Background: The coronavirus infectious disease 2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) has been a huge challenge, affecting directly or indirectly the human race worldwide. Many COVID-19-related risk factors have been identified. Various drugs, including proton pump inhibitors (PPIs), have been claimed to increase the risk of acquiring or affecting the outcome of COVID-19 prior to the availability of vaccines. The aim of this review was to summarize the uncertain role of PPI use on the SARS-CoV-2 infection and COVID-19 clinical course, including both some well-established and presumptive indications to these potent inhibitors of gastric acid secretion, before vaccinations against COVID-19 were implemented on a massive scale. Summary: Although iatrogenic hypochlorhydria alters human microbiota and impairs the nonspecific innate immunity, increasing the likelihood of gastrointestinal and pulmonary infections, published data on the relationship between SARS-CoV-2 positivity and clinical manifestations of COVID-19 are quite inconclusive. On the contrary, the beneficial role of PPI use in the prevention of the upper gastrointestinal bleeding, particularly in at-risk patients receiving dual antiplatelet treatment and/or concomitant anticoagulation, constituting a significant proportion of COVID-19 patients, has been well-documented. Furthermore, PPIs have been marked as a potential antiviral remedy, just like many already existing repurposed drugs, that merit further studies in search of successful anti-CO­VID-19 pharmacotherapy. Key Messages: The controversial role of PPIs on the SARS-CoV-2 infection and COVID-19 severity propounded in the past should not prevent the use of these potent inhibitors of gastric acid secretion in well-established clinical indications.

Severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) is a novel, rapidly spreading infectious pathogen, whereas coronavirus infectious disease 2019 (COVID-19) is a clinical manifestation of a symptomatic SARS-CoV-2 infection. COVID-19 has been declared a pandemic by the WHO on March 11, 2020. As of late February 2021, there have been more than 111 million cases of diagnosed SARS-CoV-2 infection and almost 2.5 million deaths reported worldwide. Since its outbreak, the number of people affected by the pandemic has been increasing.

SARS-CoV-2 utilizes angiotensin-converting enzyme type 2 (ACE2) as an entry receptor to infect mainly alveolar epithelial cells [1]. Typical manifestations of COVID-19 range from mild to a severe systemic disease characterized by an acute respiratory distress syndrome (ARDS), shock, and multiorgan dysfunction. While approximately 80% of symptomatic patients report mild or moderate illness, the rest develop profound hypoxemia, necessitating prompt in-hospital oxygenation. The highest mortality is reported in COVID-19 patients admitted to an intensive care unit (ICU).

Morbidity and mortality increase with age, disproportionately affecting those with preexisting conditions, for example, obesity, chronic kidney disease, diabetes mellitus, cardiovascular disease, and chronic lung disease [2]. Other COVID-19-related risk factors, including commonly used medications, are still being identified.

Proton pump inhibitors (PPIs) suppress gastric acid production by irreversibly binding to the active site of the H+/K+ ATPase of parietal cells. PPIs have been linked, among others, to gut dysbiosis, increased risk of gastrointestinal (GI) infections [3], community-acquired pneumonia [4, 5], and a higher mortality rate due to cardiovascular or chronic kidney disease [6]. PPIs raise gastric pH, which might facilitate the SARS-CoV-2 infection, whereas bacterial pneumonia or GI-derived infections may complicate the clinical course of COVID-19.

Some early retrospective studies [7‒9] raised concerns about the use of PPIs with respect to SARS-CoV-2 positivity and/or severe clinical course and fatal outcome of COVID-19. However, these preliminary observations have been much criticized [10] and have not been completely confirmed [11, 12].

Moreover, symptomatic SARS-CoV-2 infection increases the demand for nonsteroidal anti-inflammatory drugs (NSAIDs) that can cause GI damage. In addition, prophylactic or therapeutic anticoagulation due to the high prevalence of thromboembolic complications in COVID-19 patients [13, 14] may promote GI bleeding (GIB) from existing mucosal lesions. PPIs used according to the guidelines as gastroprotection decrease the risk of the upper GIB, need for urgent GI endoscopy, GIB-related hospital admissions and surgery, the prevalence of rebleeding, and GIB-related mortality rate in at-risk patients taking antiplatelet therapy, anticoagulants, or NSAIDs, including those admitted to the ICU (stress ulcer prophylaxis), that is a significant proportion of COVID-19 inpatients. Therefore, the beneficial effects of PPIs in selected SARS-CoV-2 positive individuals should not be neglected.

Despite some progress made in the symptomatic management of COVID-19, no drug with proven specific anti-SARS-CoV-2 activity has been discovered until February 2021. In addition, the initial enthusiasm on the first registered anti-COVID-19 vaccines has been soon replaced by the efficacy concerns with respect to the emerging SARS-CoV-2 mutants. Therefore, the need for successful antiviral pharmacotherapy still remains the major goal of research [15].

Interestingly, PPIs have been marked as a potential remedy for COVID-19 within the frame of the idea to use already available drugs with the ascertained mechanism of action, clinical indications, and an acceptable safety profile to prevent/treat SARS-CoV-2 infection in the lack of specific antiviral chemotherapeutics [16]. It has been assumed that PPIs-induced pH change in cells as well as their presumed immunoregulatory, anti-inflammatory, antioxidative, antiproliferative, and antifibrotic effects [17, 18] justify the hypothesis that PPIs may be of value in COVID-19 prophylaxis and/or treatment.

The aim of this review was to summarize early evidence on the questionable impact of PPI use on the SARS-CoV-2 infection and COVID-19 clinical course based on a literature review of studies published in English till the end of February 2021 (key search words and collocations: SARS-CoV-2, COVID-19, proton pump inhibitors, adverse effects, clinical outcome) found in the PubMed database. The results of this search (information available in the selected studies) and evaluation are presented and discussed in a few sections reflecting strong belief in the Janus-faced nature of PPIs found in scientific publications.

PPIs – efficient, affordable, and universally available drugs – have been listed among the most commonly used and overused medications, often prescribed and/or taken long-term without a clear indication or any medical supervision [19]. PPIs are generally considered as safe and well-tolerated treatment of gastric acid-related disorders [20]. Nevertheless, they have been associated with several adverse effects [21], including GI infections [3] and pneumonia [4, 5, 22, 23]. However, the association between PPI use and bacterial pneumonia might have been overestimated [24, 25]. An increased susceptibility to bacterial GI infections, gut dysbiosis, and small intestinal bacterial overgrowth as adverse effects of PPIs are supported by randomized trials and meta-analyses [26‒30]. In addition, Vilcu et al. [31] found that continuous use of PPIs is associated with an increased risk for an acute viral enteric infection during periods of high endemic prevalence. It has been documented that increased gastric pH may be a risk factor for rotavirus [32], norovirus [33], and influenza virus [34] infections.

Zhou et al. [35] demonstrated that human intestinal epithelial cells were highly susceptible to the Middle East respiratory syndrome coronavirus infection, and the mice pretreated with pantoprazole in order to increase the Middle East respiratory syndrome coronavirus viability in the stomach preceding intragastric administration of the virus showed more prominent inflammation and epithelial damage in the small intestine than a control group. Of interest, with the progression of the enteric infection, live viruses appeared in the lungs. Previous data revealed that the normal pH of a healthy stomach (pH ≤ 3), unlike less acidic pH achieved with PPIs therapy, inactivates SARS-CoV-1 [36]. Similarly, SARS-CoV-2 may be destroyed in pH levels of 1.5–3, but it can survive in the presence of PPI-induced hypochlorhydria. Thus, iatrogenic hypochlorhydria, which impairs the innate immune system defense mechanisms, may facilitate invasion and replication of SARS-CoV-2 within enterocytes [37].

ACE2 receptors are widely expressed in the GI tract. Once SARS-CoV-2 colonizes the GI tract, it can lead to gastroenterocolitis [38]. There is evidence for the presence of infectious virus in the faeces [39, 40]. Negative nasopharyngeal tests were found in the presence of positive fecal samples collected simultaneously [39], suggesting that fecal-oral transmission cannot be completely excluded [41]. In addition, there is concern that the SARS-CoV-2 could spread beyond the GI tract, seed infection, or promote inflammation in the other organs, including the respiratory tract via a “gut-lung axis” [42].

PPIs may also modulate the immune response of neutrophils, NK cells, and cytotoxic T lymphocytes [5]. PPI use stimulates overgrowth of gut microbiota [26], but diminishes GI microbial diversity, that renders colonization of enteropathogens [43]. The relationship between GI dysbiosis and COVID-19 severity has been raised [42, 44, 45]. PPIs-induced dysbiosis might increase the likelihood of developing enteric infections, facilitate bacterial translocation, and sepsis which could complicate the clinical course of COVID-19. The intestinal lesions may promote the release of endotoxins and microbial metabolites that subsequently trigger inflammation and extreme proinflammatory cytokine release, present in some COVID-19 inpatients, which, in turn, can lead to multiorgan dysfunction and a fatal outcome. In addition, in case of micro-aspiration episodes, the bacteria will colonize the airways. The secondary pulmonary infection increases the probability of a severe clinical course of COVID-19, ARDS, and death.

Furthermore, taking PPIs has been related with a small excess of mortality, including cardiovascular and chronic kidney disease [6]. PPIs can inhibit the activity of dimethylarginine dimethylaminohydrolase and, secondarily, nitric oxide synthase, thus promoting inflammation, thrombosis, and the development of cardiovascular diseases. The idiosyncratic effect of PPIs on the kidneys may also lead to renal injury and dyselectrolytemia [46]. Noteworthy, there is evidence that omeprazole increases the expression of ACE2 in renal tissues [47].

The COVID-19 pandemic has raised safety concerns about the influence of PPI use on the SARS-CoV-2 positivity, disease severity, and clinical outcome of COVID-19. Despite the theoretical background, several respective studies on the association between the PPI use and COVID-19 have produced conflicting results (Table 1).

An internet survey study of a voluntary subject self-reporting of COVID-19 test results, demographics, GI symptoms, and PPIs and histamine type 2 receptor antagonists (H2RAs) use disclosed that PPIs used up to once daily or twice daily increased odds (aOR) (2.15 and 3.67, respectively) for self-reporting a positive COVID-19 test when compared with PPIs nonusers [7]. These alarming results soon gained the general public interest but have been later either questioned, mainly due to a selection bias [4, 10, 48, 49, 50], or not confirmed by other researchers [8, 11, 51, 52].

It is worth to emphasize that OR >3 falls into “zone of interest,” whereas 0.33 < OR < 3 falls within the “zone of potential bias.” In addition, it is recommended to use the Bradford Hill criteria [53] to assess presumed associations between PPI use and an outcome.

In Almario et al. [7] study, information on the number of participants tested for COVID-19 is missing. It is unclear whether the control group consists of participants who were tested and were COVID-19 negative or were a mixture of tested and untested subjects. If PPI nonusers received fewer tests, it could result in fewer positive tests. Moreover, subjects taking more potent gastric acid suppressants by prescription may be more involved into the healthcare system and, therefore, more likely to get tested and hence, could report more positive results. The question is, if the study demonstrates an association between PPI use and the risk of SARS-CoV-2 infection or the likelihood of being interested in SARS-CoV-2 testing or being tested for COVID-19.

Hadi et al. [50] claimed that Almario et al. [7] included participants who were not currently using PPIs as their reference group, so they were unable to determine the association between past PPI use and the odds of reporting a positive COVID-19 test. In addition, data regarding relevant comorbidities (cardiovascular disease, diabetes mellitus, chronic respiratory, or kidney disease) are absent.

Hajifathalian and Katz [10] noted that the COVID-19 positive respondents have a substantially different demographics than the overall study participants. Almario et al. [7] explain anomalies in data by suggesting that participants might be dishonest about their education or income or might not remember their correct diagnosis, but, on the contrary, postulate that it is unlikely for respondents to be wrong about COVID-19 diagnosis or PPI use. In addition, the reason for PPI use, dose, or compliance is unknown. It is not clear why once daily PPI use for more than 6 months should have a significantly smaller effect than once daily PPI use of less than 6 months. PPIs twice daily for less than 6 months was no different than once daily PPIs, whereas the same dose for more than 6 months had a significantly larger effect than once daily dosing.

Contrarily to the Almario et al. study, Tarlow et al. [52] found OR of SARS-CoV-2 positivity with PPI use of 0.48 compared with PPIs nonusers. Interestingly, in a retrospective case-control study on 179 elderly patients, PPI users were 2.3 times less likely to be infected by SARS-CoV-2 and develop COVID-19 than PPIs nonusers [11].

Some studies showed the improved clinical outcome (a low risk of mechanical ventilation or death over a 30-day period) in hospitalized COVID-19 patients who use famotidine but not PPIs [54]. However, these initial observations were later criticized (retrospective analysis, inpatient cohort, small number of patients, inclusion of the individuals without a proven COVID-19 diagnosis, lack of a control group, any difference in the outcome between oral vs. IV administration of famotidine not addressed in the study, concomitant drugs not considered, laboratory parameters, serving as surrogate markers for disease severity not adjusted for in the analysis).

In a more complex study of 952 COVID-19 patients (5.4% with severe disease) including 26 covariates (age, sex, comorbidities, other medications, laboratory parameters), no significant association between severe COVID-19 disease and the use of famotidine or PPIs with laboratory parameters (leukocyte count >11 × 109/L and LDH >280 U/L as independent markers associated with severe COVID-19) was disclosed [12].

In the study including 9,469 participants, neither PPI nor H2RA use was associated with the risk of SARS-CoV-2 infection or death in-patients with COVID-19 [51]. The pooled OR of testing positive for SARS-CoV-2 associated with PPI use (previous and current) and current PPI use only was 1.06 and 1.03, respectively.

These results are consistent with a study from South Korea with reference to SARS-CoV-2 positivity, however, Korean patients taking PPIs (mainly short-term use of PPIs for less than a month starting within the previous 30 days) were at increased risk for severe clinical outcomes of COVID-19 (admission to the ICU, the need for invasive ventilation, or death) [8]. The relationship with past PPI use remained insignificant. The researcher admitted that: (a) PPIs prescriptions do not reflect actual drug exposure (probable noncompliance); (b) the results of SARS-CoV-2 test may be false negative/positive; (c) genetic polymorphisms of CYP2C19 and gastric H+ K+-ATPase may be a potential confounder; (d) smoking and BMI have not been included in the final analysis.

In the other study, exposure to PPIs prior to hospitalization was associated with worse clinical outcomes, including mortality in COVID-19 patients, regardless of the presence of cardiovascular diseases [9]. Despite some heterogeneity of the adverse clinical outcomes utilized in clinical trials, the meta-analysis of five studies with a total of 37,372 patients [55] disclosed increased odds for a severe or fatal course of COVID-19 (3 studies) and secondary infections (2 other studies) with the use of PPIs in COVID-19 patients compared with PPIs nonusers (pooled OR of 1.46 and 2.91, respectively). However, the increased risk for severe or fatal course of COVID-19 was small.

In the Cunye et al. [56] analysis (online publication without a peer-review) of eight studies with more than 268,683 subjects, PPI use did not influence the risk of SARS-CoV-2 infection or mortality risk of COVID-19 patients, but the risk of severe disease (OR 1.54) and secondary infection (OR 4.33) was increased. However, the authors stress the need of more high-quality studies to further clarify the relationship.

In Hariyanto et al. [57] meta-analysis, a total of 5884 COVID-19 patients (six studies) showed that PPI use was associated with an increased risk of severe COVID-19 (RR 1.35) and mortality from COVID-19 (RR 1.72), but (a) confounders such as patients’ age and comorbidities (not included in the analysis) may affect the relationship; (b) the information about the type, dose, duration, frequency, and compliance for PPI use is missing in the majority of analyzed studies; and (c) the RR values lower than 2.0 decrease the clinical meaning of the association; (d) most studies are of limited scientific value.

Meta-analysis by Li et al. [58] showed that PPI users were more likely to have severe outcomes of COVID-19 than PPI nonusers, with a pooled OR of 1.67. Past use of PPIs was not associated with increased susceptibility to SARS-CoV-2 infection (OR 0.85) or severe outcomes of COVID-19 (OR 1.03).

With reference to potential confounders, obesity, especially with BMI >40 kg/m2 is an established risk factor for severe COVID-19 and death. GERD is more prevalent in obese patients, and it is a risk factor for micro-aspiration that might worsen the clinical outcome of COVID-19, just like the majority of obesity-related comorbitities, rather than PPI use itself.

Noteworthy, according to the Spain predictors of ICU admission study, COVID-19 patients suffering from asthma or allergic diseases were often the previous PPI users [59]. On the contrary, Savarino et al. [60] addressed the issue of the risk of COVID-19 in eosinophilic GI diseases treated with PPIs, glucocorticosteroides, or biologics depending on the location and initial response to pharmacotherapy. Despite the remarkable intake of acid suppressants (67.7%), no case of COVID-19 was diagnosed.

GI symptoms are common in COVID-19 [38] and may be the first clinical manifestation of SARS-CoV-2 infection [60, 61]. GIB has been reported in 4–13.7% of COVID-19 patients with GI symptoms in association with mucosal lesions in the esophagus, peptic ulcers, and SARS-CoV-2 detection in the cytoplasm of gastric, duodenal, and rectal epithelial cells [62, 63]. Of note, the first documented emergency surgery in a SARS-CoV-2-positive patient was performed due to perforated duodenal ulcer [64]. In addition, SARS-CoV-2 may affect the liver and negatively impact coagulation [65].

The following factors are considered in the pathophysiology of GIB in COVID-19 patients: (a) direct damage to the GI epithelium by SARS-CoV-2; (b) COVID-19-associated coagulopathy; and (c) pharmacotherapy including NSAIDs, glucocorticosteroids, IL-6 inhibitors [66], antiplatelet drugs, and prophylactic/therapeutic anticoagulation in COVID-19 patients that promotes GIB from existing mucosal lesions.

GIB can be a life-threatening complication to a severely ill COVID-19 patient, especially the elderly with comorbidities. Overt GIB in a COVID-19 patient necessitating endoscopy is also challenging to the endoscopic unit staff (the risk of airborne transmission of SARS-CoV-2), especially with reference to the upper GI tract examinations [67, 68].

Risk factors for the upper GIB in patients initiated on anticoagulants include older age, a prior history of GIB, chronic renal failure, H. pylori infection, concomitant use of antiplatelet drugs, and preexisting GI tract lesions [69]. The risk of GIB increases with the number of risk factors [70]. COVID-19 and anti-IL-6 therapy might be regarded as adjunctive risk factors influencing risk stratification of GIB in hospitalized patients [71].

The incidence of hospitalization for upper GIB is lower among patients who receive PPIs [72]. PPIs promote ulcer healing, prevent ulcer recurrence, and diminish the risk of GIB in high-risk populations. Several studies have demonstrated an overall benefit with the use of prophylactic PPIs, particularly in patients taking dual antiplatelet therapy and/or anticoagulation [73]. The meta-analysis of Mo et al. [74] confirmed that PPIs are also effective in prevention of low-dose aspirin-associated upper GI ulcers and bleeding. Moayyedi et al. [75] demonstrated that pantoprazole reduced overt GIB in patients receiving low anticoagulation and/or aspirin by approximately 50%, confirming the protective effect of PPIs.

NSAIDs, often used in COVID-19, have long been associated with an increased risk of GIB, particularly in older patients (>65 years), in-patients with previous complicated ulcers, and in those receiving high-dose NSAIDs or concomitant anticoagulation, aspirin, or glucocorticosteroids [76]. If NSAIDs cannot be stopped while receiving anticoagulation, PPIs may decrease the risk of the upper GIB in this population [77, 78].

For higher-risk critically ill patients, PPIs reduce the prevalence of GIB compared with no prophylaxis [79]. For patients at low risk, the reduction in bleeding may be unimportant. Therefore, the use of PPIs for the prevention of stress ulcers in non-critically ill patients admitted to the hospital is currently not recommended [80]. On the contrary, the use of PPIs for the prevention of stress ulcers in critical care patients has been supported by evidence and is recommended for high-risk patients [81]. Meta-analysis of 19 clinical trials showed that PPIs were more effective than H2RAs in reducing the risk of clinically important and overt GIB without an increased risk of pneumonia, ICU, length of stay, or death [82]. No effect of continuous daily treatment with PPIs versus H2RAs on risk of ventilator-associated events in critically ill patients who underwent mechanical ventilation for at least 5 days has been found [83].

Undoubtedly, careful risk stratification of the upper GIB, including medications and past medical history, should be performed in COVID-19 patients. If necessary, concomitant treatment with PPIs is strongly recommended [84‒87].

The COVID-19 pandemic raised, yet again, controversy about PPIs safety but also hope for their prophylactic and therapeutic potential against SARS-CoV-2. With the respect to the risk and severity of SARS-CoV-2 infection, there is no consensus regarding whether the use of PPIs might be beneficial, harmful, or indifferent. Although an increased pH in the stomach may, theoretically, be advantageous to the SARS-CoV-2 virus, the clinical significance of this hypothesis still remains unclear.

The results of clinical studies (often of an inferior scientific quality, including no precise data as to the PPIs dose and/or duration of treatment) on the risk of SARS-CoV-2 infection, disease severity, and mortality rate in COVID-19 patients exposed to PPIs are highly inconclusive. Numerous confounding factors could bias the interpretation of available data regarding the correlation between PPI use and SARS-CoV-2 infection and disease severity.

In fact, precipitous entering “negative public relations” of PPIs into general public may result in unnecessary anxiety and needless, potentially harmful PPI withdrawal in patients who definitely should continue the treatment. It ought to be stressed that both outpatient and in-hospital PPI use may prevent the upper GIB and the need for challenging endoscopic procedures at the time of airborne pandemic. The unproven potential of PPIs’ harmful effects and safety concerns referring to SARS-CoV-2 infection should not prevent proper prophylactic or therapeutic PPI use.

Any physician prescribing PPIs should always assess the individual benefit-risk ratio. Patients with a proven indication for PPI use should continue pharmacotherapy in the lowest effective dose. Whenever possible, prophylaxis of severe clinical course of COVID-19, for example, vaccinations or weight loss in obese patients, should be recommended.

The authors have no conflicts of interest to declare.

No funding in the preparation of data or the manuscript applies to the manuscript.

Dorota Ksiądzyna contributed to the manuscript as follows: conception and design of the work, acquisition, analysis, and interpretation of the data, drafting of the manuscript, and final approval of the preliminary manuscript to be submitted. Adam Szeląg contributed to the manuscript as follows: acquisition, analysis, interpretation and critical revision of the data, and final approval of the submitted version.

1.
Zhou
P
,
Yang
XL
,
Wang
XG
,
Hu
B
,
Zhang
L
,
Zhang
W
,
.
A pneumonia outbreak associated with a new coronavirus of probable bat origin
.
Nature
.
2020
;
579
(
7798
):
270
3
.
2.
Stokes
EK
,
Zambrano
LD
,
Anderson
KN
,
Marder
EP
,
Raz
KM
,
El Burai Felix
S
,
.
Coronavirus disease 2019 case surveillance: United States, January 22–May 30, 2020
.
MMWR Morb Mortal Wkly Rep
.
2020
;
69
(
24
):
759
65
.
3.
Vaezi
MF
,
Yang
YX
,
Howden
CW
.
Complications of proton pump inhibitor therapy
.
Gastroenterology
.
2017
;
153
(
1
):
35
48
.
4.
Lambert
AA
,
Lam
JO
,
Paik
JJ
,
Ugarte-Gil
C
,
Drummond
MB
,
Crowell
TA
.
Risk of community-acquired pneumonia with outpatient proton-pump inhibitor therapy: a systematic review and meta-analysis
.
PLoS One
.
2015
;
10
(
6
):
e0128004
.
5.
Laheij
RJF
,
Sturkenboom
MCJM
,
Hassing
R-J
,
Dieleman
J
,
Stricker
BH
,
Jansen
JB
.
Risk of community-acquired pneumonia and use of gastric acid-suppressive drugs
.
JAMA
.
2004
;
292
(
16
):
1955
60
.
6.
Xie
Y
,
Bowe
B
,
Li
T
,
Xian
H
,
Yan
Y
,
Al-Aly
Z
.
Estimates of all cause mortality and cause specific mortality associated with proton pump inhibitors among US veterans: cohort study
.
BMJ
.
2019
;
365
:
l1580
.
7.
Almario
CV
,
Chey
WD
,
Spiegel
BMR
.
Increased risk of COVID-19 among users of proton pump inhibitors
.
Am J Gastroenterol
.
2020
;
115
(
10
):
1707
15
.
8.
Lee
SW
,
Ha
EK
,
Yeniova
,
Moon
SY
,
Kim
SY
,
Koh
HY
,
.
Severe clinical outcomes of COVID-19 associated with proton pump inhibitors: a nationwide cohort study with propensity score matching
.
Gut
.
2021
;
70
(
1
):
76
84
.
9.
Ramachandran
P
,
Perisetti
A
,
Gajendran
M
,
Jean-Louis
F
,
Bansal
P
,
Dwivedi
AK
,
.
Pre-hospitalization proton pump inhibitor use and clinical outcomes in COVID-19
.
Eur J Gastroenterol Hepatol
.
2022
;
34
(
2
):
137
41
.
10.
Hajifathalian
K
,
Katz
PO
.
Regarding “increased risk of COVID-19 in patients taking proton pump inhibitors”
.
Am J Gastroenterol
.
2020
;
115
(
11
):
1918
9
.
11.
Blanc
F
,
Waechter
C
,
Vogel
T
,
Schorr
B
,
Demuynck
C
,
Martin-Hunyadi
C
,
.
Interest of proton pump inhibitors in reducing the occurrence of COVID-19: a case-control study
.
Preprints
.
2020
:
2020050016
.
12.
Cheung
KS
,
Hung
IF
,
Leung
WK
.
Association between famotidine use and COVID-19 severity in Hong Kong: a territory-wide study
.
Gastroenterology
.
2021
;
160
(
5
):
1898
9
.
13.
Cui
S
,
Chen
S
,
Li
X
,
Liu
S
,
Wang
F
.
Prevalence of venous thromboembolism in patients with severe novel coronavirus pneumonia
.
J Thromb Haemost
.
2020
;
18
(
6
):
1421
4
.
14.
Middeldorp
S
,
Coppens
M
,
Haaps
TF
,
Foppen
M
,
Vlaar
AP
,
Müller
MCA
,
.
Incidence of venous thromboembolism in hospitalized patients with COVID-19
.
J Thromb Haemost
.
2020
;
18
(
8
):
1995
2002
.
15.
Pagliano
P
,
Scarpati
G
,
Sellitto
C
,
Conti
V
,
Spera
AM
,
Ascione
T
,
.
Experimental pharmacotherapy for COVID-19: the latest advances
.
J Exp Pharmacol
.
2021
;
13
:
1
13
.
16.
Ray
A
,
Sharma
S
,
Sadasivam
B
.
The potential therapeutic role of proton pump inhibitors in COVID-19: hypotheses based on existing evidences
.
Drug Res
.
2020
;
70
(
10
):
484
8
.
17.
De Milito
A
,
Iessi
E
,
Logozzi
M
,
Lozupone
F
,
Spada
M
,
Marino
ML
,
.
Proton pump inhibitors induce apoptosis of human B-cell tumors through a caspase-independent mechanism involving reactive oxygen species
.
Cancer Res
.
2007
;
67
(
11
):
5408
17
.
18.
Ghebre
YT
,
Raghu
G
.
Idiopathic pulmonary fibrosis: novel concepts of proton pump inhibitors as antifibrotic drugs
.
Am J Respir Crit Care Med
.
2016
;
193
(
12
):
1345
52
.
19.
Ksiądzyna
D
,
Szeląg
A
,
Paradowski
L
.
Overuse of proton pump inhibitors
.
Pol Arch Intern Med
.
2015
;
125
(
4
):
289
98
.
20.
Brunner
G
,
Athmann
C
,
Schneider
A
.
Long term, open-label trial: safety and efficacy of continuous maintenance treatment with pantoprazole for up to 15 years in severe acid-peptic disease
.
Aliment Pharmacol Ther
.
2012
;
36
(
1
):
37
47
.
21.
Johnson
DA
,
Katz
PO
,
Armstrong
D
,
Cohen
H
,
Delaney
BC
,
Howden
CW
,
.
The safety of appropriate use of over-the-counter proton pump inhibitors: an evidence-based review and Delphi consensus
.
Drugs
.
2017
;
77
(
5
):
547
61
.
22.
Hsu
WT
,
Lai
CC
,
Wang
YH
,
Tseng
PH
,
Wang
K
,
Wang
CY
,
.
Risk of pneumonia in patients with gastroesophageal reflux disease: a population-based cohort study
.
PLoS One
.
2017
;
12
(
8
):
e0183808
.
23.
Sarkar
M
,
Hennessy
S
,
Yang
YX
.
Proton-pump inhibitor use and the risk for community-acquired pneumonia
.
Ann Intern Med
.
2008
;
149
(
6
):
391
8
.
24.
Ma
C
,
Shaheen
AA
,
Congly
SE
,
Andrews
CN
,
Moayyedi
P
,
Forbes
N
.
Interpreting reported risks associated with use of proton pump inhibitors: residual confounding in a 10-year analysis of national ambulatory data
.
Gastroenterology
.
2020
;
158
(
3
):
780
2.e3
.
25.
Wang
CH
,
Li
CH
,
Hsieh
R
,
Fan
CY
,
Hsu
TC
,
Chang
WC
,
.
Proton pump inhibitors therapy and the risk of pneumonia: a systematic review and meta-analysis of randomized controlled trials and observational studies
.
Expert Opin Drug Saf
.
2019
;
18
(
3
):
163
72
.
26.
Lo
WK
,
Chan
WW
.
Proton pump inhibitor use and the risk of small intestinal bacterial overgrowth: a meta-analysis
.
Clin Gastroenterol Hepatol
.
2013
;
11
(
5
):
483
90
.
27.
Bavishi
C
,
Dupont
HL
.
Systematic review: the use of proton pump inhibitors and increased susceptibility to enteric infection
.
Aliment Pharmacol Ther
.
2011
;
34
(
11–12
):
1269
81
.
28.
Leonard
J
,
Marshall
JK
,
Moayyedi
P
.
Systematic review of the risk of enteric infection in patients taking acid suppression
.
Am J Gastroenterol
.
2007
;
102
(
9
):
2047
56
.
29.
Moayyedi
P
,
Eikelboom
JW
,
Bosch
J
,
Connolly
SJ
,
Dyal
L
,
Shestakovska
O
,
.
Safety of proton pump inhibitors based on a large, multi-year, randomized trial of patients receiving rivaroxaban or aspirin
.
Gastroenterology
.
2019
;
157
(
3
):
682
91.e2
.
30.
Trifan
A
,
Stanciu
C
,
Girleanu
I
,
Stoica
OC
,
Singeap
AM
,
Maxim
R
,
.
Proton pump inhibitors therapy and risk of Clostridium difficile infection: systematic review and meta-analysis
.
World J Gastroenterol
.
2017
;
23
(
35
):
6500
15
.
31.
Vilcu
AM
,
Sabatte
L
,
Blanchon
T
,
Souty
C
,
Maravic
M
,
Lemaitre
M
,
.
Association between acute gastroenteritis and continuous use of proton pump inhibitors during winter periods of highest circulation of enteric viruses
.
JAMA Netw Open
.
2019
;
2
(
11
):
e1916205
.
32.
Weiss
C
,
Clark
HF
.
Rapid inactivation of rotaviruses by exposure to acid buffer or acidic gastric juice
.
J Gen Virol
.
1985
;
66
(
12
):
2725
30
.
33.
Prag
C
,
Prag
M
,
Fredlund
H
.
Proton pump inhibitors as a risk factor for norovirus infection
.
Epidemiol Infect
.
2017
;
145
(
8
):
1617
23
.
34.
Hayase
Y
,
Tobita
K
,
Sato
H
.
Detection of type B influenza virus genes from biopsied gastric mucosa
.
J Gastroenterol
.
2002
;
37
(
2
):
101
5
.
35.
Zhou
J
,
Li
C
,
Zhao
G
,
Chu
H
,
Wang
D
,
Yan
HHN
,
.
Human intestinal tract serves as an alternative infection route for middle East respiratory syndrome coronavirus
.
Sci Adv
.
2017
;
3
(
11
):
eaao4966
.
36.
Darnell
ME
,
Subbarao
K
,
Feinstone
SM
,
Taylor
DR
.
Inactivation of the coronavirus that induces severe acute respiratory syndrome, SARS-CoV
.
J Virol Methods
.
2004
;
121
(
1
):
85
91
.
37.
Lamers
MM
,
Beumer
J
,
van der Vaart
J
,
Knoops
K
,
Puschhof
J
,
Breugem
TI
,
.
SARS-CoV-2 productively infects human gut enterocytes
.
Science
.
2020
;
369
(
6499
):
50
4
.
38.
Pan
L
,
Mu
M
,
Yang
P
,
Sun
Y
,
Wang
R
,
Yan
J
,
.
Clinical characteristics of COVID-19 patients with digestive symptoms in Hubei, China: a descriptive, cross-sectional, multicenter study
.
Am J Gastroenterol
.
2020
;
115
(
5
):
766
73
.
39.
Xiao
F
,
Sun
J
,
Xu
Y
,
Li
F
,
Huang
X
,
Li
H
,
.
Infectious SARS-CoV-2 in feces of patient with severe COVID-19
.
Emerg Infect Dis
.
2020
;
26
(
8
):
1920
2
.
40.
Wang
W
,
Xu
Y
,
Gao
R
,
Lu
R
,
Han
K
,
Wu
G
,
.
Detection of SARS-CoV-2 in different types of clinical specimens
.
JAMA
.
2020
;
323
(
18
):
1843
4
.
41.
Dibner
JJ
.
Direct COVID-19 infection of enterocytes: the role of hypochlorhydria
.
Am J Infect Control
.
2021
;
49
(
3
):
385
6
.
42.
Uzzan
M
,
Corcos
O
,
Martin
JC
,
Treton
X
,
Bouhnik
Y
.
Why is SARS-CoV-2 infection more severe in obese men? The gut lymphatics: lung axis hypothesis
.
Med Hypotheses
.
2020
;
144
:
110023
.
43.
Seto
CT
,
Jeraldo
P
,
Orenstein
R
,
Chia
N
,
DiBaise
JK
.
Prolonged use of a proton pump inhibitor reduces microbial diversity: implications for Clostridium difficile susceptibility
.
Microbiome
.
2014
;
2
(
1
):
42
.
44.
Segal
JP
,
Mak
JWY
,
Mullish
BH
,
Alexander
JL
,
Ng
SC
,
Marchesi
JR
.
The gut microbiome: an under-recognised contributor to the COVID-19 pandemic
.
Therap Adv Gastroenterol
.
2020
;
13
:
1756284820974914
.
45.
Dhar
D
,
Mohanty
A
.
Gut microbiota and Covid-19- possible link and implications
.
Virus Res
.
2020
;
285
:
198018
.
46.
Makunts
T
,
Cohen
IV
,
Awdishu
L
,
Abagyan
R
.
Analysis of postmarketing safety data for proton-pump inhibitors reveals increased propensity for renal injury, electrolyte abnormalities, and nephrolithiasis
.
Sci Rep
.
2019
;
9
(
1
):
2282
.
47.
Saheb Sharif-Askari
N
,
Saheb Sharif-Askari
F
,
Alabed
M
,
Tayoun
AA
,
Loney
T
,
Uddin
M
,
.
Effect of common medications on the expression of SARS-CoV-2 entry receptors in kidney tissue
.
Clin Transl Sci
.
2020
;
13
(
6
):
1048
54
.
48.
Aby
ES
,
Rodin
H
,
Debes
JD
.
Proton pump inhibitors and mortality in individuals with COVID-19
.
Am J Gastroenterol
.
2020
;
115
(
11
):
1918
.
49.
Boushey
H
,
Gonda
I
.
Comment on increased risk of COVID-19 among users of proton pump inhibitors
.
Am J Gastroenterol
.
2020
;
115
(
11
):
1921
.
50.
Hadi
YB
,
Naqvi
SF
,
Kupec
JT
.
Risk of COVID-19 in patients taking proton pump inhibitors
.
Am J Gastroenterol
.
2020
;
115
(
11
):
1919
20
.
51.
Fan
X
,
Liu
Z
,
Miyata
T
,
Dasarathy
S
,
Rotroff
DM
,
Wu
X
,
.
Effect of acid suppressants on the risk of COVID-19: a propensity score-matched study using UK Biobank
.
Gastroenterology
.
2021
;
160
(
1
):
455
8.e5
.
52.
Tarlow
B
,
Gubatan
J
,
Khan
MA
,
Cholankeril
G
.
Are proton pump inhibitors contributing to SARS-COV-2 infection
.
Am J Gastroenterol
.
2020
;
115
(
11
):
1920
1
.
53.
Hill
AB
.
The environment and disease: association or causation
.
Proc R Soc Med
.
1965
;
58
(
5
):
295
300
.
54.
Freedberg
DE
,
Conigliaro
J
,
Wang
TC
,
Tracey
KJ
,
Callahan
MV
,
Abrams
JA
,
.
Famotidine use is associated with improved clinical outcomes in hospitalized COVID-19 patients: a propensity score matched retrospective cohort study
.
Gastroenterology
.
2020
;
159
(
3
):
1129
31.e3
.
55.
Kow
CS
,
Hasan
SS
.
Use of proton pump inhibitors and risk of adverse clinical outcomes from COVID-19: a meta analysis
.
J Intern Med
.
2021
;
289
(
1
):
125
8
.
56.
Cunye
Y
,
Yue
C
,
Chenyu
S
,
Mubashir
AA
,
Chandur
B
,
Ce
C
,
.
Will proton pump inhibitors lead to a higher risk of COVID-19 infection and progression to severe disease? A meta-analysis
.
medRxiv
.
2020
:
1101
.
57.
Hariyanto
TI
,
Prasetya
IB
,
Kurniawan
A
.
Proton pump inhibitor use is associated with increased risk of severity and mortality from coronavirus disease 2019 (COVID-19) infection
.
Dig Liver Dis
.
2020
;
52
(
12
):
1410
2
.
58.
Li
GF
,
An
XX
,
Yu
Y
,
Jiao
LR
,
Canarutto
D
,
Yu
G
,
.
Do proton pump inhibitors influence SARS-CoV-2 related outcomes? A meta-analysis
.
Gut
.
2020
;
70
(
9
):
1806
8
.
59.
García-Menaya
JM
,
Cordobés-Durán
C
,
Rangel-Mayoral
JF
,
García-Martín
E
,
Agúndez
JAG
.
Outcomes and laboratory and clinical findings of asthma and allergic patients admitted with Covid-19 in a Spanish University Hospital
.
Front Pharmacol
.
2020
;
11
:
570721
.
60.
Savarino
E
,
Lorenzon
G
,
Ghisa
M
,
Laserra
G
,
Barberio
B
,
Maniero
D
,
.
Lack of complications in patients with eosinophilic gastrointestinal diseases during SARS-CoV-2 outbreak
.
J Allergy Clin Immunol Pract
.
2020
;
8
:
2790
2.e1
.
61.
Pazgan-Simon
M
,
Rorat
M
,
Buczyńska
I
,
Zińczuk
A
,
Simon
K
.
Gastrointestinal symptoms as the first, atypical indication of severe acute respiratory syndrome coronavirus 2 infection
.
Pol Arch Intern Med
.
2020
;
130
(
4
):
338
9
.
62.
Tian
Y
,
Rong
L
,
Nian
W
,
He
Y
.
Review article: gastrointestinal features in COVID-19 and the possibility of faecal transmission
.
Aliment Pharmacol Ther
.
2020
;
51
(
9
):
843
51
.
63.
Xiao
F
,
Tang
M
,
Zheng
X
,
Liu
Y
,
Li
X
,
Shan
H
.
Evidence for gastrointestinal infection of SARS-CoV-2
.
Gastroenterology
.
2020
;
158
(
6
):
1831
3.e3
.
64.
He
L
,
Zhao
W
,
Zhou
W
,
Pang
P
,
Liao
Y
,
Liu
J
.
An emergency surgery in severe case infected by COVID-19 with perforated duodenal bulb ulcer
.
Ann Surg
.
2020
;
272
(
1
):
e35
7
.
65.
Kukla
M
,
Skonieczna-Żydecka
K
,
Kotfis
K
,
Maciejewska
D
,
Łoniewski
I
,
Lara
LF
,
.
COVID-19, MERS and SARS with concomitant liver injury-systematic review of the existing literature
.
J Clin Med
.
2020
;
9
(
5
):
1420
.
66.
Curtis
JR
,
Lanas
A
,
John
A
,
Johnson
DA
,
Schulman
KL
.
Factors associated with gastrointestinal perforation in a cohort of patients with rheumatoid arthritis
.
Arthritis Care Res
.
2012
;
64
(
12
):
1819
28
.
67.
Marek
T
,
Reguła
J
,
Kamiński
MF
,
Rydzewska
G
.
Polish Society of Gastroenterology and gastroenterology national consultant guidelines concerning digestive endoscopy during COVID-19 pandemic
.
Gastroenterol Klin
.
2020
;
12
:
1
6
.
68.
Barkun
AN
,
Almadi
M
,
Kuipers
EJ
,
Laine
L
,
Sung
J
,
Tse
F
,
.
Management of nonvariceal upper gastrointestinal bleeding: guideline recommendations from the international consensus group
.
Ann Intern Med
.
2019
;
171
(
11
):
805
22
.
69.
Vaduganathan
M
,
Pareek
M
,
Bhatt
DL
.
Gastroprotection with proton-pump inhibitors in high-risk cardiovascular patients: who to target and for how long
.
Expert Opin Drug Saf
.
2016
;
15
(
11
):
1451
3
.
70.
Abraham
NS
,
Hlatky
MA
,
Antman
EM
,
Bhatt
DL
,
Bjorkman
DJ
,
Clark
CB
,
.
ACCF/ACG/AHA 2010 expert consensus document on the concomitant use of proton pump inhibitors and thienopyridines: a focused update of the ACCF/ACG/AHA 2008 expert consensus document on reducing the gastrointestinal risks of antiplatelet therapy and NSAID use: a report of the American College of Cardiology foundation task force on expert consensus documents
.
Circulation
.
2010
;
122
(
24
):
2619
33
.
71.
Agnes
A
,
La Greca
A
,
Tirelli
F
,
Papa
V
.
Duodenal perforation in a SARS-CoV-2-positive patient with negative PCR results for SARS-CoV-2 in the peritoneal fluid
.
Eur Rev Med Pharmacol Sci
.
2020
;
24
(
23
):
12516
21
.
72.
Ray
WA
,
Chung
CP
,
Murray
KT
,
Smalley
WE
,
Daugherty
JR
,
Dupont
WD
,
.
Association of oral anticoagulants and proton pump inhibitor cotherapy with hospitalization for upper gastrointestinal tract bleeding
.
JAMA
.
2018
;
320
(
21
):
2221
30
.
73.
Ray
WA
,
Chung
CP
,
Murray
KT
,
Smalley
WE
,
Daugherty
JR
,
Dupont
WD
,
.
Association of proton pump inhibitors with reduced risk of warfarin-related serious upper gastrointestinal bleeding
.
Gastroenterology
.
2016
;
151
(
6
):
1105
12.e10
.
74.
Mo
C
,
Sun
G
,
Lu
ML
,
Zhang
L
,
Wang
YZ
,
Sun
X
,
.
Proton pump inhibitors in prevention of low-dose aspirin-associated upper gastrointestinal injuries
.
World J Gastroenterol
.
2015
;
21
(
17
):
5382
92
.
75.
Moayyedi
P
,
Eikelboom
JW
,
Bosch
J
,
Connolly
SJ
,
Dyal
L
,
Shestakovska
O
,
.
Pantoprazole to prevent gastroduodenal events in patients receiving rivaroxaban and/or aspirin in a randomized, double-blind, placebo-controlled trial
.
Gastroenterology
.
2019
;
157
(
2
):
403
12.e5
.
76.
Lanza
FL
,
Chan
FK
,
Quigley
EM
;
Practice Parameters Committee of the American College of Gastroenterology
.
Guidelines for prevention of NSAID-related ulcer complications
.
Am J Gastroenterol
.
2009
;
104
(
3
):
728
38
.
77.
Regula
J
,
Butruk
E
,
Dekkers
CPM
,
de Boer
SY
,
Raps
D
,
Simon
L
,
.
Prevention of NSAID-associated gastrointestinal lesions: a comparison study pantoprazole versus omeprazole
.
Am J Gastroenterol
.
2006
;
101
(
8
):
1747
55
.
78.
Scheiman
JM
,
Yeomans
ND
,
Talley
NJ
,
Vakil
N
,
Chan
FKL
,
Tulassay
Z
,
.
Prevention of ulcers by esomeprazole in at-risk patients using non-selective NSAIDs and COX-2 inhibitors
.
Am J Gastroenterol
.
2006
;
101
(
4
):
701
10
.
79.
Wang
Y
,
Ye
Z
,
Ge
L
,
Siemieniuk
RAC
,
Wang
X
,
Wang
Y
,
.
Efficacy and safety of gastrointestinal bleeding prophylaxis in critically ill patients: systematic review and network meta-analysis
.
BMJ
.
2020
;
368
:
l6744
.
80.
Grube
RRA
,
May
DB
.
Stress ulcer prophylaxis in hospitalized patients not in intensive care units
.
Am J Health Syst Pharm
.
2007
;
64
(
13
):
1396
400
.
81.
Alhazzani
W
,
Alshamsi
F
,
Belley-Cote
E
,
Heels-Ansdell
D
,
Brignardello-Petersen
R
,
Alquraini
M
,
.
Efficacy and safety of stress ulcer prophylaxis in critically ill patients: a network meta-analysis of randomized trials
.
Intensive Care Med
.
2018
;
44
:
1
11
.
82.
Alshamsi
F
,
Belley-Cote
E
,
Cook
D
,
Almenawer
SA
,
Alqahtani
Z
,
Perri
D
,
.
Efficacy and safety of proton pump inhibitors for stress ulcer prophylaxis in critically ill patients: a systematic review and meta-analysis of randomized trials
.
Crit Care
.
2016
;
20
(
1
):
120
.
83.
Li
X
,
Klompas
M
,
Menchaca
JT
,
Young
JG
.
Effects of daily treatment with acid suppressants for stress ulcer prophylaxis on risk of ventilator-associated events
.
Infect Control Hosp Epidemiol
.
2020
;
41
(
2
):
187
93
.
84.
Cavaliere
K
,
Levine
C
,
Wander
P
,
Sejpal
DV
,
Trindade
AJ
.
Management of upper GI bleeding in patients with COVID-19 pneumonia
.
Gastrointest Endosc
.
2020
;
92
(
2
):
454
5
.
85.
Gadiparthi
C
,
Perisetti
A
,
Sayana
H
,
Tharian
B
,
Inamdar
S
,
Korman
A
.
Gastrointestinal bleeding in patients with severe SARS-CoV-2
.
Am J Gastroenterol
.
2020
;
115
(
8
):
1283
5
.
86.
Patel
P
,
Sengupta
N
.
PPIs and beyond: a framework for managing anticoagulation-related gastrointestinal bleeding in the era of COVID-19
.
Dig Dis Sci
.
2020
;
65
(
8
):
2181
6
.
87.
Hunt
RH
,
East
JE
,
Lanas
A
,
Malfertheiner
P
,
Satsangi
J
,
Scarpignato
C
,
.
COVID-19 and gastrointestinal disease: implications for the gastroenterologist
.
Dig Dis
.
2021
;
39
(
2
):
119
39
.