You are on page 1of 13

809927

review-article2019
TAW0010.1177/2042098618809927Therapeutic Advances in Drug SafetyM Jaynes and AB Kumar

Therapeutic Advances in Drug Safety Review

The risks of long-term use of proton


Ther Adv Drug Saf

2019, Vol.10: 1­–13

pump inhibitors: a critical review DOI: 10.1177/


https://doi.org/10.1177/2042098618809927
https://doi.org/10.1177/2042098618809927
2042098618809927

© The Author(s), 2019.


Article reuse guidelines:
Megan Jaynes and Avinash B. Kumar sagepub.com/journals-
permissions

Abstract: Proton pump inhibitors (PPIs) are among the most frequently prescribed
medications. Their use is likely even higher than estimated due to an increase in the
number of PPIs available without a prescription. Appropriate indications for PPI use include
Helicobacter pylori infection, erosive esophagitis, gastric ulcers, and stress ulcer prevention
in high-risk critically ill patients. Unfortunately, PPIs are often used off-label for extended
periods of time. This increase in PPI usage over the past two decades has called into question
the long-term effects of these medications. The association between PPI use and infection,
particularly Clostridium difficile and pneumonia, has been the subject of several studies. It’s
proposed that the alteration in gastrointestinal microflora by PPIs produces an environment
conducive to development of these types of infections. At least one study has suggested that
long-term PPI use increases the risk of dementia. Drug interactions are an important and
often overlooked consideration when prescribing any medication. The potential interaction
between PPIs and antiplatelet agents has been the subject of multiple studies. One of
the more recent concerns with PPI use is their role in the development or progression of
chronic kidney disease. There is also some literature suggesting that PPIs contribute to
the development of various micronutrient deficiencies. Most of the literature examining the
potential adverse effects of PPI use is composed of retrospective, observation studies. There
is a need for higher quality studies exploring this relationship.

Keywords: PPIs, proton pump inhibitors, side effects

Received: 23 July 2018; revised manuscript accepted: 28 September 2018.

Introduction narrow approval of the US Food and Drug Correspondence to:


Avinash B. Kumar
The management of acid peptic disease was revo- Administration (FDA). The ‘off-label’ use of Division of Critical
lutionized by the introduction of proton pump medications is prevalent in the intensive care Care, Department of
Anesthesiology, Vanderbilt
inhibitors (PPIs) into clinical practice almost environment, and PPIs account for the highest University, Nashville, TN
three decades ago. Today, PPIs remain among off-label use (as high as 55% prevalence) in inten- 37212, USA
avinash.b.kumar@
the most widely prescribed medications in the sive care units.4 The widespread and often open- vanderbilt.edu
world. PPIs as a class of medication also have a ended use of medications, even those with a Megan Jaynes
high prevalence of being prescribed for poorly relatively safe profile, can have negative, unin- Division of Critical Care,
Department of Pharmacy,
defined reasons or for conditions where PPIs have tended consequences in the long term. Some of Vanderbilt University
not been shown to be beneficial. The current evi- the main inappropriate uses of PPIs are for the Medical Center, Nashville,
TN, USA
dence suggests PPIs are often overused with 25– prevention of gastroduodenal ulcers in low-risk
70% of prescriptions having no appropriate patients, low-dose steroid therapy without addi-
indication.1,2 In the United States (US) alone, tional risk factors, systemic anticoagulation with-
PPIs account for >$10 billion in healthcare costs out additional risk factors for gastroduodenal
and the global costs exceed $25 billion/year.3 injury, and the overtreatment of functional dys-
Clinicians can legally prescribe medications based pepsia. The US FDA and the National Institute
on their individual interpretation of the scientific for Clinical Excellence in the United Kingdom
evidence or clinical judgment regardless of the published guidelines on the indications for

journals.sagepub.com/home/taw 1

Creative Commons Non Commercial CC BY-NC: This article is distributed under the terms of the Creative Commons Attribution-NonCommercial 4.0 License
(http://www.creativecommons.org/licenses/by-nc/4.0/) which permits non-commercial use, reproduction and distribution of the work without further permission
provided the original work is attributed as specified on the SAGE and Open Access pages (https://us.sagepub.com/en-us/nam/open-access-at-sage).
Therapeutic Advances in Drug Safety 10

prescribing PPIs (especially in hospitalized The mechanistic basis of this association is not
patients). The major indications included erosive clear at this time. Especially because the associa-
esophagitis, nonsteroidal anti-inflammatory drug tion was based on retrospective observational
(NSAID)-induced dyspepsia and sequelae, criti- data. Thus not completing eliminating confound-
cally ill patients on mechanical ventilation, and ers in the analyses.
the treatment of Helicobacter pylori (H. pylori) in
conjunction with antibiotics.5 Postulated mechanisms linking CDI with PPI
use. There are a number of potential pathophysi-
A large number of mainly observational studies ologic mechanisms that have been described in
on a wide range of possible associations with the the literature. Although C. difficile spores are rela-
use of PPIs have been published in the past dec- tively resistant to gastric hydrochloric acid, the
ade. Additionally, a number of meta-analyses on long-term gastric acid suppression with PPIs may
the same subject have almost doubled the num- alter the colonic microbiome to decrease coloniza-
ber of publications reporting on the long-term tion resistance or other normal barriers to C. dif-
effect of PPI use in varied patient populations in ficile proliferation.9 A small number of studies that
the last decade. have evaluated the gut microbiome using high-
throughput genomic sequencing have shown
In this review, we sought to evaluate the major marked decreases in the diversity of the bacterial
reported associations and gain some clarity on the flora within 30 days of starting PPIs. This loss of
long-term effects of PPIs. microbial diversity is a consistent feature in CDI
patients. This loss of diversity may eliminate nutri-
The major associations reported in the literature ent competition between the gut microbiome and
about the potential adverse effects of the long- favor the growth of C. difficile in the utilization of
term use of PPIs are outlined below: available amino acids (especially monomeric glu-
cose, N-acetylglucosamine, and sialic acids).12
i. PPIs and Clostridium difficile (C. difficile) Other potential host and microbiological path-
infection ways are yet to be clearly understood in the patho-
ii. PPIs and dementia genesis of CDI in the PPI-exposed cohorts.
iii. PPIs and pneumonia
iv. PPIs and antiplatelet agents Summary. The various strategies in the preven-
v. PPIs and kidney disease tion of CDI should begin with the cessation of the
vi. PPIs and micronutrient deficiency medications without strong indications and close
vii. PPIs and bone mineral density reassessment of PPI use, especially in the inten-
sive care patient population

i. Long-term use of PPIs and the risk of C.


difficile infection ii. Long-term use of PPIs and the risk of
The introduction of PPIs into clinical practice dementia
revolutionized the management of acid peptic Dementia is a silent and progressive disorder
disease and gastroesophageal reflux disease characterized by deterioration in cognitive ability
(GERD). The use of PPIs has increased several- that severely debilitates the individual and affects
fold over the last two decades and one of the their ability to live independently. It is a disorder
inappropriate indications often attributed to of age, with the incidence increasing as age
this rise is the use of PPIs for the prevention of advances and more importantly does not have a
gastroduodenal ulcers in low-risk patients. As cure at this time. Besides the tremendous social,
the data accumulated with years of usage, an emotional and caregiver burden that dementia
epidemiologic association between the use of imposes, the associated worldwide financial costs
hypochlorhydric agents and the increased risk of of dementia patients were estimated at greater
acquired enteric infections such as C. difficile than 600 billion dollars in 2010.13 With world-
emerged.6,7 A brief summary of the recent stud- wide efforts directed at primary prevention, the
ies exploring the relationship between PPI expo- focus on modifiable risk factors becomes key.
sure and the development of C. difficile infection One such factor is long-term medication use for
(CDI) are shown in Table 1. Table 2 summa- other conditions. One of the early, large epide-
rizes studies exploring the role of PPIs and the miological studies based on the German ageing,
recurrence of CDI.8–11 cognition and dementia databases showed a

2 journals.sagepub.com/home/taw
Table 1. Studies evaluating the association between PPI use and the risk of developing C. difficile infections.

Authors Paper Type Number of Pooled Results Conclusion


patients studies

Arriola47 Assessing the risk of Meta-analysis 186,033 23 Pooled OR Increased risk of hospital-acquired C.
hospital-acquired C. difficile 1.81 difficile

journals.sagepub.com/home/taw
infection with PPI use

Deshpande48 Association between PPI Meta-analysis 202,965 33 (25 OR 2.15 Increased risk of C. difficile
therapy and C. difficile CC+cohort)
infection in a meta-analysis

Roughhead49 PPIs and risk of C. difficile Sequence 54957 Health ASR 2.40 Increased risk of C. difficile
infection: a multi-country symmetry Canada and
study using sequence ASPEN
symmetry

Ro50 Risk of C. difficile infection Retrospective 1005 (6.7% versus PPI therapy is associated with a higher
with the use of a PPI for 1.8%) risk of SUP-related CDI than H2RA
stress ulcer prophylaxis in therapy in critically ill patients. (6.7%
critically ill patients versus 1.8%)

Dos Santos- Recurrence and death after Retrospective 373 Pre-existing PPI therapy may increase
Schalle8 C. difficile infection: gender monocentric the risk of recurrence or death in male
dependent influence of PPI cohort study patients with a toxicogenic CDI
therapy

Barletta51 PPIs increase the risk for Retrospective, 408 OR 2.03 (CI Proton pump inhibitors are independent
hospital-acquired C. difficile case-control 1.23–3.36) risk factors for the development of CDI
in critically ill study in ICU patients

McDonald10 Continuous PPI therapy and Retrospective 754 Two university Recurrence Patients with continuous PPI use
associated risk of recurrent cohort hospitals in HR 1.5 (95% remained at elevated risk of CDI
C. difficile Canada CI, 1.1–2.0) recurrence

Stevens52 Differential risk of C. difficile Retrospective 10,154 HR 4.95 The effect of PPI on the risk of CDI
infection with PPI use and cohort is significantly modified by antibiotic
ABX exposure exposure
Gordon53 Incidence of C. difficile Retrospective 20,944 OR: 2.2; 95% The use of PPIs together with high-
infection in patients receiving cohort CI 1.52–3.23 risk antibiotics was associated with a
high risk ABX with or without significantly higher incidence of CDI
PPI

ABX, Antibiotics; ASR, adjusted sequence ratio; CDI, C. difficile infection; CI, confidence interval; H2RA, histamine-2 receptor antagonist; HR, hazard ratio; ICU, intensive care unit;
OR, odds ratio; PPI, proton pump inhibitor; SUP, stress ulcer prophylaxis.

3
M Jaynes and AB Kumar
Therapeutic Advances in Drug Safety 10

Table 2. Studies evaluating the association between PPI use and the risk of developing recurrent C. difficile
infections.

Authors Paper Type Number Conclusion


of
patients

McDonald and Continuous PPI therapy Retrospective 754 PPI use remained at elevated
colleagues10 and associated risk of cohort risk of CDI recurrence.
recurrent CDI infection. Cessation of unnecessary PPI
JAMA Intern Med 2015; use should be considered at
175(5): 784–791 the time of CDI diagnosis.

Freedberg and PPIs and risk for Retrospective 894 Receipt of PPIs concurrent
colleagues9 recurrent CDI among with CDI treatment was
inpatients. Am J NOT associated with CDI
Gastroenterol 2013; recurrence.
108(11): 1794–1801.

Linsky and PPIs and risk for Retrospective 1154 PPI use during incident CDI
colleagues54 recurrent CDI infection. treatment was associated
Arch Intern with a 42% increased risk of
Med 2010; 170(9): recurrence
772–778.

Kim and PPI as a risk factor for Retrospective 125 Advanced age, serum
colleagues55 recurrence of CDI- albumin level <2.5 g/dl, and
associated diarrhea. concomitant use of PPIs were
World J Gastroenterology found to be significant risk
2010; 16: 3573–3577. factors for CDI recurrence

Cadle and Association of PPI with Retrospective 140 PPI therapy was associated
colleagues56 outcomes in CDI colitis. with an increased risk of
Am J Health Syst Pharm recurrent CDI colitis
2007; 64: 2359–2363.

Lupse and J Gastrointestin Liver Dis Retrospective 306 The risk of first recurrence was
colleagues11 2013; 22: 397–403. significantly higher in patients
older than 70 who also received
PPI treatment
Dos Santos- Recurrence and death Retrospective 373 Pre-existing PPI therapy may
Schaller and after CDI infection: increase the risk of recurrence
colleagues8 SpringerPlus 2016; 5: or death in male patients with a
430. toxicogenic CDI

CDI, C. difficile infection; PPI, proton pump inhibitor.

significantly elevated risk of developing dementia cells use enzymes such as V-ATPase to degrade
in patients exposed to long-term PPI therapy.13 A and scavenge beta amyloid. Murine models sug-
subsequent study conducted on a longitudinal gest that PPIs interfere with the activity of scaven-
sample of elderly patients from the largest German ger enzymes such as V-ATPase leading to the
statutory health insurer also showed an increased accumulation of beta amyloid.15 Further studies
risk of developing dementia compared with are needed to elucidate the mechanism linking
patients with no exposure to PPIs.14 PPI usage with dementia in humans.

Postulated mechanism linking dementia and PPI Summary. These two large observational studies
use. The buildup of beta amyloid has been impli- show a small effect size but have a low quality of
cated in the progression and pathogenesis of evidence. The ongoing need for PPI therapy
dementia syndromes such as Alzheimer’s disease should be closely evaluated particularly in elderly
in humans. Central nervous system microglial patients. It is important to note that in these large

4 journals.sagepub.com/home/taw
Table 3. Studies evaluating the association between PPI use and the risk of developing nosocomial pneumonia.

Authors Paper Type Number of Pooled Results Conclusion


patients studies

Momosaki24 PPIs versus H2RAs and risk Retrospective observation 77,890 NA OR 1.10 No significant difference in the
of pneumonia in patients with study 95% CI incidence of PNA between users of

journals.sagepub.com/home/taw
acute stroke 0.99–1.21 PPIs and H2RAs after acute stroke.

Arai23 Histamine H2-blocker and Retrospective 132 NA RR 2.07 The incidence of pneumonia was
PPI use and the risk of observational study 95% CI higher in patients exposed to PPIs.
pneumonia in acute stroke: 1.13–3.62 The incidence of pneumonia in
a retrospective analysis of patients exposed to H2 blockers
susceptible patients was not higher than in patients
not exposed to acid suppression
therapy.

Alshamsi57 Efficacy and safety of PPIs Meta-analysis 1571 13 RR 1.12 PPI use does not significantly
for stress ulcer prophylaxis 95% CI increase risk of pneumonia in
in critically ill patients: a 0.86–1.46 critically ill patients.
systematic review and meta- p = 0.39
analysis of randomized trials

Lambert18 Risk of community acquired Meta-analysis 6,351,656 26 OR 1.49 Outpatient PPI use is associated
pneumonia with outpatient CAP = 95% CI with an increased risk of CAP
PPI therapy 226,769 1.16–1.92

MacLaren20 H2RAs versus PPIs on GI tract Pharmacoepidemiological 35,312 NA OR 2.3 PPIs are associated with higher
hemorrhage and infectious cohort study (71 hospitals) 95% CI rate of pneumonia than H2RA in
complications in the ICU 1.03–1.41 mechanically ventilated patients.

Herzig19 Acid-suppressive medication Pharmacoepidemiological 56,330 NA OR 2.8 Use of acid-suppressive therapy


use and the risk of hospital- cohort study 95% CI was associated with increased risk
acquired pneumonia (non ICU patients) 2.5–3.1 of hospital-acquired pneumonia.

Khorvash58 The comparison between PPIs Single-center, randomized 137 NA 14.1% versus Rate on pneumonia significantly
and sucralfate in incidence of controlled trial 36.4% lower in patients receiving
VAP in critically ill patients p < 0.0001 sucralfate than pantoprazole.
Batemen22 Type of stress ulcer Retrospective cohort study 21,214 NA RR 1.19 Patients treated with PPIs had
prophylaxis and risk of 95% CI a small increase in the risk of
nonsocomial PNA in cardiac 1.03–1.38 postoperative PNA compared with
surgery patients: cohort study patients treated with H2RAs.

CAP, Community acquired Pneumonia; CDI, C. difficile infection; CI, confidence interval; GI, gastrointestinal; H2RA, histamine-2 receptor antagonist; HR, hazard ratio; ICU, intensive care
unit; NA, not applicable; OR, odds ratio; PNA, Pneumonia; PPI, proton pump inhibitor; RR, relative risk; VAP, ventilator-associated pneumonia.

5
M Jaynes and AB Kumar
6
Table 4. Studies evaluating the association between PPI use and chronic kidney disease.

Authors Paper Type Number of Pooled Results Conclusion


patients studies

Lazarus and PPI use and risk of chronic Prospective, 11,656 NA AKI PPI use but not H2RA use
colleagues32 kidney disease cohort study HR 1.72 was an independent risk
CI 1.28–2.3 factor for AKI and CKD
p < 0.001
CKD
HR 1.45
CI 1.11–1.9
p = 0.006

Klatte and Association between proton Observational, 114,883 NA SCr Doubling PPI use was associated
colleagues36 pump inhibitor use and risk cohort study HR 1.26 with higher risk of CKD
of progression of chronic CI 1.05–1.51 progression than H2RA
Therapeutic Advances in Drug Safety 10

kidney disease >30% Decline in use


GFR
HR 1.26
CI 1.16–1.36

Arora59 Proton pump inhibitors are Retrospective 99,269 NA CKD PPI use is associated
associated with increased case-control HR 1.1 with increased odds of
risk of development of study CI 1.05–1.16 developing CKD and death
chronic kidney disease p < 0.0001
Mortality
HR 1.76
CI 1.68–1.84
p < 0.0001

Xie and Long-term kidney Retrospective, 144,032 NA CKD Proton pump inhibitor use
colleagues37 outcomes among users cohort study HR 1.29 is associated with CKD in
of proton pump inhibitors CI 1.22–1.36 patients without prior AKI.
without intervening acute
kidney injury

Xie and Proton pump inhibitors and Retrospective, 193,591 NA CKD PPI exposure is
colleagues35 risk of incident CKD and cohort study HR 1.28 associated with an
progression to ESRD CI 1.23–1.34 increased risk of incident
CKD prog/ESRD CKD, CKD progression,
HR 1.47 and ESRD.
CI 1.38–1.57
Wijarnpreecha Associations of proton- Meta-Analysis 536,902 5 CKD or ESRD There is an increased
and pump inhibitors and H2 RR 1.33 risk of CKD and ESRD in
colleagues33 receptor antagonists with CI 1.18–1.51 patients using PPIs.
chronic kidney disease: a
meta-analysis

AKI, acute kidney injury; CI, confidence interval; CKD, Chronic kidney disease; ESRD, End stage renal disease; GFR, glomerular filtration rate; HR; Hazard ratio; PPI, proton pump

journals.sagepub.com/home/taw
inhibitor; RR, relative risk; Scr, Serum Creatinine.
M Jaynes and AB Kumar

population-based studies, the PPIs were not first 14 days of hospital admission. The incidence
assigned at random but were related to patient of pneumonia was higher in patients receiving
characteristics (e.g. PPIs prescribed because of PPIs versus no acid suppression therapy, but there
older age and NSAID-associated dyspepsia). This was no difference in the incidence of pneumonia
results in differences between PPI users and non- between patients receiving PPIs versus H2RAs.23
users in factors that may impact study outcomes Together, these studies suggest that the relation-
and confound results (residual bias).16,17 ship between PPI use and pneumonia is not lim-
ited to a specific subgroup of hospitalized patients.

iii. Long-term PPI use and the risk of Postulated mechanism of HAP/VAP in patients on
pneumonia long-term PPI use. By suppressing gastric acid
Healthcare-acquired pneumonia (HAP) and ven- release, acid suppressants increase gastric pH,
tilator-associated pneumonia (VAP) are a signifi- which may promote bacterial overgrowth lead-
cant source of morbidity and mortality in critically ing to tracheal colonization and pneumonia.23,24
ill patients. In the late 1990s to early 2000s, sev- Evidence also indicates that PPIs may impair
eral studies were published suggesting an associa- immune cell function, increasing the risk of
tion between the use of PPIs in the outpatient infectious complications.22
setting and development of community-acquired
pneumonia. A meta-analysis of these studies Summary. On the surface, based on the currently
found that the risk of pneumonia was increased as available literature, there appears to be an associa-
early as the first month of therapy.18 This sparked tion between the long-term use of PPIs and devel-
an interest in the potential relationship between opment of HAP/VAP. Although the mechanism
PPI usage in the inpatient setting and the devel- and association seems biologically plausible, the
opment of pneumonia. One of the early studies overall quality of evidence is low. The risk of devel-
examining this relationship evaluated the associ- oping VAP/HAP does not seem to be substantially
ated between any type of acid suppression ther- increased and was observed in studies where resid-
apy and development of HAP. They found the ual confounding is highly likely. Thus, in reality, the
incidence of HAP was higher in patients receiving clinical relevance of the observed association is
acid suppression therapy. A stratified analysis probably less significant. For now, providers should
according to subcategories of acid suppression aim to limit the use of acid suppression therapy to
therapy, found this association was maintained in patients with a clear indication.
patients receiving PPIs but not in patients receiv-
ing histamine-2 receptor antagonists (H2RAs).19
Several studies have compared the risk of pneu- iv. Long-term use of PPIs and antiplatelet
monia in patients receiving PPIs versus H2RAs in agents
the inpatient setting. In one large pharmacoepi- One of the early concerns surrounding the use of
demiologic cohort study, including more than PPIs was a potential interaction with the antiplate-
35,000 patients, investigators found that PPIs let agent clopidogrel, a prodrug requiring activa-
were associated with higher rates of pneumonia tion by the CYP2C19 enzyme system. The active
than H2RAs in mechanically ventilated patients.20 metabolite of clopidogrel is responsible for irre-
versibly binding to the ADP receptors on platelets,
There have also been several recent studies exam- inhibiting their aggregation.25 PPIs competitively
ining the association between acid suppression inhibit CYP2C19 to varying degrees, with ome-
therapy and pneumonia in specific patient popu- prazole likely the most significant inhibitor.
lations. In patients with nontraumatic intracranial Pantoprazole does not inhibit CYP2C19. A 2009
hemorrhage, PPI prophylaxis was associated with study, including 105 patients undergoing high-
an increased risk of HAP versus no gastrointesti- risk angioplasty, found that platelet reactivity was
nal prophylaxis.21 A retrospective cohort study of ~25% higher in patients receiving a PPI in addi-
cardiac surgery patients found that patients tion to clopidogrel than in patients not receiving
treated with PPIs had an increased risk of postop- concomitant PPI therapy.26 Several other studies
erative pneumonia compared with patients who suggesting a potential interaction between PPIs,
received H2RAs.22 A retrospective study of primarily omeprazole, and clopidogrel prompted a
patients with acute stroke, primarily ischemic 2009 US FDA label warning, recommending the
stroke, examined patient exposure to PPIs and avoidance of concomitant administration of clopi-
H2RAs and the incidence of pneumonia in the dogrel and omeprazole due to a concern of a

journals.sagepub.com/home/taw 7
Therapeutic Advances in Drug Safety 10

reduction in clopidogrel’s active metabolite levels interaction, multiple other smaller or retrospec-
and antiplatelet activity. tive studies have been conducted with varying
results. The two meta-analyses, each including over
Since the US FDA warning, multiple studies have 80,000 patients, found a higher rate of adverse car-
explored the safety of the concomitant administra- diovascular events in patients receiving concomi-
tion of PPIs and clopidogrel. A retrospective cohort tant PPI-clopidogrel therapy.31 While it appears
study of ~8200 patients assessed the risk of adverse there is certainly a pharmacodynamic interaction
outcomes associated with concomitant use of PPIs between PPIs and clopidogrel, the clinical signifi-
and clopidogrel following acute coronary syn- cance of this interaction for most patients is not
drome. It found an increased risk of adverse out- well elucidated. Given the conflicting data cur-
comes in patients receiving clopidogrel with a PPI rently available, further study is indicated.
versus clopidogrel alone.27 Perhaps the most well-
known study exploring the clopidogrel–omepra-
zole relationship is the COGENT study. It was a v. Long-term PPI use and the risk of kidney
multicenter, double-blind, placebo-controlled trial disease
that found no difference in the composite cardio- Chronic kidney disease (CKD) affects more than
vascular endpoint between patients who received 10% of the US population, and is associated with
omeprazole versus placebo. While this was a large, significant morbidity and mortality, as well as a
well-designed trial, it had several limitations worth considerable economic burden to the healthcare
noting. First, it was terminated prematurely due to system.32,33 Shortly after the introduction of PPIs,
a small event number. It also used a single clopi- case reports suggesting an association between
dogrel-omeprazole pill with pharmacokinetics that their use and the development of acute interstitial
were different from either of the commercially nephritis (AIN) emerged.35 This association has
available agents.28 been substantiated by further studies, but it is
only in the last decade that the potential relation-
As previously mentioned, different PPIs inhibit ship between PPIs and the development or pro-
CYP2C19 to varying degrees, with omeprazole gression of CKD has been examined. The first
being the most potent inhibitor and pantoprazole large-scale study published examining the rela-
being the least potent. A cohort study including tionship between PPI use and CKD included two
over 20,000 patients hospitalized with myocardial individual patient cohorts intended to represent
infarction, coronary artery revascularization, or the general population. In each group both the
unstable angina did not find a significant relation- adjusted and unadjusted analysis found a signifi-
ship between PPI use and an increased risk of cant positive relationship between PPI use and
serious cardiovascular disease. It is noteworthy the development of CKD. Interestingly, H2RA
that the majority of patients in this study were use was not found to be associated with CKD in
receiving pantoprazole and fewer than 10% were either cohort.32 The results of a similarly designed
receiving omeprazole.29 A small, prospective trial study by Xie and colleagues paralleled those of
of patients with acute myocardial infarction found the Lazarus study. They found that PPI use was
that platelet function, assessed using the Verify associated with a higher incidence of CKD, kid-
Now system, was significant higher in patients ney disease progression and end-stage renal dis-
who received omeprazole versus pantoprazole.30 ease (ESRD).35 They also found that increased
duration of exposure was associated with an
Postulated mechanism of relationship between increased risk of adverse renal outcomes up to
PPIs and antiplatelet agents. PPI have been 720 days of exposure; after 720 days, this associa-
shown to inhibit certain CYP enzymes; the degree tion disappeared. A subsequent large, Swedish
of inhibition varies among the different agents cohort study also found an association between
within the class. Some antiplatelet agents rely on cumulative PPI use and CKD progression.36 The
these CYP enzymes to be metabolized to their most recent evaluation of PPI exposure and CKD
active form. In theory, the use of a PPI could pre- by Li and colleagues using complex pharmacoep-
vent an antiplatelet agent from being activated idemiologic tools to estimate the effect of unmeas-
and decrease the antiplatelet effects. ured/unknown confounders on the relationship of
PPI use and risk of CKD suggests that confound-
Summary. While the COGENT study remains ing factors alone were unlikely to explain the
the only large-scale prospective randomized con- reported association. Please refer to table 4 for
trolled trial exploring the clopidogrel–omeprazole summary of the studies.

8 journals.sagepub.com/home/taw
M Jaynes and AB Kumar

Postulated mechanism of CKD in patients on long- the risk of hypomagnesemia.39 Another study
term PPI use. The mechanism responsible for the examined this relationship in over 400 hemodial-
association between PPI use and CKD is not well ysis patients. Serum magnesium levels were sig-
understood. While the risk of AIN-induced acute nificantly lower in PPI users than nonusers (0.94
kidney injury with PPI use is well established, versus 1.03, p < 0.0001).43
whether or not this is the sole mechanism by
which PPIs are associated with CKD remains Hypomagnesemia often goes undetected in the
unanswered. Other proposed mechanisms by general population; thus, the true incidence of
which PPIs may cause renal dysfunction include PPI-related hypomagnesemia is difficult to deter-
abnormalities in lysosomal acidification hydro- mine and is likely under reported.41 Most evi-
gen/potassium adenosine triphosphatase enzyme dence suggests that hypomagnesemia is a problem
system, decreased regeneration of renal tubular for chronic PPI users, rather than patients who
cells, increased oxidative stress, and altered gene receive them for short-term gastrointestinal
expression.37 Further study is needed to gain a prophylaxis in the intensive care unit. Patients
better understanding of this relationship. with CKD and those receiving medications
known to lower magnesium concentration, such
Summary. Given the current evidence, providers as diuretics, are also thought to be at higher risk.
should be prudent in evaluating the need for ini-
tiation and continuation of PPIs, particularly in Other micronutrient deficiencies. Calcium: The
patients with known CKD or risk factors for low, acid-rich pH in the stomach facilitates the
development of CKD. release of ionized calcium from insoluble calcium
salts. Long-term PPI use has been linked to a
decrease in the enteral absorption of calcium. It is
vi. Long-term PPI use and the risk worth noting that his mechanism does not influ-
hypomagnesemia ence the absorption of water-soluble calcium salts
Hypomagnesemia occurs in up to 65% of criti- or calcium when it is contained in complex food
cally ill patients and may increase the risk of both like milk or cheese.15 Omeprazole specifically has
short and long-term complications.38,39 Acutely, been evaluated with tracer methods and found
magnesium has been shown to decrease inflam- that it may impede the absorption of calcium car-
mation, reduce platelet aggregation, and prevent bonate in older women. There are insufficient
arrhythmias.38 Chronic hypomagnesemia may data at this time to make recommendations about
increase the risk of cardiovascular disease, diabe- the need for calcium supplementation in patients
tes, and osteoporosis.39 While hypomagnesemia on PPI therapy.
in critically ill patients is often multifactorial, the
use of PPIs is a proposed contributor. The patho- Vitamin B12: Gastric acidity is important for
physiology behind the relationship of PPI use and the absorption of dietary vitamin B12. The acidic
hypomagnesemia is not well understood. One milieu in the native stomach helps release the
hypothesis is that the pH change induced by PPI food-bound B12 and facilitates binding with
use alters the affinity of magnesium transport intrinsic factors to be absorbed downstream in
receptors for magnesium, decreasing the active the terminal ileum. The association between the
transport of magnesium across the intestinal decreased absorption of B12 and PPI use was
lumen.40,41 shown in short-term studies but the effect was
not consistent in patients with long-term PPI
In 2011, the US FDA issued a drug safety com- use.15,17 Based on the current state of knowledge
munication regarding the potential association that PPIs do not completely inhibit acid secre-
between PPI use and hypomagnesemia.42 A 2014 tion, and the generally sufficient physiologic
meta-analysis by Park and colleagues including reserves of B12 in the general population, routine
nine studies examining the relationship between checks of B12 levels with PPI usage may not be
PPI use and hypomagnesemia found a higher necessary at this time.17
incidence of hypomagnesemia in PPI users than
nonusers.40 In an effort to further explore the Summary. PPI use may be associated with vari-
relationship between PPI use and hypomagne- ous micronutrient deficiencies. The development
semia, Kieboom and colleagues performed a pro- of these deficiencies is likely also highly correlated
spective cohort study including approximately with additional patient risk factors, rather than
9000 patients. They found that PPI use increased being singly attributed to PPI use. Further study

journals.sagepub.com/home/taw 9
Therapeutic Advances in Drug Safety 10

into the long-term effects and clinical implica- between PPI use and an increased risk of all-cause
tions of PPI-related micronutrient deficiency is mortality (hazard ratio 1.25, confidence interval
warranted. 1.23–1.28). The study however did not offer a
clear biological mechanism for this association
but speculated on the potential role of heme oxy-
vii. Bone density, fracture risk and PPIs genase-1, oxidative stress and accelerated senes-
In 2010, the US FDA revised the labeling of cence of human endothelial cells.
PPIs to include a warning about the possible
increased risk of bone fractures with their use.
This warning was retracted from the OTC PPI A word of caution about false alarms and
in 2011 due to insufficient evidence that short- broad conclusions
term, low-dose OTC PPI use was associated Despite a large number of studies, the overall qual-
with bone density changes. These negative ity of evidence for adverse effects of long-term use
effects on bone health may be related to nutri- of PPIs is low to very low. Overzealous conclusions
tional deficiencies. As discussed previously, the based on weak associations can be problematic
increase in gastric pH with the use of PPIs may and maybe partly responsible to the growing alarm
impede calcium absorption.15 There is currently about the prescription of PPIs today. It is worth
insufficient evidence to support routine moni- noting that the ‘guilty by association’ problem fac-
toring of bone mineral density in patients on ing PPIs is not unique; in fact, Hill eloquently
PPIs without other indications for monitoring. addressed issues with observational studies in as
early as 1965.45 He proposed a set of nine ‘aspects
of association’ (strength of association, consist-
viii. Spontaneous bacterial peritonitis ency, specificity, temporality, biological gradient,
The data suggesting an association come primar- plausibility, coherence, experiment, and analogy)
ily from observational studies, where causality to evaluate countless hypothesized relationships
cannot be established. This spontaneous bacterial between exposures and disease outcomes.
peritonitis concern was highlighted especially in
the liver disease and cirrhosis patient cohort.15 Observational studies, no matter how well per-
The biologic plausibility of secondary bacterial formed, may be inherently incapable of accurately
infections in patients with long-term PPI expo- discerning weak associations from null effects due
sure stems from the role of hypochlorhydria and to their susceptibility to systematic errors of bias/
subsequent pH changes in the colon (downstream confounding and other methodological weak-
effect) allowing for bacterial translocation and nesses.46 Also, statistical significance only takes
colonic transmigration leading to gram negative random errors related to sample size into consid-
peritonitis. The current level of evidence does not eration; it ignores systematic errors.
allow for broad recommendations and certainly
does not support withholding PPIs, if indicated, Laine and colleagues recently reported on the dif-
in patients with liver disease. ficulty in eliminating residual bias in observa-
tional studies even with statistical adjustment,
because all confounding factors are not recorded
Long-term PPI use and overall risk of death or even known.16 This may be especially impor-
from all causes tant when effect sizes are small (odds/hazard ratio
Some investigators have evaluated the role of < 2), and so it may not be possible to determine
PPIs and all-cause mortality following prolonged whether the association is valid or the result of
exposure rather than individual organ system dys- residual bias.
function. Xie and colleagues evaluated patient
data from the US Department of Veterans Affairs
as a longitudinal observational cohort study.44 Conclusion
This study used complex statistical analysis We must be cautious about drawing broad con-
including time-dependent propensity score- clusions on the current level of evidence with the
matched cohorts and high-dimensional propen- long-term use of PPIs. This is especially impor-
sity score-adjusted models to reduce the potential tant because the conclusions are overwhelmingly
confounding bias. In the primary cohort of new based on observational studies and meta-analy-
users of acid suppression therapy followed for a ses, which frequently include the same observa-
median of 5.71 years, they showed an association tional studies.

10 journals.sagepub.com/home/taw
M Jaynes and AB Kumar

PPIs have had a profound impact on the out- proton pump inhibitor therapy. Springerplus 2016;
comes of patients with acid peptic disease since 5: 430.
their introduction into clinical practice in the 9. Freedberg DE, Salmasian H, Friedman C, et al.
late 1990s. They continue to have a strong posi- Proton pump inhibitors and risk for recurrent
tive impact when used appropriately for the rec- Clostridium difficile infection among inpatients.
ognized indications. The optimal strategy for Am J Gastroenterol 2013; 108: 1794–1801.
PPI prescription at this time is for patients with 10. McDonald EG, Milligan J, Frenette C, et al.
clear indications, avoiding broad off-label use Continuous proton pump inhibitor therapy and
and to have a prudent time-limited endpoint of the associated risk of recurrent Clostridium
prescription. difficile infection. JAMA Intern Med 2015; 175:
784–791.
Funding
11. Lupse M, Flonta M, Cioara A, et al. Predictors of
The study was supported by the Department of first recurrence in Clostridium difficile-associated
Anesthesiology ( Division of Critical Care) only. disease. A study of 306 patients hospitalized in a
Romanian tertiary referral center. J Gastrointestin
Conflict of interest statement Liver Dis 2013; 22: 397–403.
The authors declare that there is no conflict of
12. Seto CT, Jeraldo P, Orenstein R, et al. Prolonged
interest.
use of a proton pump inhibitor reduces microbial
diversity: implications for Clostridium difficile
susceptibility. Microbiome 2014; 2: 42.

References 13. Haenisch B, von Holt K, Wiese B, et al. Risk


1. Nardino RJ, Vender RJ and Herbert PN. Overuse of dementia in elderly patients with the use of
of acid-suppressive therapy in hospitalized proton pump inhibitors. Eur Arch Psychiatry Clin
patients. Am J Gastroenterol 2000; 95: 3118– Neurosci 2015; 265: 419–428.
3122. 14. Gomm W, von Holt K, Thome F, et al.
2. Batuwitage BT, Kingham JGC, Morgan NE, Association of proton pump inhibitors with risk of
et al. Inappropriate prescribing of proton pump dementia: a pharmacoepidemiological claims data
inhibitors in primary care. Postgrad Med J 2007; analysis. JAMA Neurol 2016; 73: 410–416.
83: 66–68. 15. Freedberg DE, Kim LS and Yang YX. The risks
3. Shaheen NJ, Hansen RA, Morgan DR, et al. and benefits of long-term use of proton pump
The burden of gastrointestinal and liver diseases, inhibitors: expert review and best practice advice
2006. Am J Gastroenterol 2006; 101: 2128–2138. from the american gastroenterological association.
Gastroenterology 2017; 152: 706–715.
4. Barletta JF, Lat I, Micek ST, et al.; Critical Care
Pharmacotherapy Trials Network. Off-label use 16. Laine L and Nagar A. Long-term PPI use:
of gastrointestinal medications in the intensive balancing potential harms and documented
care unit. J Intensive Care Med 2015; 30: 217– benefits. Am J Gastroenterol 2016; 111: 913–915.
225. 17. Savarino V, Dulbecco P and Savarino E. Are
5. Savarino V, Dulbecco P, de Bortoli N, et al. The proton pump inhibitors really so dangerous? Dig
appropriate use of proton pump inhibitors (PPIs): Liver Dis 2016; 48: 851–859.
Need for a reappraisal. Eur J Intern Med 2017; 18. Lambert AA, Lam JO, Paik JJ, et al. Risk of
37: 19–24. community-acquired pneumonia with outpatient
6. Clooney AG, Bernstein CN, Leslie WD, et al. proton-pump inhibitor therapy: a systematic
A comparison of the gut microbiome between review and meta-analysis. PLoS One 2015; 10(6):
long-term users and non-users of proton pump e0128004.
inhibitors. Aliment Pharmacol Ther 2016; 43:
19. Herzig SJ, Howell MD, Ngo LH, et al. Acid-
974–984.
suppressive medication use and the risk for
7. Kwok CS, Arthur AK, Anibueze CI, et al. hospital-acquired pneumonia. JAMA 2009;
Risk of Clostridium difficile infection with acid 301(20): 2120–2128.
suppressing drugs and antibiotics: meta-analysis.
20. MacLaren R, Reynolds PM and Allen RR.
Am J Gastroenterol 2012; 107: 1011–1019.
Histamine-2 receptor antagonists vs proton pump
8. Dos Santos-Schaller O, Boisset S, Seigneurin inhibitors on gastrointestinal tract hemorrhage
A, et al. Recurrence and death after Clostridium and infectious complications in the intensive care
difficile infection: gender-dependant influence of unit. JAMA Intern Med 2014; 174(4): 564–574.

journals.sagepub.com/home/taw 11
Therapeutic Advances in Drug Safety 10

21. Ran L, Khatibi NH, Qin X, et al. Proton 32. Lazarus B, Chen Y, Wilson FP, et al. Proton
pump inhibitor prophylaxis increases the risk pump Inhibitor use and the risk of chronic
of nosocomial pneumonia in patients with an kidney disease. JAMA Intern Med 2016; 176:
intracerebral hemorrhagic stroke. Acta Neurochir 238–246.
Suppl 2011; 111: 435–439.
33. Wijarnpreecha K, Thongprayoon C, Chesdachai
22. Bateman BT, Bykov K, Choudhry NK, et al. S, et al. Associations of proton-pump inhibitors
Type of stress ulcer prophylaxis and risk and h2 receptor antagonists with chronic kidney
of nosocomial pneumonia in cardiac disease: a meta-analysis. Dig Dis Sci 2017; 62:
surgical patients: cohort study. BMJ 2013; 2821–2827.
347: f5416.
34. Li T, Xie Y and Al-Aly Z. The association of
23. Arai N, Nakamizo T, Ihara H, et al. Histamine proton pump inhibitors and chronic kidney
H2-blocker and proton pump inhibitor use disease: cause or confounding? Curr Opin Nephrol
and the risk of pneumonia in acute stroke: a Hypertens 2018; 27: 182–187.
retrospective analysis on susceptible patients.
35. Xie Y, Bowe B, Li T, et al. Proton pump
PLoS One 2017; 12(1): e0169300.
inhibitors and risk of incident CKD and
24. Momosaki R, Yasunaga H, Matsui H, et al. progression to ESRD. J Am Soc Nephrol 2016;
Proton pump inhibitors versus histamine-2 27: 3153–3163.
receptor antagonists and risk of pneumonia in
36. Klatte DCF, Gasparini A, Xu H, et al.
patients with acute stroke. J Stroke Cerebrovasc
Association between proton pump inhibitor use
Dis 2016; 25(5): 1035–1040.
and risk of progression of chronic kidney disease.
25. Juurlink DN, Gomes T, Ko DT, et al. A Gastroenterology 2017; 153: 702–710.
population-based study of the drug interaction
37. Xie Y, Bowe B, Li T, et al. Long-term kidney
between proton pump inhibitors and clopidogrel.
outcomes among users of proton pump inhibitors
CMAJ 2009; 180: 713–718.
without intervening acute kidney injury. Kidney
26. Gilard M, Arnaud B, Cornily JC, et al. Int 2017; 91(6): 1482–1494.
Influence of omeprazole on the antiplatelet
38. Cheungpasitporn W, Thongprayoon C,
action of clopidogrel associated with aspirin: the
Kittanamongkolchai W, et al. Proton pump
randomized, double-blind OCLA (Omeprazole
inhibitors linked to hypomagnesemia: a
CLopidogrel Aspirin) study. J Am Coll Cardiol
systematic review and meta-analysis of
2008; 51: 256–260.
observational studies. Ren Fail 2015; 37(7):
27. Ho PM, Maddox TM, Wang L, et al. Risk of 1237–1241.
adverse outcomes associated with concomitant
39. Kieboom BC, Kiefte-de Jong JC, Eijgelsheim
use of clopidogrel and proton pump inhibitors
M, et al. Proton pump inhibitors and
following acute coronary syndrome. JAMA 2009;
hypomagnesemia in the general population: a
301: 937–944.
population-based cohort study. Am J Kidney Dis
28. Bhatt DL, Cryer BL, Contant CF, et al. 2015; 66(5): 775–782.
Clopidogrel with or without omeprazole in
40. Park CH, Kim EH, Roh YH, et al. The
coronary artery disease. N Engl J Med 2010; 363:
association between the use of proton pump
1909–1917.
inhibitors and the risk of hypomagnesemia: a
29. Ray WA, Murray KT, Griffin MR, et al. systematic review and meta-analysis. PLoS One
Outcomes with concurrent use of clopidogrel 2014; 9(11): e112558.
and proton-pump inhibitors: a cohort study. Ann
41. Famularo G, Gasbarrone L and Minisola G.
Intern Med 2010; 152: 337–345.
Hypomagnesemia and proton-pump inhibitors.
30. Fontes-Carvalho R and Albuquerque A. Expert Opin Drug Saf 2013; 12(5): 709–716.
Clopidogrel–proton pump inhibitors drug
42. FDA Safety Announcement, https://www.fda.gov/
interaction: implications to clinical practice. Rev
drugs/drugsafety/ucm245011.htm (accessed 3
Port Cardiol 2010; 29: 1555–1567.
February 2011).
31. Niu Q, Wang Z, Zhang Y, et al. Combination
43. Mikolasevic I, Milic S, Stimac D, et al. Is there
use of clopidogrel and proton pump inhibitors
a relationship between hypomagnesemia and
increases major adverse cardiovascular events
proton-pump inhibitors in patients on chronic
in patients with coronary artery disease: a
hemodialysis? Eur J Intern Med 2016; 30: 99–103.
meta-analysis. J Cardiovasc Pharmacol Ther.
Epub ahead of print 20 August 2016. DOI: 44. Xie Y, Bowe B, Li T, et al. Risk of death among
10.1177/1074248416663647. users of proton pump inhibitors: a longitudinal

12 journals.sagepub.com/home/taw
M Jaynes and AB Kumar

observational cohort study of United States with proton pump inhibitor use by level of
veterans. BMJ Open 2017; 7(6): e015735. antibiotic exposure. Pharmacoepidemiol Drug Saf
2011; 20(10): 1035–1042.
45. Hill AB. The environment and disease:
association or causation? Proc R Soc Med 1965; 53. Gordon D, Young LR, Reddy S, et al. Incidence
58: 295–300. of Clostridium difficile infection in patients
receiving high-risk antibiotics with or without a
46. Vaezi MF, Yang YX and Howden CW. proton pump inhibitor. J Hosp Infect 2016; 92(2):
Complications of proton pump inhibitor therapy. 173–177.
Gastroenterology 2017.
54. Linsky A, Gupta K, Lawler EV, et al. Proton
47. Arriola V, Tischendorf J, Musuuza J, pump inhibitors and risk for recurrent
et al. Assessing the risk of hospital-acquired Clostridium difficile infection. Arch Intern Med
Clostridium difficile infection with proton pump 2010; 170(9): 772–778.
inhibitor use: a meta-analysis. Infect Control Hosp
55. Kim JW, Lee KL, Jeong JB, et al. Proton pump
Epidemiol 2016; 37(12): 1408–1417.
inhibitors as a risk factor for recurrence of
48. Deshpande A, Pant C, Pasupuleti V, et al. Clostridium-difficile-associated diarrhea. World
Association between proton pump inhibitor journal of gastroenterology 2010; 16(28): 3573–
therapy and Clostridium difficile infection in 3577.
a meta-analysis. Clinical gastroenterology and
56. Cadle RM, Mansouri MD, Logan N, et al.
hepatology : the official clinical practice journal of
Association of proton-pump inhibitors with
the American Gastroenterological Association 2012;
outcomes in Clostridium difficile colitis.
10(3): 225–233.
Am J Health Syst Pharm 2007; 64(22):
49. Roughead EE, Chan EW, Choi NK, et al. Proton 2359–2363.
pump inhibitors and risk of Clostridium difficile
57. Alshamsi F, Belley-Cote E, Cook D, et al.
infection: a multi-country study using sequence
Efficacy and safety of proton pump inhibitors for
symmetry analysis. Expert Opin Drug Saf 2016;
stress ulcer prophylaxis in critically ill patients: a
15(12): 1589–1595.
systematic review and meta-analysis of randomized
50. Ro Y, Eun CS, Kim HS, et al. Risk of Clostridium trials. Crit Care 2016; 20(1): 120.
difficile infection with the use of a proton pump
58. Khorvash F, Abbasi S, Meidani M, et al. The
inhibitor for stress ulcer prophylaxis in critically ill
comparison between proton pump inhibitors and
patients. Gut Liver 2016, 10(4): 581–586.
sucralfate in incidence of ventilator associated
51. Barletta JF and Sclar DA. Proton pump pneumonia in critically ill patients. Adv Biomed
inhibitors increase the risk for hospital-acquired Res 2014; 3: 52.
Clostridium difficile infection in critically ill
59. Arora P, Gupta A, Golzy M, et al. Proton pump Visit SAGE journals online
patients. Crit Care 2014; 18(6): 714. journals.sagepub.com/
inhibitors are associated with increased risk of
home/taw
52. Stevens V, Dumyati G, Brown J, et al. development of chronic kidney disease. BMC
Differential risk of Clostridium difficile infection Nephrol 2016; 17(1): 112. SAGE journals

journals.sagepub.com/home/taw 13

You might also like