drug by the reporter despite the (HR <2.0) is of low magnitude, but awareness of PPIs as
fact that concomitant medication was used. The strength a possible cause for renal disease is important. In patients
of the association between PPI intake and myopathies with new-onset AKI due to AIN, early withdrawal of
is based on the temporality, that is, that the occurrence PPIs is mandatory to restore kidney function. In patients
of myopathy is associated with the timing of when PPIs at increased risk of PPI-induced AIN (that is, patients
are taken. In a further report, the same patient took three >60 years), monitoring of renal function in the first few
different PPIs (lansoprazole, esomeprazole and rabepra- months might be worth considering.
zole) at different time periods, with myalgia and muscle
weakness occurring with all three drugs71. Furthermore, Death. In older patients (≥65 years) discharged from
the time to onset after PPI intake was provided in 17 of the acute care hospitals, the association between PPI intake
cases of patients with rhabdomyolysis. The complication and 1‑year mortality at low doses of PPIs was not statis­
occurred during the first week in nine cases and between tically significant, whereas high-dose PPI treatment was
14 days to 3 months after treatment in a further three associated with an increased 1‑year mortality (OR 2.59)81.
patients. Notably, in 12 of these patients, a concomi­tant PPIs in high doses are considered an indicator of high
intake of a 3‑hydroxy‑3‑­methylglutaryl-coenzyme A morbidity and polymorbidity with an associated higher
(HMG‑CoA) reductase inhibitor (­statin) was recorded. risk of mortality 81. A direct causality has not been
established. In a meta-analysis of 20 randomized clin-
Kidney disease. PPI intake has been associated with ical trials, there was no difference in all-cause mortal-
acute interstitial nephritis (AIN). Since the first report ity between patients receiving PPI therapy and those
of omeprazole-induced AIN in 1992, other case reports receiving placebo82.

6 | ADVANCE ONLINE PUBLICATION www.nature.com/nrgastro


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Vitamin and mineral deficiency databank with ~1% of the patients taking long-term
Vitamin B12 deficiency. In a case–control study within PPI med­ication. The issue here is possible confound-
the Kaiser Permanente Northern California popula- ing factors (that is, impaired renal function or the use
tion, a ≥2‑year supply of PPIs was dispensed to 12% of diuretics)90. The homeostasis of the serum magne­
of patients with vitamin B12 deficiency compared with sium level is balanced by intestinal absorption and
7.2% of patients in the control group without vitamin B12 renal excretion91. The postulated patho­physiological
deficiency 83. A ≥2‑year supply of PPIs (OR 1.65, 95% CI background of PPI-associated hypomagnesaemia is
1.58–1.73) was associated with an increased risk of vita- apparently caused by reduced intestinal magnesium
min B12 deficiency. There is a plausible mech­anism for absorption by inhib­ition of the TRPM6 and TRPM7
some patients with an inadequate nutritional supply: channel-dependent paracellular magnesium transpor-
gastric acid is important for the release of vitamin B12 tation92. In patients with chronic renal insufficiency and
from ingested nutrients84. Gastric parietal cells are also receiving haemo­dialysis, renal excretion is addition-
the source of intrinsic factor, which is essential for B12 ally disturbed, and magnesium homeostasis is mainly
binding 85; therefore, high PPI doses might contribute to regu­lated by intestinal reabsorption93. Diuretics repre-
the critical reduction of intrinsic factor. Determination sent independent risk factors for hypomagnesaemia,
of serum vitamin B12 levels (including peripheral red as they increase, per se, the renal loss of magnesium
blood cell analysis) at biannual intervals seems appro- and other electrolytes. Large cross-­sectional trials sug-
priate in long-term PPI users, especially if they are on gest that PPI-associated hypo­magnesaemia can occur,
high-dose PPIs and have dietary restrictions (such as ­especially in patients with chronic renal insufficiency
strict vegetarians and vegans). on diuretic therapy 94–97.
In patients without chronic renal insufficiency and
Hypomagnesaemia. Clinical manifestations of hypo- without concomitant diuretic therapy, the associ­ation
magnesaemia include neuromuscular disturbances of PPIs and hypomagnesaemia has not been proven98.
(for example, tetany and seizures) and cardiac arrhyth- In line with these results, a case–control study includ-
mias86. Hypomagnesaemia with depletion of total body ing 154 outpatients on long-term PPI therapy and
magnesium stores in patients with long-term PPI use 84 matched controls not using PPIs showed no differ­
deserves consideration, because if it occurs, it is of ences in serum magnesium levels99. Thus, evidence
important clinical concern. Initial case reports were is lacking to recommend routine screening for hypo­
published in 2006, followed by other single case reports magnesaemia in patients without risk factors on PPI
and case series87–89. The prevalence of hypomagnesae- therapy or before initiating long-term PPI use. Monitor­
mia follow­ing long-term PPI use might have been ing for hypomagnesaemia is worth considering for
underestimated, especially in male patients, as reported those patients with additional risk factors (that is, renal
in a critical assessment of the FDA adverse event insufficiency or concomitant diuretic therapy) and is

Antrum-predominant gastritis Develops into corpus-predominant gastritis

PPIs

• ↑ IL-1β
• ↑ NH4
• ↑ other microorganisms
• Accelerate development of atrophy
Helicobacter in the fundus–corpus region
pylori

Figure 3 | Effect of acid inhibition with PPIs on Helicobacter pylori gastritis.


Nature Reviews | Gastroenterology
A shift from & Hepatology
antrum-predominant
inflammation to fundus–corpus-predominant inflammation occurs with acid inhibition via proton-pump inhibitors (PPIs).
The occurrence of Helicobacter pylori gastritis varies according to H. pylori virulence and host susceptibility genes and
is related to diverse clinical outcomes. Corpus-predominant gastritis is associated with a markedly higher risk of
developing malignancy than antrum-predominant gastritis. PPIs alter the gastric milieu by contributing to increased
IL‑1β, a potent pro-inflammatory factor, and ammonia (NH4). The catalytic activity of H. pylori urease on urea leads to
the formation of NH4, and PPIs could further promote the production of ammonia through the biological nitrogen
cycle involving other gut bacteria. IL‑1β and NH4 contribute to as well as result from the fact that H. pylori shifts from
the antrum to the corpus. Consequently, H. pylori might even totally disappear from the antrum, but it increases the
inflammation in the fundus–corpus.

NATURE REVIEWS | GASTROENTEROLOGY & HEPATOLOGY ADVANCE ONLINE PUBLICATION | 7


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

recommended in those suspected to have symptoms homeo­stasis are still lacking. However, in the analysis of
potentially due to hypomagnesaemia, such as cramps, the LOTUS and SOPRAN cohorts comparing anti-reflux
paresthesias and cardiac arrhythmias. surgery with long-term PPI intake for 5 and 12 years,
respectively, no differences in calcium and vitamin D
Hypocalcaemia. PPI-induced hypocalcaemia is serum levels were observed between groups with or
described in case reports related to PPI-associated without PPI use108. Thus, the available evidence currently
hypomagnesaemia100,101. The clinical importance of does not support that impaired calcium absorption is a
PPI-associated hypocalcaemia has been emphasized relevant PPI-associated mechanism.
and critically discussed in studies showing an increased
risk of bone fractures among patients on long-term PPI Fractures and osteoporosis. The association of PPI
therapy 102. The existing data are conflicting, and the intake with impaired bone mineralization has been the
exact pathophysiological mechanism of PPI-induced subject of many investigations with disparate outcomes
hypocalcaemia is still unclear. Experimental evidence (TABLE 2). An increased risk of osteoporosis with hip
indicates that PPIs might inhibit paracellular calcium fractures was first reported in two observational studies
transport by increasing the luminal pH, similar to the from the UK and Denmark in 2006 (REF. 109). An early
interference with the intestinal absorption of magne- study reported an adjusted odds ratio of 1.44 (95% CI
sium103–105. With short-term acid inhibition, PPI intake 1.30–1.59) for hip fracture in patients >65 years of age
does not influence calcium absorption. In a randomized, on PPIs for >1 year 110. The findings were strengthened
placebo-controlled trial including 12 healthy volunteers by the dose-dependent and time-dependent effect of
in a crossover design, inhibition of gastric acid secretion PPI intake (adjusted OR 2.65, 95% CI 1.80–3.90). In a
by PPI intake did not affect the absorption of calcium. large retrospective Canadian study of 15,792 patients
All patients received standardized meals with defined with osteoporosis-related fractures and 47,289 matched
amounts of protein, phosphate and calcium. Gastric controls, no increased risk of osteoporotic fractures was
acid suppression under PPI intake was monitored by observed in patients taking PPIs for <6 years. For PPI
gastric pH-metry 106. Another prospective study included intake >6 years, a 1.62‑fold risk of hip fracture (95% CI
21 postmenopausal women and did not find effects on 1.02–2.58) was calculated111.
calcium absorption after 30 days of PPI-induced gas- Two different meta-analyses confirmed only a mar-
tric acid inhibition107. Population-based studies investi­ ginally increased risk of hip fractures (OR 1.25) in older
gating the long-term effect of PPI therapy on calcium patients on long-term PPI therapy. A meta-analysis of

Table 2 | Association between long-term PPI intake and osteoporotic changes and risk of fractures*
Reference End point of the study and results Type of study
Yang et al. 110
• Risk of hip fractures Observational,
• OR 1.44 (95% CI 1.30–1.59): older patients (>65 years); duration of PPI case–control study
intake >1 year
• Dose- and time-dependent effects: adjusted OR 2.65 (95% CI 1.8–3.90)
Vestergaard • Risk of hip fractures Observational,
et al.109 • OR 1.45 (95% CI 1.28–1.65) for PPI intake within the last year case–control study
• OR 0.69 (95% CI 0.57–0.84) for H2 receptor antagonist intake within
the last year
Targownik et al.111 • Risk of hip fractures Observational,
• OR 1.62 (95% CI 1.02–2.58); only in patients on PPIs >6 years case–control study
• No risk for patients on PPIs <6 years
Ye et al.112 • Risk of hip fractures Meta-analysis, seven
• OR 1.24 (95% CI 1.15–1.34) observational studies
Ngamruengphong • Risk of hip fractures Meta-analysis, ten
et al.113 • OR 1.25 (95% CI 1.14–1.37) observational studies
Corley et al.114 • Risk of hip fractures Observational,
• OR 1.30 (95% CI 1.21–1.39): on PPIs and H2 receptor antagonists; case–control study
only in the presence of other risk factors (that is, pre-existing osteoporosis
or chronic steroid therapy)
Kaye & Jick115 • Risk of hip fractures Observational,
• No risk in the absence of other risk factors case–control study
Targownik et al.116 • Risk of osteoporotic changes and bone mineral density loss Observational,
• No effect of PPIs on bone mineral density case–control study
Chen et al.117 • Risk of osteoporosis Observational,
• HR 1.5 (95% CI 1.39–1.62) for osteoporosis case–control study
• No risk of fractures
*Authors’ bias of most important studies investigating the association between PPI intake and risk of fractures and
osteoporotic changes based on study design, enrolled patients and robustness of data. HR, hazard ratio; OR, odds ratio;
PPI, proton-pump inhibitor.

8 | ADVANCE ONLINE PUBLICATION www.nature.com/nrgastro


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

a Clopidogrel (Prodrug) therapy should be aware of this risk association and can
consider on an individual basis whether measures (that
Cytochrome P450 Oxidation is, bone mineral density measurement and adequate
(CYP2C19) Hydrolysis
therapy) are required. We caution against the general
Active metabolite unjustified alarm of a risk to bone health by PPI intake.

b Drug interactions
Clopidogrel (Prodrug) Interaction of PPIs with clopidogrel. A number of
Cytochrome P450 Oxidation retrospective analyses suggest an association of PPI
(CYP2C19) Hydrolysis ­co‑medication with an increased rate of adverse cardio­
vascular events — composite ischaemic end points,
Loss-of-function mutations Active metabolite
all-cause mortality, non-fatal myocardial infarction,
stroke, revascularization and stent thrombosis — in
c PPIs Clopidogrel (Prodrug) patients on clopidogrel 118–120. PPIs and clopidogrel
are both metabolized via the same cytochrome P450
Cytochrome P450 Oxidation pathway (CYP2C19) and, therefore, a competition for
Hydroxy-metabolites (CYP2C19) Hydrolysis
and the same pathway has been hypothesized121,122 (FIG. 4).
5-O-desmethyl-metabolites Active metabolite Ex vivo studies showed that, in contrast to omeprazole,
neither pantoprazole nor esomeprazole or lansoprazole
Normal activation Reduced activation influence the antiplatelet effects of clopidogrel123,124.
These data suggest that drug interaction between clopi-
Figure 4 | Activation of clopidogrel via cytochrome
Nature ReviewsP450. a | Normal activation
| Gastroenterology of
& Hepatology
dogrel and PPIs might be agent-specific rather than
clopidogrel occurs via CYP2C19. b | Loss‑of‑function mutation of CYP2C19 leads to
reduced activation of clopidogrel. c | A possible reduced activation of clopidogrel has class-­specific. Consequently, the FDA and European
been suggested in patients on proton-pump inhibitor (PPI) therapy (particularly for Medicines Agency warned in 2009 against combin-
omeprazole) due to competition for the same pathway. ing clopidogrel with PPIs (particularly omeprazole).
However, in randomized controlled trials, omepra-
zole did not increase the risk of adverse cardiovascular
seven observational studies found a pooled odds ratio events compared with placebo but did reduce upper
of 1.24 (95% CI 1.15–1.34)112. This finding was repli- gastrointestinal bleeding 125,126. Despite these new data,
cated in another meta-analysis that included ten studies on 10 March 2016, the FDA published a reminder to
(OR 1.25, 95% CI 1.14–1.37)113. avoid the concomitant use of clopidogrel and omepra-
Another large case–control study similarly des­ zole127, emphasizing that this r­ ecommendation applies
cribed a low increased risk of hip fractures in patients only to omeprazole and not to all PPIs.
on PPI medication as well as on H2 receptor antagonists The interindividual CYP2C19 genetic variability is
(OR 1.30, 95% CI 1.21–1.39) but only in patients with at an additive factor responsible for a reduced activation
least one other risk factor (pre-existing osteoporosis or of clopidogrel, with a number of loss‑of‑function muta-
chronic steroid therapy). In the absence of other addi- tions of the CYP2C19 gene128. The frequencies of these
tional risk factors, no association between PPI intake polymorphic alleles show marked interethnic variation
and an increased risk of hip fractures was observed114,115. (up to 35% in the Chinese, Japanese and Indian popu­
The discussion on the possible association between lation and up to 15% in the white European popula-
PPI intake and an increased risk of both osteoporosis tion)129,130. This aspect was not taken into consideration
and accelerated loss of bone mineral density remains in many of the early studies on the interaction between
open. In a North American study, PPI use was not clopidogrel and PPIs. Data from the TRIUMPH obser-
associated with either an increased risk of osteoporosis vational cohort study showed an association between
or an accelerated loss of bone mineral density 116. The PPI use and the risk of cardiac rehospitalization among
authors concluded that hip fractures in older patients clopidogrel-treated white patients who had a myo­cardial
on long-term PPI therapy might occur independently infarction and were carriers of the CYP2C19*17 variant
from the occurrence of osteoporotic changes. In con- (HR 1.62, 95% CI 1.19–2.19)131; therefore, ­genotyping
trast to that study, a Taiwanese investigation including could be considered for all patients.
>10,000 patients with GERD on long-term therapy The conflicting results with respect to the interaction
with PPIs and 20,000 controls matched by sex-, age- between clopidogrel and omeprazole can be summarized
and comorbidities without GERD found an increased as follows: ex vivo analyses showed that omeprazole (but
risk of osteoporosis in patients with GERD taking PPIs not other PPIs) influenced the antiplatelet effects of
(HR 1.5, 95% CI 1.39–1.62), but no association with clopidogrel; retrospective studies suggested that all PPIs
hip fractures117. (class-specific effect) increased cardiovascular events
All studies show important limitations of retro­ in patients on clopidogrel; and prospective trials found
spective, observational studies. No prospective no interaction between omeprazole and clopidogrel.
studies evaluating the pharmacological effect of acid-­ Thus, the debate on whether PPIs (including omepra-
suppressive therapy on hip fractures are available. zole) increase the risk of coronary events in patients on
At present, phys­icians involved in the medical care of clopidogrel is far from resolved. Both clopidogrel and
patients with severe comorbidities on long-term PPI omeprazole have short plasma half-lives (1–2 h)130,132;

NATURE REVIEWS | GASTROENTEROLOGY & HEPATOLOGY ADVANCE ONLINE PUBLICATION | 9


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Table 3 | Key recommendations for the management of PPI-related adverse effects


Issue, concern or effect Management
Long-term PPI intake • Keep dose and duration to the essential to reduce the risk of possible adverse effects
• Test for and treat Helicobacter pylori gastritis to prevent the shift from
antrum-predominant to corpus-predominant gastritis
AIN • After early detection of AIN, PPI withdrawal leads to recovery of renal function
• Avoid PPI re‑exposure
CKD • Search for known aetiologies of CKD before assuming a role for PPI intake
• PPI withdrawal could be considered in individual cases
Hypomagnesaemia • Consider PPIs as a cause of hypomagnesaemia and hypocalcaemia in patients with CKD
and hypocalcaemia on diuretic therapy
• Consider monitoring for hypomagnesaemia and electrolytes in patients with risk factors
and long-term PPI intake
• In patients with risk factors for osteoporotic changes (that is, pre-existing osteoporosis,
steroid therapy or malabsorption), calcium and vitamin D supplementation is required
Fractures and • Consider PPI intake at the minimally effective dose, especially in elderly patients
osteoporosis • Vitamin D supplementation in case of deficiency
Myopathies • Exclude other aetiological factors
• In patients who recover from an episode of rhabdomyolysis possibly related to PPI
intake and have a definite indication for ongoing PPI treatment, the PPI should not
be coadministered with statins
Cardiovascular risk • Prescribe a PPI other than omeprazole to patients on clopidogrel
AIN, acute interstitial nephritis; CKD, chronic kidney disease; PPI, proton-pump inhibitor.

therefore, separating the timing of clopidogrel and clinically meaningful drug–drug interaction between
omepra­zole administration has been proposed as a PPIs and warfarin138.
strategy to attenuate the potential for drug–drug com- PPI intake impairs tyrosine kinase inhibitor efficacy
petition. However, the success of this strategy is under- by reducing drug absorption. In a retrospective study
mined by the increased bioavailability of omeprazole due of nearly 200 patients with advanced renal cell cancer
to repeated dosing. The FDA discourages this strategy treated with sunitinib, coadministration of PPIs was
as well133. For the time being, and taking into consider- associ­ated with poorer progression-free survival and
ation the lack of updated clinical guidelines, adherence overall survival than in those not taking PPIs139. A simi­lar
to authorities’ recommendations is best practice. PPIs finding was demonstrated in >500 patients with advanced
other than omeprazole can be prescribed to patients non-small-cell lung cancer treated with erlotinib140.
on clopidogrel whenever indicated and at the lowest Furthermore, effects related to tyrosine kinase inhibition
effective dose. were markedly less toxic in patients with non-small-cell
lung cancer concomitantly receiving PPIs with erlo-
Drug absorption. A systematic review identified tinib than in patients not taking PPIs. Interestingly,
16 high-­quality randomized crossover studies investi- erlotinib absorption in patients on PPIs was improved
gating whether acid suppression during anti-­secretory when administering the drug with an acidic beverage,
therapy through PPIs or H 2 receptor antagonists such as cola141.
might impair absorption of coadministered drugs134. A possible negative effect of PPIs on the absorption
Suppression of gastric acidity reduced the absorption of the oral chemotherapeutic agent capecitabine in the
of ketoconazole, itraconazole, atazanavir, cefpodoxime, context of a complex chemotherapeutic regimen has
L‑thyroxin, α‑methyldopa, enoxacin and dipyridamole been suggested to be responsible for the observed statis-
(with a median reduction of the maximum observed tically significant decrease in progression-free survival
concentration of 66.5%). Furthermore, anti-secretory and reduced overall survival in patients with advanced
therapy enhanced the absorption of nifedipine and gastro-oesophageal cancer 142. This finding has not been
digoxin (with an increase in the median area under the observed with oral fluoropyrimidine S-1 (REF. 143). These
curve (AUC) of 10%) and induced a twofold increase emerging data demand high-quality studies in this area.
in alendronate bioavailability 134. PPIs are reported
to potentiate the anticoagulation effect of vitamin K PPIs and endothelial homeostasis
antagonists by enhancing absorption as a consequence According to experimental data, the off-target effects of
of elevated intragastric pH135,136. However, in healthy PPIs observed in vitro provide new insights into the direct
volunteers, dexlansoprazole once daily, compared with effects of PPIs on vascular homeostasis, which could
placebo, did not influence the peak plasma concentra- account for cardiovascular, renal and neuro­vascular
tion or AUC of warfarin nor international normalized complications. The presumed mechanism is a direct
ratio values137. Furthermore, although PPIs and warfarin interference of PPIs with the endothelial homeo­stasis
are both metabolized by hepatic CYP enzymes, a study of asymmetric dimethylarginine (ADMA), an endo­
including 305 warfarin users found no evidence of a genous inhibitor of nitric oxide synthase (NOS)144.

10 | ADVANCE ONLINE PUBLICATION www.nature.com/nrgastro


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Several clinical studies have indicated that elevated reported in association with long-term PPI intake, only
ADMA plasma levels are associated with increased a few have proven causality and/or are based on potential
cardio­vascular risk. ADMA correlates with disease pathophysiological mechanisms induced by PPIs (FIG. 5).
severity, major adverse cardiovascular events and all- The effects of PPIs on their therapeutic target organ, the
cause mortality in patients with peripheral arterial dis- stomach, and the consequences of acid suppression on
ease and is discussed as an independent risk factor for the gastric mucosa structure and physiology are plausible
mortality in patients with peripheral arterial disease145,146. and evidence-based. The few reports on an increased risk
Studies have examined ADMA and arginine deriv­ of gastric and other gastrointestinal malignancies due to
atives in relation to vascular function in the general PPIs have important biases and confounding variables.
popu­lation. In a controlled trial, 4 weeks of PPI use did The association of PPIs with a variety of gastrointestinal
not markedly increase serum ADMA levels compared infections has been established, and the mechanism is
with placebo; however, substantial changes might occur plausible. Furthermore, PPIs induce changes in the gut
with long-term PPI treatment 147. Another mechanism microbiota composition with unknown health-related
that might explain the association of chronic PPI intake consequences. Several studies have failed to confirm
with cardiovascular, renal and neurological adverse an increased risk of community- and hospital-acquired
effects is an acceleration of endothelial senescence by pneumonia in patients taking PPIs.
impaired lysosomal activity, disrupted proteostasis in PPIs rarely affect vitamin B12 absorption, magne-
endothelial cells and telomere shortening 148–151. The sium and calcium homeostasis. The association of
in vitro effects of PPIs on endothelial cell ageing prov­ osteoporosis and bone fractures with PPI intake is of
ides stimulating hypotheses, but they are not ready for low degree and mostly confounded by clinically signifi­
translation into a clinical setting. cant comorbidities. PPIs are a rare cause of AIN and
other idiosyncrasy-­related events. Omeprazole (but
Conclusions not other PPIs) can increase the risk of coronary events
In acid-related diseases, the benefits of PPI intake out- in patients on clopidogrel. Beyond metabolic drug–
weigh their potential harms. TABLE 3 outlines key recom­ drug ­interactions, PPIs also impair the absorption of
mendations on how to manage and possibly prevent certain drugs.
PPI-related complications. PPIs should not be withheld In spite of the fact that most associations between
in patients with an appropriate indication, but dose and PPI intake and adverse events have substantial intrinsic
duration should be kept to the minimum. Among the weaknesses due to the inclusion of retrospective cohorts
long and continuously increasing list of adverse events and insufficient adjustment for comorbidities, co‑­
medications and the dose and duration of PPI exposure,
we should remain alert to capture any possible relevant
Acute kidney event associated with PPI intake. However, premature
Structural and injury Chronic kidney conclusions should not be drawn before these associ­
functional changes disease
in the gastric mucosa
ations are investigated in depth and a causal relationship
has been considered.
Dementia
Enteric infections*
In clinical practice, it is important to carefully assess
the individual management of our patients requiring
Gastrointestinal
SIBO malignancies long-term PPI therapy without causing alarm about
PPIs possible dangers induced by PPIs. This aspect is
Clostridium difficile Cardiovascular events, particu­larly important in the context of the u ­ ncritical
infection negative effect release of media reports on the inherent risks of PPI
on clopidogrel‡ intake. For the clinical management of patients on
Vitamin B12 long-term PPI therapy, general surveillance of blood
deficiency Pneumonia chemistry para­meters is not mandatory, but we sug-
gest monitoring renal function, serum electrolytes and
Hypomagnesaemia Osteoporosis micronutrients according to the management of their
Hypocalcaemia Bone fractures
individual comorbidities. H. pylori gastritis should be
cured in patients requiring long-term PPI therapy to
Causal relationship prevent progression to ­preneoplastic changes in the
Low evidence for causality gastric mucosa.
Insufficient evidence for causality There is a continued need for strong acid suppression
now and in the future to control the substantial mor-
Figure 5 | PPIs and adverse events with proven
Nature and unproven
Reviews causality. The
| Gastroenterology number
& Hepatology bidity and mortality in acid-related diseases. Clinical
of complications associated with proton-pump inhibitors (PPIs) is high. This figure shows studies will have to include prospective population-­
the adverse events linked to PPI use, with indicators of the evidence supporting causality.
based outcome studies in patients on long-term PPI
Those events with proposed major importance in clinical practice are shown according
to the scientific evidence and grading as per TABLE 1. *Enteric infections with treatment, starting by including patients at the time
Salmonella enterica subsp. enterica serovar Enteritidis, Salmonella enterica subsp. enterica when they are first exposed to a PPI. These studies
serovar Typhimurium and Campylobacter jejuni. ‡Association restricted to omeprazole; should include pharmacogenomics and should investi­
the FDA recommends avoiding the concomitant use of clopidogrel and omeprazole127. gate adverse events by involving interdisciplinary
SIBO, small intestinal bacterial overgrowth. medical expertise.

NATURE REVIEWS | GASTROENTEROLOGY & HEPATOLOGY ADVANCE ONLINE PUBLICATION | 11


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

1. Turner, T. Proton pump inhibitors. Drugwatch https:// effect of gastrin. J. Physiol. Pharmacol. 66, 159–167 44. Lo, W. K. & Chan, W. W. Proton pump inhibitor use
www.drugwatch.com/proton-pump-inhibitors/ (2017). (2015). and the risk of small intestinal bacterial overgrowth:
2. Statista. Top 20 pharmaceutical products by sales Experimental evidence of anti-carcinogenetic a meta-analysis. Clin. Gastroenterol. Hepatol. 11,
worldwide in 2014 (in billions US dollars). Statista effects of PPIs — these results are awaiting clinical 483–490 (2013).
https://www.statista.com/statistics/258022/top-10- confirmation. 45. Jacobs, C., Coss Adame, E., Attaluri, A., Valestin, J.
pharmaceutical-products-by-global-sales-2011/ (2017). 23. Poulsen, A. H. et al. Proton pump inhibitors and risk & Rao, S. S. Dysmotility and proton pump
3. Lessell, S. Omeprazole (correction of omepraxole) of gastric cancer: a population-based cohort study. inhibitor use are independent risk factors for small
and ocular damage. Concerns on safety of drug are Br. J. Cancer. 100, 1503–1507 (2009). intestinal bacterial and/or fungal overgrowth.
unwarranted. BMJ 316, 7124–7167 (1998). 24. Singh, M., Dhindsa, G., Friedland, S. Aliment. Pharmacol. Ther. 37, 1103–1111 (2013).
4. Stolte, M., Bethke, B., Rühl, G. & Ritter, M. & Triadafilopoulos, G. Long-term use of proton pump 46. Giamarellos-Bourboulis, E. J., Pyleris, E.,
Omeprazole-induced pseudohypertrophy of gastric inhibitors does not affect the frequency, growth, Barbatzas, C., Pistiki, A. & Pimentel, M. Small
parietal cells. Z. Gastroenterol. 30, 134–138 (1992). or histologic caracteristics of colon adenomas. intestinal bacterial overgrowth is associated with
5. Fiocca, R. et al. Gastric exocrine and endocrine cell Aliment. Pharmacol. Ther. 26, 1051–1061 (2007). irritable bowel syndrome and is independent of proton
morphology under prolonged acid inhibition therapy: 25. Robertson, D. J. et al. Proton pump inhibitor use pump inhibitor usage. BMC Gastroenterol. 16, 67
results of a 5‑year follow‑up in the LOTUS trial. and risk of colorectal cancer: a population-based, case- (2016).
Aliment. Pharmacol. Ther. 36, 959–971 (2012). control study. Gastroenterology 133, 755–760 (2007). 47. Leonard, J., Marshall, J. K. & Moayyedi, P.
A controlled study on the long-term effect of PPIs 26. Kearns, M. D., Boursi, B. & Yang, Y. X. Proton pump Systematic review of the risk of enteric
on endocrine and exocrine gastric cells. inhibitors on pancreatic cancer risk and survival. infection in patients taking acid suppression.
6. Cats, A. et al. Parietal cell protrusions and fundic Cancer Epidemiol. 46, 80–84 (2017). Am. J. Gastroenterol. 102, 2047–2056 (2007).
gland cysts during omeprazole maintenance 27. Kuipers, E. J. et al. Increase of Helicobacter pylori- 48. Doorduyn, Y., Van Den Brandhof, W. E.,
treatment. Hum. Pathol. 31, 684–690 (2000). associated corpus gastritis during acid suppressive Van Duynhoven, Y. T., Wannet, W. J. & Van Pelt, W.
7. Lamberts, R., Brunner, G. & Solcia, E. Effects of very therapy: implications for long-term safety. Risk factors for Salmonella Enteritidis and
long (up to 10 years) proton pump blockade on human Am. J. Gastroenterol. 90, 1401–1406 (1995). Typhimurium (DT104 and non‑DT104) infections
gastric mucosa. Digestion 64, 205–213 (2001). 28. Malfertheiner, P., Chan, F. K. & McColl, K. E. Peptic in the Netherlands: predominant roles for raw eggs in
8. Lundell, L., Vieth, M., Gibson, F., Nagy, P. ulcer disease. Lancet 374, 1449–1461 (2009). Enteritidis and sandboxes in Typhimurium infections.
& Kahrilas, P. J. Systematic review: the effects of 29. Hunt, R. H. et al. The stomach in health and disease. Epidemiol. Infect. 134, 617–626 (2006).
long‑term proton pump inhibitor use on serum gastrin Gut 64, 1650–1668 (2015). 49. Bavishi, C. & Dupont, H. L. Systematic review: the use
levels and gastric histology. Aliment. Pharmacol. Ther. A comprehensive update on all aspects of gastric of proton pump inhibitors and increased susceptibility
42, 649–663 (2015). functions. to enteric infection. Aliment. Pharmacol. Ther. 34,
This systematic review reports on long-term 30. Logan, R. P. et al. Changes in the intragastric 1269–1281 (2011).
PPI-induced moderate hypergastrinaemia in distribution of H. pylori during treatment with 50. Hassing, R. J., Verbon, A., de Visser, H., Hofman, A.
most patients and an increased prevalence of omeprazole. Gut 36, 12–16 (1995). & Stricker, B. H. Proton pump inhibitors and
enterochromaffin-like cell hyperplasia; Helicobacter 31. Stolte, M., Meining, A., Schmitz, J. M., Alexandridis, T. gastroenteritis. Eur. J. Epidemiol. 31, 1057–1063
pylori-positive patients receiving long-term PPI & Seifert, E. Changes in Helicobacter pylori-induced (2016).
therapy were exposed to a higher risk of corpus gastritis in the antrum and corpus during 12 months 51. Kwok, C. S. et al. Risk of Clostridium difficile infection
atrophy than were H. pylori-negative patients. of treatment with omeprazole and lansoprazole in with acid suppressing drugs and antibiotics: meta-
9. Schubert, M. L. Gastric acid secretion. Curr. Opin. patients with gastro-oesophageal reflux disease. analysis. Am. J. Gastroenterol. 107, 1011–1019
Gastroenterol. 32, 452–460 (2016). Aliment. Pharmacol. Ther. 12, 247–253 (1998). (2012).
10. Reimer, C., Søndergaard, B., Hilsted, L. & Bytzer, P. 32. Kuipers, E. J. et al. Atrophic gastritis and Helicobacter 52. Tleyjeh, I. M. et al. Association between proton pump
Proton-pump inhibitor therapy induces acid-related pylori infection in patients with reflux esophagitis inhibitor therapy and clostridium difficile infection:
symptoms in healthy volunteers after withdrawal treated with omeprazole or fundoplication. N. Engl. a contemporary systematic review and meta-analysis.
of therapy. Gastroenterology 137, 80–87 (2009). J. Med. 334, 1018–1022 (1996). PLoS ONE 7, e50836 (2012).
11. Hongo, M., Fujimoto, K. & Gastric Polyps Study 33. Eissele, R., Brunner, G., Simon, B., Solcia, E. 53. Arriola, V. et al. Assessing the risk of hospital-acquired
Group. Incidence and risk factor of fundic gland polyp & Arnold, R. Gastric mucosa during treatment Clostridium difficile infection with proton pump
and hyperplastic polyp in long-term proton pump with lansoprazole: Helicobacter pylori is a risk factor inhibitor use: a meta-analysis. Infect. Control Hosp.
inhibitor therapy: a prospective study in Japan. for argyrophil cell hyperplasia. Gastroenterology 112, Epidemiol. 28, 1–10 (2016).
J. Gastroenterol. 45, 618–624 (2010). 707–717 (1997). A meta-analysis suggesting an almost twofold
12. Martin, F. C., Chenevix-Trench, G. & Yeomans, N. D. 34. Lundell, L. et al. Changes of gastric mucosal increased risk of hospital-acquired Clostridium
Systematic review with meta-analysis: fundic architecture during long-term omeprazole difficile infection in patients on PPIs.
gland polyps and proton pump inhibitors. therapy: results of a randomized clinical trial. 54. Kandel, C. E., Gill, S. E.,McCready, J., Matelski, J.
Aliment. Pharmacol. Ther. 44, 915–925 (2016). Aliment. Pharmacol. Ther. 23, 639–647 (2006). & Powis, J. E. Reducing co‑administration of proton
13. Tran-Duy, A., Spaetgens, B., Hoes, A. W., de Wit, N. J. 35. Malfertheiner, P. et al. Management of Helicobacter pump inhibitors and antibiotics using a computerized
& Stehouwer, C. D. Use of proton pump inhibitors pylori infection-the Maastricht V/Florence Consensus order entry alert and prospective audit and feedback.
and risks of fundic gland polyps and gastric Report. Gut 66, 6–30 (2017). BMC Infect. Dis. 16, 355 (2016).
cancer: systematic review and meta-analysis. This manuscript presents the latest 55. Chande, N. & Driman, D. K. Microscopic colitis
Clin. Gastroenterol. Hepatol. 14, 1706–1719 (2016). recommendations on the management of associated with lansoprazole: report of two cases
The latest systematic review and meta-analysis Helicobacter pylori infection and the role of PPIs. and a review of the literature. Scand. J. Gastroenterol.
reporting that PPI use increases the incidence 36. Sanduleanu, S., Jonkers, D., De Bruïne, A., 42, 530–533 (2007).
of fundic gland polyps; a small effect on gastric Hameeteman, W. & Stockbrügger, R. W. Double 56. Wilcox, G. M. & Mattia, A. R. Microscopic colitis
cancer is biased by confounding factors. gastric infection with Helicobacter pylori and non- associated with omeprazole and esomeprazole
14. Worthley, D. L. et al. Gastric adenocarcinoma and Helicobacter pylori bacteria during acid-suppressive exposure. J. Clin. Gastroenterol. 43, 551–553
proximal polyposis of the stomach (GAPPS): a new therapy: increase of pro-inflammatory cytokines and (2009).
autosomal dominant syndrome. Gut 61, 774–779 development of atrophic gastritis. Aliment. Pharmacol. 57. Keszthelyi, D. et al. Proton pump inhibitor use is
(2012). Ther. 8, 1163–1175 (2001). associated with an increased risk for microscopic
15. Yanaru-Fujisawa, R. et al. Familial fundic gland 37. Schulz, C., Koch, N., Schütte, K., Pieper, D. H. colitis: a case-control study. Aliment. Pharmacol. Ther.
polyposis with gastric cancer. Gut 61, 1103–1104 & Malfertheiner, P. H. pylori and its modulation of 32, 1124–1128 (2010).
(2012). gastrointestinal microbiota. J. Dig. Dis. 16, 109–117 58. Verhaegh, B. P. et al. High risk of drug-induced
16. Sanduleanu, S. et al. Serum chromogranin A as a (2015). microscopic colitis with concomitant use of NSAIDs
screening test for gastric enterochromaffin-like cell 38. Freedberg, D. E. et al. Proton pump inhibitors and proton pump inhibitors. Aliment. Pharmacol. Ther.
hyperplasia during acid-suppressive therapy. alter specific taxa in the human gastrointestinal 43, 1004–1013 (2016).
Eur. J. Clin. Invest. 31, 802–811 (2001). microbiome: a crossover trial. Gastroenterology 149, 59. Xu, H. B. et al. Proton pump inhibitor use and risk of
17. Savarino, V., Dulbecco, P. & Savarino, E. Are proton 883–885 (2015). spontaneous bacterial peritonitis in cirrhotic patients:
pump inhibitors really so dangerous? Dig. Liver Dis. 39. Jackson, M. A. et al. Proton pump inhibitors alter the a systematic review and meta-analysis. Genet. Mol.
48, 851–859 (2016). composition of the gut microbiota. Gut 65, 749–756 Res. 14, 7490–7501 (2015).
18. Waldum, H. L., Hauso, Ø., Brenna, E., Qvigstad, G. (2016). 60. Terg, R. et al. Proton pump inhibitor therapy does
& Fossmark, R. Does long-term profound inhibition References 37 and 38 provide novel insights into not increase the incidence of spontaneous bacterial
of gastric acid secretion increase the risk of ECL cell- the effect of PPIs on gut microbiota composition. peritonitis in cirrhosis: a multicenter prospective
derived tumors in man? Scand. J. Gastroenterol. 51, 40. Imhann, F. et al. Proton pump inhibitors affect the gut study. J. Hepatol. 62, 1056–1060 (2015).
767–773 (2016). microbiome. Gut 65, 740–748 (2016). 61. Dam, G., Vilstrup, H., Watson, H. & Jepsen, P.
19. Jianu, C. S. et al. Gastric carcinoids after long-term 41. Clooney, A. G. et al. A comparison of the gut Proton pump inhibitors as a risk factor for hepatic
use of a proton pump inhibitor. Aliment. Pharmacol. microbiome between long-term users and non-users encephalopathy and spontaneous bacterial peritonitis
Ther. 36, 644–649 (2012). of proton pump inhibitors. Aliment. Pharmacol. Ther. in patients with cirrhosis with ascites. Hepatology 64,
20. Pregun, I. et al. Effect of proton-pump inhibitor 43, 974–984 (2016). 1265–1272 (2016).
therapy on serum chromogranin a level. Digestion 84, 42. Lombardo, L., Foti, M., Ruggia, O. & Chiecchio, A. A relevant study showing that PPIs increase
22–28 (2011). Increased incidence of small intestinal bacterial the risk of developing hepatic encephalopathy
21. Wong, H. et al. PPI-delayed diagnosis of gastrinoma: overgrowth during proton pump inhibitor therapy. and spontaneous bacterial peritonitis in patients
oncologic victim of pharmacologic success. Clin. Gastroenterol. Hepatol. 8, 504–508 (2010). with liver cirrhosis.
Pathol. Oncol. Res. 16, 87–91 (2010). 43. Ratuapli, S. K. et al. Proton pump inhibitor therapy 62. Fohl, A. L. & Regal, R. E. Proton pump inhibitor-
22. Han, Y. M. et al. Role of proton pump inhibitors use does not predispose to small intestinal bacterial associated pneumonia: not a breath of fresh air after
in preventing hypergastrinemia-associated overgrowth. Am. J. Gastroenterol.107, 730–735 all? World J. Gastrointest. Pharmacol. Ther. 2, 17–26
carcinogenesis and in antagonizing the trophic (2012). (2011).

12 | ADVANCE ONLINE PUBLICATION www.nature.com/nrgastro


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

63. Alshamsi, F. et al. Efficacy and safety of proton pump 84. McColl, K. E. Effect of proton pump inhibitors on 106. Wright, M. J. et al. Inhibiting gastric acid production
inhibitors for stress ulcer prophylaxis in critically ill vitamins and iron. Am. J. Gastroenterol. 104, S5–S9 does not affect intestinal calcium absorption in young,
patients: a systematic review and meta-analysis (2009). healthy individuals: a randomized, crossover,
of randomized trials. Crit. Care 20, 120 (2016). 85. Venerito, M. et al. Oxyntic gastric atrophy in controlled clinical trial. J. Bone Miner. Res. 25,
64. Laheij, R. J., Van Ijzendoorn, M. C., Janssen, M. J. Helicobacter pylori gastritis is distinct from 2205–2211 (2010).
& Jansen, J. B. Gastric acid-suppressive therapy autoimmune gastritis. J. Clin. Pathol. 69, 677–685 107. Hansen, K. E. et al. Do proton pump inhibitors
and community-acquired respiratory infections. (2016). decrease calcium absorption? J. Bone Miner. Res. 25,
Aliment. Pharmacol. Ther. 18, 847–851 (2003). 86. al‑Ghamdi, S. M., Cameron, E. C. & Sutton, R. A. 2786–2795 (2010).
65. Eom, C. S. et al. Use of acid-suppressive drugs and risk Magnesium deficiency: pathophysiologic and clinical 108. Attwood, S. E. et al. Long-term safety of proton pump
of pneumonia: a systematic review and meta-analysis. overview. Am. J. Kidney Dis. 24, 737–752 (1994). inhibitor therapy assessed under controlled,
CMAJ 183, 310–319 (2011). 87. Epstein, M., McGrath, S. & Law, F. Proton-pump randomised clinical trial conditions: data from the
66. Filion, K. B. et al. Proton pump inhibitors and the risk inhibitors and hypomagnesemic hypoparathyroidism. SOPRAN and LOTUS studies. Aliment. Pharmacol.
of hospitalisation for community-acquired pneumonia: N. Engl. J. Med. 355, 1834–1836 (2006). Ther. 41, 1162–1174 (2015).
replicated cohort studies with meta-analysis. Gut 63, 88. Hoorn, E. J. et al. A case series of proton pump This study shows data from controlled randomized
552–558 (2014). inhibitor-induced hypomagnesemia. Am. J. Kidney Dis. trials with reassuring findings on long-term PPI use,
67. Estborn, L. & Joelson, S. Frequency and time to onset 56, 112–116 (2010). electrolytes and micronutrients.
of community-acquired respiratory tract infections 89. Mackay, J. D. & Bladon, P. T. Hypomagnesaemia due 109. Vestergaard, P., Rejnmark, L. & Mosekilde, L. Proton
in patients receiving esomeprazole: a retrospective to proton-pump inhibitor therapy: a clinical case pump inhibitors, histamine H2 receptor antagonists,
analysis of patient-level data in placebo-controlled series. QJM 103, 387–395 (2010). and other antacid medications and the risk of fracture.
studies. Aliment. Pharmacol. Ther. 42, 607–613 90. Luk, C. P., Parsons, R., Lee, Y. P. & Hughes, J. D. Calcif. Tissue Int. 79, 76–83 (2006).
(2015). Proton pump inhibitor-associated hypomagnesemia: 110. Yang, Y. X., Lewis, J. D., Epstein, S. & Metz, D. C.
A large cohort study of a PPI (esomeprazole) what do FDA data tell us? Ann. Pharmacother. 47, Long-term proton pump inhibitor therapy and risk
that disproves the risk of community-acquired 773–780 (2013). of hip fracture. JAMA 296, 2947–2953 (2006).
pneumonia. 91. Quamme, G. A. Recent developments in intestinal 111. Targownik, L. E. et al. Use of proton pump inhibitors
68. Abramowitz, J. et al. Adverse event reporting for magnesium absorption. Curr. Opin. Gastroenterol. 24, and risk of osteoporosis-related fractures. CMAJ 179,
proton pump inhibitor therapy: an overview of 230–235 (2008). 319–326 (2008).
systematic reviews. Otolaryngol. Head Neck Surg. 92. Voets, T. et al. TRPM6 forms the Mg2+ influx channel 112. Ye, X. et al. Proton pump inhibitors therapy and
155, 547–554 (2016). involved in intestinal and renal Mg2+ absorption. risk of hip fracture: a systematic review and meta-
69. Gomm, W. et al. Association of proton pump inhibitors J. Biol. Chem. 279, 19–25 (2004). analysis. Eur. J. Gastroenterol. Hepatol. 9, 794–800
with risk of dementia: a pharmacoepidemiological 93. Schlingmann, K. P. et al. Hypomagnesemia with (2011).
claims data analysis. JAMA Neurol. 73, 410–416 secondary hypocalcemia is caused by mutations in 113. Ngamruengphong, S., Leontiadis, G., Radhi, S.,
(2016). TRPM6, a new member of the TRPM gene family. Dentino, A. & Nugent, K. Proton pump inhibitors and
This study was an adverse event report showing a Nat. Genet. 31, 166–170 (2002). risk of fracture: a systematic review and meta-analysis
significantly increased risk of incident dementia in 94. Zipursky, J. et al. Proton pump inhibitors and of observational studies. Am. J. Gastroenterol. 106,
conjunction with PPI use; this report caused great hospitalization with hypomagnesemia: a population- 1209–1218 (2011).
concern in the general public. based case-control study. PLoS Med. 11, e1001736 114. Corley, D. A., Kubo, A., Zhao, W. & Quesenberry, C.
70. Booker, A., Jacob, L. E., Rapp, M., Bohlken, J. (2014). Proton pump inhibitors and histamine‑2 receptor
& Kostev, K. Risk factors for dementia diagnosis in A population-based case–control study showing antagonists are associated with hip fractures among
German primary care practices. Int. Psychogeriatr. 28, an increased risk of hospitalization with at‑risk patients. Gastroenterology 139, 93–101
1059–1065 (2016). hypomagnesaemia in patients on PPIs that (2010).
71. Clark, D. W. & Strandell, J. Myopathy including are also receiving diuretics. 115. Kaye, J. A. & Jick, H. Proton pump inhibitor use and
polymyositis: a likely class adverse effect of proton 95. Sumukadas, D., McMurdo, M. E. T. & Habicht, D. risk of hip fractures in patients without major risk
pump inhibitors? Eur. J. Clin. Pharmacol. 62, Proton pump inhibitors are associated with lower factors. Pharmacotherapy 28, 951–959 (2008).
473–479 (2006). magnesium levels in older people with chronic kidney 116. Targownik, L. E., Lix, L. M., Leung, S. & Leslie, W. D.
72. Ruffenach, S. J., Siskind, M. S. & Lien, Y. H. Acute disease. J. Am. Geriatr. Soc. 60, 392–393 (2012). Proton-pump inhibitor use is not associated with
interstitial nephritis due to omeprazole. Am. J. Med. 96. Danziger, J. et al. Proton-pump inhibitor use is osteoporosis or accelerated bone mineral density loss.
93, 472–473 (1992). associated with low serum magnesium concentrations. Gastroenterology 138, 896–904 (2010).
73. Perazella, M. A. & Markowitz, G. S. Drug-induced Kidney Int. 83, 692–699 (2013). An accurate study shedding light on the association
acute interstitial nephritis. Nat. Rev. Nephrol. 6, 97. Misra, P. S., Alam, A., Lipman, M. L. & Nessim, S. J. between PPIs and osteoporosis or accelerated
461–470 (2010). The relationship between proton pump inhibitor bone mineral density loss.
74. Muriithi, A. K. et al. Biopsy-proven acute interstitial use and serum magnesium concentration among 117. Chen, C. H., Lin, C. L. & Kao, C. H. Gastroesophageal
nephritis, 1993‑2011: a case series. Am. J. Kidney hemodialysis patients: a cross-sectional study. reflux disease with proton pump inhibitor use is
Dis. 64, 558–566 (2014). BMC Nephrol. 16, 136 (2015). associated with an increased risk of osteoporosis:
75. Moledina, D.,G. & Perazella, M. A. PPIs and kidney 98. Sharara, A. I. et al. Low prevalence of a nationwide population-based analysis.
disease: from AIN to CKD. J. Nephrol. 29, 611–616 hypomagnesemia in long-term recipients of proton Osteoporos. Int. 27, 2117–2126 (2016).
(2016). pump inhibitors in a managed care cohort. 118. Serbin, M. A., Guzauskas, G. F. & Veenstra, D. L.
76. Blank, M. L., Parkin, L., Paul, C. & Herbison, P. Clin. Gastroenterol. Hepatol. 14, 317–321 (2016). Clopidogrel-proton pump inhibitor drug-drug
A nationwide nested case-control study indicates An important study showing that, in the absence interaction and risk of adverse clinical outcomes
an increased risk of acute interstitial nephritis with of known precipitating factors (concomitant diuretic among PCI-treated ACS patients: a meta-analysis.
proton pump inhibitor use. Kidney Int. 86, 837–844 intake, chronic diarrhoea, chronic kidney disease J. Manag. Care Spec. Pharm. 22, 939–947 (2016).
(2014). and malignancies), chronic PPI use is not A meta-analysis of randomized controlled trials
77. Antoniou, T. et al. Proton pump inhibitors and the risk associated with hypomagnesaemia. and observational studies, showing that
of acute kidney injury in older patients: a population- 99. Biyik, M. et al. Hypomagnesemia among outpatient concomitant clopidogrel–PPI therapy following
based cohort study. CMAJ Open 3, E166–E171 long-term proton pump inhibitor users. Am. J. Ther. percutaneous coronary intervention is significantly
(2015). 24, e52–e55 (2017). associated with adverse cardiovascular events.
78. Geevasinga, N., Coleman, P. L., Webster, A. C. 100. Negri, A. L. & Valle, E. E. Hypomagnesaemia/ 119. Sherwood, M. W. et al. Individual proton pump
& Roger, S. D. Proton pump inhibitors and acute hypokalemia associated with the use of esomeprazole. inhibitors and outcomes in patients with coronary
interstitial nephritis. Clin. Gastroenterol. Hepatol. 4, Curr. Drug Saf. 6, 204–206 (2011). artery disease on dual antiplatelet therapy:
597–604 (2006). 101. Deroux, A., Khouri, C., Chabre, O., Bouillet, L. a systematic review. J. Am. Heart Assoc. 4, e002245
79. Lazarus, B. et al. Proton pump inhibitor use and & Casez, O. Severe acute neurological symptoms (2015).
the risk of chronic kidney disease. JAMA Intern. Med. related to proton pump inhibitors induced 120. Melloni, C. et al. Conflicting results between
176, 238–246 (2016). hypomagnesemia responsible for profound randomized trials and observational studies on the
This study report comprehensive data showing hypoparathyroidism with hypocalcemia. Clin. Res. impact of proton pump inhibitors on cardiovascular
that PPI use is associated with a low risk of incident Hepatol. Gastroenterol. 38, e103–e105 (2014). events when coadministered with dual antiplatelet
chronic kidney disease. 102. Leontiadis, G. I. & Moayyedi, P. Proton pump therapy: systematic review. Circ. Cardiovasc. Qual.
80. Xie, Y. et al. Proton pump inhibitors and risk of inhibitors and risk of bone fractures. Curr. Treat. Outcomes 8, 47–55 (2015).
incident CKD and progression to ESRD. J. Am. Options Gastroenterol. 12, 414–423 (2014). 121. Gilard, M., Arnaud, B., le Gal, G., Abgrall, J. F.
Soc. Nephrol. 10, 3153–3163 (2016). 103. Thongon, N. & Krishnamra, N. Apical acidity decreases & Boschat, J. Influence of omeprazol on the
81. Maggio, M. et al. Proton pump inhibitors and risk of inhibitory effect of omeprazole on Mg(2+) absorption antiplatelet action of clopidogrel associated to aspirin.
1‑year mortality and rehospitalization in older patients and claudin‑7 and -12 expression in Caco‑2 J. Thromb. Haemost. 4, 2508–2509 (2006).
discharged from acute care hospitals. JAMA Intern. monolayers. Exp. Mol. Med. 44, 684–693 (2012). 122. Focks, J. J. et al. Concomitant use of clopidogrel
Med. 173, 518–523 (2013). 104. Lameris, A. L. L., Hess, M. W., van Kruijsbergen, I., and proton pump inhibitors: impact on platelet
82. Leontiadis, G. I., Sharma, V. K. & Howden, C. W. Hoenderop, J. G. J. & Bindels, R. J. M. Omeprazole function and clinical outcome: a systematic review.
Proton pump inhibitor therapy for peptic ulcer enhances the colonic expression of the Mg(2+) Heart 99, 520–527 (2013).
bleeding: Cochrane collaboration meta-analysis of transporter TRPM6. Pflugers Arch. 465, 1613–1620 123. Siller-Matula, J. M. et al. Effects of pantoprazole and
randomized controlled trials. Mayo Clin. Proc. 82, (2013). esomeprazole on platelet inhibition by clopidogrel.
286–296 (2007). 105. Hess, M. W., de Baaij, J. H. F., Gommers, L. M. M., Am. Heart J. 157, 148.e1–148.e5 (2009).
83. Lam, J. R., Schneider, J. L., Zhao, W. & Corley, D. A. Hoenderop, J. G. J. & Bindels, R. J. M. Dietary inulin 124. Small, D. S. et al. Effects of the proton pump inhibitor
Proton pump inhibitor and histamine 2 receptor fibers prevent proton-pump inhibitor (PPI)-induced lansoprazole on the pharmacokinetics and
antagonist use and vitamine B12 deficiency. JAMA hypocalcemia in mice. PLoS ONE 10, e0138881 pharmacodynamics of prasugrel and clopidogrel.
310, 2435–2442 (2013). (2015). J. Clin. Pharmacol. 48, 475–484 (2008).

NATURE REVIEWS | GASTROENTEROLOGY & HEPATOLOGY ADVANCE ONLINE PUBLICATION | 13


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

125. Bhatt, D. L., et al. Clopidogrel with or without crossover, placebo-controlled, single-centre studies. 149. Goligorsky, M. S. Pathogenesis of endothelial cell
omeprazole in coronary artery disease. N. Engl. Clin. Drug Investig. 29, 35–50 (2009). dysfunction in chronic kidney disease: a retrospective
J. Med. 363, 1909–1917 (2010). 138. Henriksen, D. P. et al. The potential drug-drug and what the future may hold. Kidney Res. Clin. Pract.
126. Gao, Q. P. Sun, Y., Sun, Y. X., Wang, L. F. & Fu, L. interaction between proton pump inhibitors 34, 76–82 (2015).
Early use of omeprazole benefits patients with acute and warfarin. Pharmacoepidemiol. Drug Saf. 24, 150. Di Marco, L. Y. et al. Vascular dysfunction in the
myocardial infarction. J. Thromb. Thrombolysis 28, 1337–1340 (2015). pathogenesis of Alzheimer’s disease — a review
282–287 (2009). 139. Ha, V. H. et al. Does gastric acid suppression affect of endothelium-mediated mechanisms and
127. FDA. FDA reminder to avoid concomitant use of Plavix sunitinib efficacy in patients with advanced or ensuing vicious circles. Neurobiol. Dis. 82, 593–606
(clopidogrel) and omeprazole. FDA https://www.fda.gov/ metastatic renal cell cancer? J. Oncol. Pharm. Pract. (2015).
Drugs/DrugSafety/ucm231161.htm (2016). 21, 194–200 (2015). 151. Flammer, A. J. et al. The assessment of endothelial
128. Simon, T. et al. Genetic determinants of response to 140. Chu, M. P. et al. Gastric acid suppression is function: from research into clinical practice.
clopidogrel and cardiovascular events. N. Engl. J. Med. associated with decreased erlotinib efficacy in non- Circulation 126, 753–767 (2012).
360, 363–375 (2009). small-cell lung cancer. Clin. Lung Cancer 16, 33–39 152. Malfertheiner, P. et al. Management of Helicobacter
129. Rosemary, J. & Adithan, C. The pharmacogenetics of (2015). pylori infection — the Maastricht IV/ Florence
CYP 2C9 and CYP2C19: ethnic variation and clinical 141. van Leeuwen, R. W. et al. Influence of the acidic Consensus Report. Gut 61, 646–664 (2012).
significance. Curr. Clin. Pharmacol. 2, 93–109 beverage cola on the absorption of erlotinib in
(2007). patients with non-small-cell lung cancer. J. Clin. Oncol. Author contributions
130. Venerito, M., Kandulski, A. & Malfertheiner, P. 34, 1309–1314 (2016). All authors contributed equally to the writing and reviewing
Dilemma between gastroprotection and 142. Chu, M. P. et al. Association of proton pump of the draft manuscript.
cardiovascular prevention. Dtsch. Med. Wochenschr. inhibitors and capecitabine efficacy in advanced
135, 2193–2198 (2010). gastroesophageal cancer: secondary analysis Competing interests statement
131. Depta, J. P. et al. Clinical outcomes associated with of the TRIO‑013/LOGiC randomized clinical trial. P.M. is a member of the advisory boards for Allergan, Alfa
proton pump inhibitor use among clopidogrel-treated JAMA Oncol. 3, 767–773 (2017). Wassermann and Bayer and has taken part in speakers’
patients within CYP2C19 genotype groups following A secondary analysis of a phase III randomized bureaus for Allergan, AstraZeneca, Reckitt Benckiser
acute myocardial infarction. Pharmacogenomics J. 15, trial, showing that PPIs negatively affect and Takeda. M.V. is a member of the advisory boards for
20–25 (2015). capecitabine efficacy, possibly by raising gastric Amgen, Lilly and Nordic and has taken part in a speakers’
132. Laine, L. & Hennekens, C. Proton pump inhibitor pH levels; PPI-treated patients with advanced bureau for Bayer and Merck Serono. A.K. declares no
and clopidogrel interaction: fact or fiction? gastro-oesophageal cancer receiving competing interests.
Am. J. Gastroenterol. 105, 34–41 (2010). capecitabine-based polychemotherapy had poorer
133. FDA. Information for healthcare professionals: update progression-free survival and overall survival than Publisher’s note
to the labeling of clopidogrel bisulfate (marketed as those not treated with PPIs. Springer Nature remains neutral with regard to jurisdictional
Plavix) to alert healthcare professionals about a drug 143. Scheulen, M. E. et al. Effect of food and a proton pump claims in published maps and institutional affiliations.
interaction with omeprazole (marketed as Prilosec inhibitor on the pharmacokinetics of S-1 following oral
and Prilosec OTC). FDA https://www.fda.gov/Drugs/ administration of S-1 in patients with advanced solid Review criteria
DrugSafety/PostmarketDrugSafetyInformationfor tumors. Cancer Chemother. Pharmacol. 69, 753–761 Bibliographic searches in PubMed, Embase and Medline for
PatientsandProviders/DrugSafetyInformationforHeath (2012). literature published until 31 Dec 2016 were performed using
careProfessionals/ucm190787.htm (2009). 144. Ghebremariam, Y. T. et al. Unexpected effect of proton the term “PPI” with various complications: “gastric”, “intesti-
134. Lahner, E., Annibale, B. & Delle Fave, G. Systematic pump inhibitors: elevation of the cardiovascular risk nal”, “infections”, “cardiovascular”, “renal”, “osteological”,
review: impaired drug absorption related to the factor asymmetric dimethylarginine. Circulation 128, “metabolic” and “neurologic”. Some new reports were
co‑administration of antisecretory therapy. 845–853 (2013). included during the revision of the paper in April 2017.
Aliment. Pharmacol. Ther. 29, 1219–1229 (2009). 145. Wilson, A. M. et al. Asymmetric dimethylarginine Systematic reviews since 2010 reporting on adverse events
135. Julkunen, R. J. The absorption of warfarin from the rat correlates with measures of disease severity, major related to PPI use were also included. Only human studies
stomach in situ. Med. Biol. 54, 260–263 (1976). adverse cardiovascular events and all-cause mortality published in English were considered; selected experimental
136. Scarpignato, C. et al. Effective and safe proton pump in patients with peripheral arterial disease. Vasc. Med. studies were cited to explain mechanisms leading to compli-
inhibitor therapy in acid-related diseases - a position 15, 267–274 (2010). cations. The strength of the supporting evidence for the asso-
paper addressing benefits and potential harms of acid 146. Böger, R. H. et al. Asymmetric dimethylarginine as an ciation between PPI intake and various complications was
suppression. BMC Med. 14, 179 (2016). independent risk marker for mortality in ambulatory graded according to the system used in previous reports152
A position statement on the benefits, harms patients with peripheral arterial disease. J. Intern. (listed in Table 1).
and appropriateness of PPI use. Med. 269, 349–361 (2011).
137. Vakily, M., Lee, R. D., Wu, J., Gunawardhana, L. 147. Ghebremariam, Y. T. et al. Proton pump inhibitors and
& Mulford, D. Drug interaction studies with vascular function: a prospective cross-over pilot study. DATABASES
dexlansoprazole modified release (TAK‑390MR), Vasc. Med. 4, 309–316 (2015). VigiBase: https://www.who-umc.org/vigibase/vigibase
a proton pump inhibitor with a dual delayed-release 148. Yepuri, G. et al. Proton pump inhibitors accelerate ALL LINKS ARE ACTIVE IN THE ONLINE PDF
formulation: results of four randomized, double-blind, endothelial senescence. Circ. Res. 118, e36–e42 (2016).

14 | ADVANCE ONLINE PUBLICATION www.nature.com/nrgastro


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.

You might also like