Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Sep 3.
Published in final edited form as: JAMA. 2012 Jan 25;307(4):373–381. doi: 10.1001/jama.2011.2035

Randomized Clinical Trial of Lansoprazole for Poorly Controlled Asthma in Children

The American Lung Association Asthma Clinical Research Centers*

Janet T Holbrook, Robert A Wise, Benjamin D Gold, Kathryn Blake, Ellen D Brown, Mario Castro, Allen J Dozor, John Lima, John G Mastronarde, Marianna Sockrider, W Gerald Teague
PMCID: PMC4153372  NIHMSID: NIHMS512330  PMID: 22274684

Abstract

Context

Asymptomatic gastroesophageal reflux (GER) is prevalent in children with asthma. It is not known whether treatment of GER with a proton-pump inhibitor (PPI) improves asthma control.

Objective

To determine whether lansoprazole is effective in reducing asthma symptoms in children without overt GER.

Design, Setting, and Patients

A multicenter, randomized, masked, placebo controlled, parallel clinical trial comparing lansoprazole to placebo in children with poor asthma control on inhaled corticosteroid treatment conducted at 18 academic clinical centers. Participants were followed for 24 weeks. A subgroup had an esophageal pH study before randomization.

Intervention

Children received either lansoprazole (15 mg daily < 30 kg; 30 mg ≥ 30 kg) or placebo, 1:1 allocation ratio.

Main outcome

The primary outcome was the change in Asthma Control score (ACQ, range from 0 to 6). Secondary outcomes included lung function measures, asthma-related quality of life and acute episodes of poor asthma control.

Results

306 children were enrolled from April 2011 to August 2010, the median age was 11. The mean change (95% confidence interval (CI)) in the ACQ score was −0.1 (−0.2, 0.1) and −0.2 (−0.4, −0.1) units for the lansoprazole and placebo groups, respectively (P=0.12). There were no detectable treatment differences in secondary outcomes (mean (95% CI) for FEV1(0.00 (−0.08, 0.08)), asthma quality of life (−0.1 (−0.4, 0.1) or episodes of poor asthma control, hazard ratio of 1.18 (95% CI 0.91, 1.53). Among the 115 children with esophageal pH studies, the prevalence of GER was 43%. In the subgroup with a positive pH study, no treatment effect for lansoprazole versus placebo was observed for any asthma outcome. Children treated with lansoprazole reported more upper respiratory infections (63% vs 49%, P=0.02), sore throats (52% vs 39%, P=0.02), and bronchitis (7% vs 2%, P=0.05).

Conclusion

Among children with poorly controlled asthma without symptoms of GER who were using inhaled corticosteroids, the addition of lansoprazole, as compared to placebo, did not improve symptoms nor lung function but was associated with increased adverse events.

Introduction

Asthma and gastroesophageal reflux (GER) are both common disorders in children and symptoms of GER are frequently reported among children with asthma.1,2,3,4 GER identified by esophageal pH monitoring often presents with respiratory symptoms5,6,7 and frequently occurs in children without characteristic gastrointestinal symptoms.1,8,9 Untreated GER has been postulated to be a cause of inadequate asthma control in children despite inhaled corticosteroid (ICS) treatment. Additionally, expert panels have indicated that the utility of testing and treating children with refractory asthma symptoms for asymptomatic GER has been inadequately studied.3,10 Hence, it is of clinical importance to determine whether anti-reflux therapy, the most common being proton pump inhibitors (PPI), improves the control of asthma in children.

In adults, it appears that PPIs may be helpful for asthma in some patients who manifest reflux symptoms, but are not helpful for those with asymptomatic GER. 11,12,13 An open-label prospective study evaluated 44 children with moderate, persistent asthma and GERD who showed clinical improvement in asthma symptoms after 1 year of treatment with a esomeprazole and metoclopramide. Children who continued the combination treatment for six months had fewer asthma exacerbations than those who switched to ranitidine.14

we conducted a trial to study the efficacy of PPIs in children with poor asthma control without symptomatic GER. The primary hypothesis was that children with symptomatic asthma taking inhaled corticosteroids have improved asthma control. with lansoprazole treatment compared to placebo. We also investigated whether asymptomatic GER as identified by pH probe testing would identify children who respond to PPI treatment.

Methods

Study Design

The Study of Acid Reflux in Children with Asthma (SARCA) was a randomized (allocation ratio 1:1) double masked, placebo-controlled, parallel clinical trial designed to evaluate the effectiveness of lansoprazole (Prevacid ®, TAKEDA Pharmaceuticals North America) for treatment of asthma in children (clinicaltrial.gov NCT00442013). The study was conducted at 19 American Lung Association's Asthma Clinical Research Center (ALA-ACRC) sites from to April 2007 to April 2011. The study was approved by the institutional review board at each center, and the legal guardians signed informed consent statement and the participants signed assent statement according to local regulatory policy.

Children were randomly assigned to receive either lansoprazole (15 mg daily for children < 30 kg; 30 mg daily for children ≥ 30 kg) or a matching placebo. A permuted block treatment assignment schedule stratified by clinical center was used. Treatment assignments were masked throughout the study. After the screening and randomization visits, participants returned to the clinical centers for assessments every 4 weeks for 24 weeks. Throughout the study children kept daily diaries to record morning peak expiratory flow, asthma symptoms, nocturnal awakenings, use of short-acting β-agonists (excluding routine use before exercise), oral corticosteroid use, and unscheduled health care visits for asthma symptoms. Ambulatory esophageal pH monitoring prior to randomization was conducted in a subgroup of children who agreed to the procedure at 13 clinical centers with the capability of doing pH probe studies for the study. Participants were paid, on average, ($ ) for each study visit.

Participant Selection

Participants were between 6 and 17 years of age, had physician-diagnosed asthma, and labile airways function defined as a 12% or greater increase in FEV1 post-bronchodilator, a methacholine challenge PC20 of < 16 mg/mL, or a positive exercise-bronchoprovocation test of a 20% or greater fall in FEV1 post-exercise demonstrated within the prior 12 months. Participants were treated with inhaled corticosteroids (≥ 176 mcg per day fluticasone equivalents) and had no change in controller therapies for at least 8 weeks prior to enrollment. Poor asthma control was defined as any one of the following: use of short-acting β-agonists for asthma symptoms two or more times per week; nocturnal awakenings with asthma symptoms more than once per week over the month before enrollment; two or more emergency department visits, unscheduled physician visits, prednisone courses, or hospitalizations for asthma in the prior year; or a score of 1.25 or higher on the ACQ at the screening visit. Recruits were excluded from the study if they exhibited any of the following: symptoms of GER requiring treatment, treatment with a PPI or other reflux medications (other than occasional oral antacids); history of anti-reflux surgery or a previous trachea-esophageal fistula repair; an FEV1 less than 60% predicted; history of neonatal respiratory distress or prematurity less than 33 weeks gestational age; or other major chronic illnesses. Children with a known sensitivity or intolerance to lansoprazole or aspartame were not enrolled. Other exclusion criteria were non-adherence, (below 80% completion of daily diaries during run-in), inability to take study medications, perform baseline measurements or be contacted by telephone, or pregnancy. Participant characteristics related to race, cigarette or cigar smoke exposure, past asthma treatments and asthma triggers were collected via interview. Children were followed between 2 to 8 weeks before randomization and were eligible for randomization if they completed 80% of daily diary cards, maintained a stable dose of inhaled corticosteroids, had a percent predicted FEV1 of 60% or greater and were not pregnant were eligible for randomization. After eligibility was confirmed, children were assigned to the next random treatment assignment via computer.

Outcome Measures

The primary outcome for the trial was the change in Asthma Control Questionnaire (ACQ) at the 24 week visit. 16 The ACQ integrates indicators of asthma control including use of bronchodilators, nocturnal symptoms, cough, activity level, and pulmonary function and has a range of 0 to 6 (higher values indicate worse asthma control). A 0.5 change in ACQ reflects a clinically meaningful difference in asthma control.

Secondary outcomes included the rate of acute episodes of poor asthma control (EPACs),14 Asthma Symptom Utility Index (ASUI) score (range 0 to 1.0, ?.? ,15 Asthma Control Test (ACT16/cACT17; range 5-25/0-27, respectively, meaningful difference is 3), asthma-specific quality of life for children (AQLQ; range 1-7, meaningful difference is 0.5) and their caregivers (CAQLQ; range 1-7, meaningful difference is 0.5),18 methacholine PC20,19 spirometry,20 exhaled nitric oxide (eNO), gastrointestinal symptoms (GSAS; range 0-441, meaningful difference not defined) and nocturnal awakenings. We defined two types of EPACs derived from the daily diaries: an EPAC1 was a decrease ≥ 30% in morning peak flow rate from personal best (assessed during run-in) for two consecutive days, or addition of an oral corticosteroid to treat asthma symptoms, or unscheduled contact with a health care provider for asthma symptoms. An EPAC2 also included increased use of short-acting β agonists from baseline (4 or more additional puffs of rescue medication or 2 or more additional nebulizer treatments in one day). Participants were questioned about potential adverse effects of treatment at each visit. Subsequent to an FDA advisory on the risk of bone fractures in adults,21 clinics were asked to review participant records for fractures.

Esophageal pH studies were done prior to randomization at 13 clinical centers according to a standard protocol and centrally reviewed. At least 16 hours of monitoring was required for evaluation. The thresholds used for the definition of pathological GER were: esophageal pH ≤ 4 for 6% or greater of the time for a 6-11 year old, and for 4% or greater of the time for a 12-17 year old.

Sample Size

The planned sample size of 300 participants provided 90% power to detect a 0.6 unit change in the primary outcome, ACQ, assuming a standard deviation of 1.5 with a two-sided type I error rate of 5%. The sample size was inflated by 5% to account for missing data. We estimated 80 to 90% power to detect a difference of 0.85 to 1.0 in ACQ score in the subgroup of participants who underwent pH probes.

Data Analysis

All analyses were conducted according to treatment assignment and all available data were incorporated. Longitudinal models estimated the change from baseline to 6 months in a measurement using generalized estimating equations (GEE) with an unstructured or exchangeable covariance matrix to adjust for repeated measures22 All participants with baseline or any follow-up data were included in the models. Negative binomial regression models were used to evaluate differences in the rate of EPACs;23 participants without diary card data were excluded from these analyses. We planned to evaluate the treatment effect in subgroups defined by pH probe test results; a differential effect of treatment was evaluated by a test of interaction. There was one interim analysis during the trial after approximately 50% of the participants completed follow-up. P-values were two-sided and a P-value ≤0.05 was considered statistically significant; P-value were not adjusted for multiple looks or multiple comparisons. Data were analyzed using SAS (version 9.2) and Stats (version 10).

Results

Recruitment

2,453 children were screened for eligibility; 306 children were randomized, 157 were assigned to receive placebo and 149 were assigned to receive lansoprazole (Figure 1). More than 88% of participants completed the study and 94% of follow-up visits were completed. Self-reported compliance to study treatments was high in both groups, study drug was taken on 90 to 91% of follow-up days.

Figure 1.

Figure 1

CONSORT Diagram

Features of the Study Participants

The demographic and asthma characteristics at baseline were similar in the two treatment groups (Table 1). The children had a median age of 11 years, included more males than females, and 50% of participants were African American. Most participants required an intervention (urgent care, oral prednisone course or frequent use of rescue medication) for asthma symptoms in the year prior to enrollment. Mean baseline FEV1 was 99% of predicted in both treatment groups. Children treated with lansoprazole had greater change in FEV1 in response to a bronchodilator, 10.9% versus 8.5% in the placebo-treated group (P=0.02). Among the subgroup with methacholine provocation results at randomization, bronchial responsiveness was elevated in both groups, but relatively less in the lansoprazole group (PC20 3.4 mg/mL versus 2.5 mg/mL, P=0.04).

Table 1.

Characteristics of Study Participants

Treatment Assignment
Placebo Lansoprazole
N 157 149
Age at randomization – year (mean ± standard deviation) 11 ± 3 11 ± 3
Male (%) 65 58
Race or ethnic group (%)
    White 33 34
    Black 50 50
    Hispanic 11 11
    Other 6 5
pH probe test results
      N completing pH probe test 53 62
      Positive test result (%) 38 47
Exposed to secondhand smoke (%) 18 17
Asthma Characteristics
    Age of asthma onset – year (mean ± standard deviation) 3 ± 3 4 ± 3
    Unscheduled health care for asthma in past year (%) 69 77
    Oral corticosteroids for asthma in past year (%) 62 72
    Use of rescue inhaler 2 or more times per week (%) 75 73
    Daily use of ICS/LABA (%) 60 59
    Daily use of anti-leukotriene (%) 54 55
Inhaled corticosteroid treatment at enrollment
    Fluticasone/salmeterol (%) 52 53
      Fluticasone (%) 22 17
    Budesonide (%) 10 14
    Budesonide/formoterol (%) 6 5
    Beclomethasone (%) 7 5
    Mometasone or triamcinolone (%) 3 6
Asthma questionnaire scores (mean ± standard deviation)
      ACQ (↓score range: 0-6) at screening 1.6 ± 0.8 1.6 ± 0.8
      ACQ (↓score range: 0-6) at randomization 1.2 ± 0.8 1.2 ± 0.8
      ASUI score (↑score range: 0-1) 0.82 ± 0.14 0.82 ± 0.15
      ACT (age 12-17, n=139)) (↑score range: 5-25) 19 ± 4 19 ± 4
      C-ACT (age 6-11, n=164)) (↑score range: 0-27) 20 ± 4 20 ± 4
      Mini-AQLQ score (↑) (score range: 1-7) 5.5 ± 1.1 5.4 ± 1.2
      Caregiver's AQOL (↑) (score range: 1-7) 5.4 ± 1.2 5.6 ± 1.2
GERD Symptom Assessment (↓)(median, interquartile range)
      Median for score (score range: 0-441) 9 (2, 220) 7 (2 ,28)
      Median for number of symptom (range: 0-9) 2 (1, 4) 3 (1, 4)
Pulmonary Function (mean ± standard deviation)
      Pre-bronchodilator FEV1 (% predicted)* 92.4 ± 14.6 91.3 ± 17.1
      Post-bronchodilator FEV1 (% predicted)* 99.7 ± 14.1 99.6 ± 15.4
      Pre-bronchodilator FVC (% predicted)* 101.1 ± 14.6 100.4 ± 13.8
      Post-bronchodilator FVC (% predicted)* 102.9 ± 15.1 103.0 ± 13.2
      Peak flow (% predicted)* 97.3 ± 22.1 92.7 ± 20.0
      %Change in FEV1 after bronchodilator 8.5 ± 10.9 10.9 ± 11.3
      %Change in FVC after bronchodilator 1.5 ± 6.0 3.0 ± 5.2
      Exhaled NO (median, IQR) 35 (14, 59) 26 (10, 48)
      Exhaled breath condensate pH 6.1 ± 1.0 6.2 ± 1.0
    N with PC20 108 107
    PC20 (mg/mL) 2.5 ± 3.7 3.4 ± 4.0
Other Conditions (%)
    GERD 1 2
    Eczema 39 48
    Sinusitis 28 30
    Rhinitis 51 68
    Food Allergies 27 26
    Allergies worsen asthma 79 81

Abbreviations: ICS=inhaled corticosteroid, LABA=long-acting beta-agonist, PC20=provocative dose of methacholine to reduce percent predicted FEV1 by 20%; ACQ=Asthma Control Questionnaire; ACT=Asthma Control Test; C-ACT=Child's Asthma Control Test; AQLQ=Asthma Quality of Life Questionnaire; GERD=Gastro-esophageal reflux disease; FEV1=force expiratory volume in 1 second; FVC=forced vital capacity

*

predicted values of Hankinson et al.

↓ lower value indicates less severe asthma/GERD; ↑ higher value indicates less severe asthma

Number with data on pH probe results or PC20 at baseline

GER Status

152 participants underwent 24 hour esophageal pH monitoring studies. Of these 115 were adequate for interpretation. Forty-nine participants (43%) had abnormal esophageal acid exposure as assessed by frequent acid reflux events, prolonged acid reflux episodes (longer than 5 minutes), or overall 24 hour esophageal acid relux (reflux index). GSAS scores (mean (95% CI) were not different between patients with normal versus abnormal pH probe studies (15 (10,20) vs 20 (12, 27)), respectively.

Effects of Treatment on Asthma Outcomes

At the screening visit the mean ACQ was high and consistent with poor asthma control in both treatment groups (1.6 for both groups, P=0.96). Between the screening and randomization visits, the ACQ fell in both groups, indicating improvement (Table 1) but there was no difference between treatment groups; the mean difference (95% CI) for lansoprazole-placebo was 0.2 (−0.0,0.3), Table 2, Figure 2). There was no significant treatment effect for any of the secondary indices of asthma control: ASUI, ACT for adolescents 12-17, cACT for children ages 6-11, AQLQ, and the cAQLQ for caregivers (Table 2). Likewise there was no significant treatment effect on lung function, including pre-bronchodilator FEV1 or FVC over the six- month follow-up (Table 2). The magnitude of bronchial hyperresponsiveness increased slightly (decrease in methacholine PC20) in the lansoprazole treatment group from baseline to month 6 of the study but there was no treatment effect. There was no significant treatment effect on EPAC 1 or EPAC 2 rates (Table 3). EPAC type 1 excluded use of rescue medications because we were concerned that heartburn symptoms could be mistaken for asthma symptoms and results in increased rescue medication use.

Table 2.

Questionnaire scores and lung function measures at 24 weeks of follow-up

Value at 24 weeks Change from baseline Treatment Effect P-value
Placebo Lansoprazole Placebo Lansoprazole Diff Change Lansoprazole-Placebo Treatment Effect* Interaction with GER
N --------------------------Mean (95% Confidence Interval)------------------------
Asthma Control Questionnaires
    Asthma Control Questionnaire 263 1.0 (0.9, 1.1) 1.1 (0.9, 1.2) −0.2 (−0.4, 0.1) −0.1 (−0.2, 0.1) 0.2 (−0.0, 0.3) 0.12 0.15
    Asthma Symptom Utility Index 266 0.88 (0.86, 0.90) 0.86 (0.83,0.88) 0.06 (0.03, 0.09) 0.03 (0.01, 0.06) −0.03 (−0.07, 0.01) 0.14 0.38
    Asthma Control Test 12-17 years 126 20.2 (19.4, 21.2) 20.2 (19.3, 21.1) 1.6 (0.5, 2.7) 1.0 (0.2, 1.7) −0.6 (−2.0, 0.7) 0.37 0.86
    Asthma Control Test 6-11 years 126 21.9 (21.1, 22.8) 20.9 (19.9, 21.9) 2.0 (1.0, 2.9) 1.2 (0.3, 2.1) −0.7 (−2.0, 0.6) 0.29 0.29
Quality of Life Questionnaires
    Pediatric QoL 262 6.0 (5.9, 6.0) 5.8 (5.7, 6.0) 0.5 (0.3, 0.7) 0.4 (0.3, 0.6) −0.1 (−0.3, 0.2) 0.65 0.48
    Caregiver QoL 261 6.0 (5.8, 6.2) 6.0 (5.8, 6.2) 0.5 (0.3, 0.7) 0.4 (0.2, 0.6) −0.1 (−0.4, 0.1) 0.28 0.46
Pulmonary function
    Pre-bronchodilator FEV1 (L) 259 2.3 (2.1, 2.4) 2.2 (2.1, 2.4) 0.03 (−0.03, 0.08) 0.03 (−0.03, 0.09) 0.00 (−0.08, 0.08) 0.99 0.27
    FEV1 bronchodilator Response (%) 120 10.8 (7.8, 14.0) 10.7 (7.0, 14.5) 2.2 (−1.2, 5.7) −0.1 (−4.0, 3.9) −2.3 (−7.6, 2.9) 0.38 0.002
    Pre-bronchodilator FVC (L) 259 2.9 (2.7, 3.1) 2.9 (2.7, 3.0) 0.07 (0.02, 0.11) 0.05 (0.01, 0.10) −0.01 (−0.08, 0.05) 0.69 0.22
    PC20 (mg/mL) 119 2.5 (1.7, 3.4) 2.6 (2.09, 3.4) 0.0 (−0.9, 1.0) 0.1 (−0.8, 1.1) −0.8 (−2.1, 0.4) 0.20 0.80
*

P-values for treatment effect based on GEE models including all data points

For ACQ and PC20 a negative number favors lansoprazole, for all other measures a positive treatment effect favors lansoprazole

P-values for a differential treatment effect in patients with positive pH probe status in subgroup of patients (n=115) with those data

Figure 2.

Figure 2

Change in asthma control score in children with poor asthma control treated with lansoprazole and placebo.

Table 3.

Respiratory infections

Treatment Group
Placebo Lansoprazole P-value*
N 150 147
N (%)
Upper respiratory 74 (49) 93 (63) 0.02
Sore throat 59 (39) 77 (52) 0.02
Strep throat 11 (7) 6 (4) 0.23
Bronchitis 3 (2) 10 (7) 0.04
Pneumonia 5 (3) 4 (3) 0.76
Otis media 10 (7) 12 (8) 0.62
Acute sinusitis 17 (11) 16 (11) 0.90
*

Based on Chi-square test or Fisher's Exact test

Sub-analysis of Children with Abnormal Esophageal pH Studies

Among the 115 children with adequate 24 hour esophageal monitoring studies, 42% (n= 49) were positive for GER; 38% who received placebo had GER versus 47% who received lansoprazole (P=0.33). In a sub-analysis of children with GER, there was no significant effect of lansoprazole treatment on any of the study outcomes, including the ACQ, ASUI, ACT, or cACT, asthma related quality of life, EPAC rates, lung function, or bronchial hyperresponsiveness (Supplemental tables 1 and 2).

Adverse Events

Ten participants in the lansoprazole group and 9 in the placebo group had one or more serious adverse events. The most common serious adverse event in both groups was asthma exacerbations (15 of 25 reports). Among all reported adverse events, treatment with lansoprazole was associated with a greater prevalence of upper respiratory infections, sore throats, and episodes of bronchitis (Table 4). Activity-related bone fractures in children treated with lansoprazole versus placebo (6 vs 1, P=0.06) was not different. The children experiencing fractures were between the ages of 7 and 14 and all had been receiving inhaled corticosteroids throughout the trial; 2 (one in each group) had also received a course of oral prednisone during the trial. One fracture, in the lansoprazole group, occurred on the day the patient was randomized. The others occurred after 2 months (1), 5 months (3) and 6 months (2) of follow-up.

Discussion

In children with poorly controlled asthma lansoprazole treatment had no effect on asthma control measures. This was the case even though GER was prevalent (42% as evidenced by positive esophageal pH tests) in the study sample. In a sub-analysis of the effects of lansoprazole restricted to participants with documented GER, we found no effect of lansoprazole on any of the aforementioned indicators of asthma control. Although GER is prevalent in children with poorly controlled asthma, the results of this clinical trial are uniformly negative regarding the benefit of acid suppression therapy on symptom relief, lung function, airways reactivity or quality of life.

Previous clinical trials in children with symptomatic GER and respiratory symptoms have not shown a clear benefit of PPI treatment on respiratory outcomes. In one uncontrolled study of children, 85% with demonstrable GER, lansoprazole improved asthma symptoms in a majority24 or improved airways reactivity.25 However, a placebo-controlled trial of omeprazole in 38 children with asthma and GER showed no significant effect of omeprazole on asthma outcomes.26 These results, in conjunction with ours, indicate that PPI therapy for poorly controlled asthma is not warranted.

While our results are robust there are hypotheses about the role of GER and asthma we did not address. We focused on patients without symptomatic GER because these children do not have an independent indication for PPI treatment. There are preliminary data that suggest non-acid reflux may result in respiratory symptoms.27 The possible role of non-acid reflux in worsening asthma control is unclear, and larger, well controlled studies are needed to validate this hypothesis. It is also possible that acid reflux was a cause of asthma symptoms but the FDA-approved dose of lansoprazole that we used may not have achieved adequate acid suppression in all children.

Our study does not refute the possibility that GER may trigger chronic cough in children.28 In a recent study in non-asthmatic children with chronic cough, episodes of GER preceded cough in 22 of 26 patients.29 In adults, proximal reflux has been reported to be associated with worse asthma control and health-related quality of life despite lack of physiologic impairment or decrease in asthma symptoms.30

We confirmed previous data suggesting a high prevalence of GER in asthmatic children, and our results do not support routine esophageal pH testing to identify children who respond to PPIs, nor do they support trials of PPI for poorly controlled asthma.

Our study raises important questions about adverse effects of lansoprazole treatment of children with asthma. The lansoprazole group had significantly more self-reported episodes of upper respiratory symptoms, including sore throats, and bronchitis. In a clinical trial of lansoprazole vs placebo in infants, lansoprazole was associated with more lower respiratory infections.31 In additions, PPIs use has been associated with increased risk of community-acquired pneumonia in adults and children,32,33,34,35 which is thought to be related to a reduction in the host defense against bacterial colonization imparted by low gastric acid.36,37,38

The use of proton pump inhibitors in children has increased dramatically in the past decade, going from about 875,000 prescriptions in 2002 to 2.6 million in 2009.39 The increase, especially among infants, along with safety signals in adults, has resulted in two FDA advisory board reviews of existing data related to the use of proton-pump inhibitors in children and infants within the past two years.40,41 At the time of those reviews, there were two reports of fractures related to proton pump inhibitors in the FDA Adverse Event Reporting System. However, both committees recognized that there were limited data mostly from short-term studies and the Pediatric committee voted to receive updated reports on the safety of PPI use in children.40 Our results reinforce the need for continued study of PPI safety in children, especially those with other risk factors, like inhaled corticosteroid use, which may already place them at higher risk for adverse events.

In conclusion the results of our study indicate that PPI treatment of children with poorly controlled asthma without symptomatic GER was not an effective therapy for asthma and there may be significant safety concerns for long term PPI use in children that warrants further study.

Supplementary Material

01

Acknowledgments

American Lung Association Asthma Clinical Research Centers

Baylor College of Medicine, Houston: N. A. Hanania (principal investigator), M. Sockrider (co-principal investigator), L. Bertrand (principal clinic coordinator), M. Atik, L. Giraldo, B, Flores (coordinators);

Columbia University–New York University Consortium, New York: J. Reibman (principal investigator), E. DiMango, L. Rogers (co-principal investigators), C. Cammarata and K. Carapetyan (clinic coordinators at New York University), J. Sormillon and E. Simpson (clinic coordinators at Columbia University);

Duke University Medical Center, Durham, N.C.: L. Williams (principal investigator), J. Sundy (co-principal investigator), G. Dudek (principal clinic coordinator), R. Newton and A. Dugdale (coordinators);

Emory University School of Medicine, Atlanta: W.G. Teague (principal investigator), Anne Fitzpatrick, Sumita Khatri (co-principal investigators), R. Patel (principal clinic coordinator), J. Peabody, E Hunter, D Whitlock (coordinators);

Illinois Consortium, Chicago: L. Smith (principal investigator), J. Moy, E. Naureckas, A. Prestridge (co-principal investigators), J. Hixon (principal clinic coordinator), A. Brees, J. Judge (coordinators);

Indiana University, Asthma Clinical Research Center, Indianapolis: M. Busk (principal investigator), P. Puntenney (principal clinic coordinator), N. Busk, J. Hutchins (coordinators);

University of Pennsylvania, Philadelphia: F. Leone (principal investigator), M. Hayes-Hampton (principal clinic coordinator);

National Jewish Health, Denver: R. Katial (principal investigator), M. Krawiecz (co-principal investigator), H. Currier (principal clinic coordinator);

Nemours Children's Clinic–University of Florida Consortium, Jacksonville: J. Lima (principal investigator), K. Blake (co-principal investigator), J Lang (co-principal investigator), K Schaeffer (investigator), A Santos (principal coordinator), M McRae (coordinator)

Hofstra University School of Medicine (formerly North Shore–Long Island Jewish Health System), New Hyde Park, N.Y.: R. Cohen (principal investigator), M. Santiago (co-principal investigator), R. Ramdeo (principal clinic coordinator);

Northern New England Consortium (formerly Vermont Lung Center at the University of Vermont), Colchester, Vt.: C.G. Irvin (principal investigator), A.E. Dixon, D.A. Kaminsky, T. Lahiri, P.A. Shapero (co-principal investigators), R. Colletti (GI consultant), S.M. Burns, L.V. Griffes, R. Pratt, M. Doucette, P. Oertel (coordinators);

The Ohio State University Medical Center/Columbus Children's Hospital, Columbus: J. Mastronarde (principal investigator), K. McCoy (co-principal investigator), J. Parsons (co-investigator), J. Drake (principal clinic coordinator), R. Compton, L. Raterman, D. Cosmar (coordinators);

Maria Fareri Children's Hospital at Westchester Medical Center and New York Medical College: A. Dozor (principal investigator), I. Gherson (principal clinic coordinator), L. Monchil (research coordinator).

University of Alabama at Birmingham, Birmingham: L.B. Gerald (principal investigator), W.C. Bailey, R. Grad (co-principal investigators), S. Erwin (principal clinic coordinator), A. Kelley, D. Laken (coordinators);

University of Miami, Miami–University of South Florida, Tampa: A. Wanner (principal investigator, Miami), R. Lockey (principal investigator, Tampa), E. Mendes (principal clinic coordinator for University of Miami), S. McCullough (principal clinic coordinator for University of South Florida), M. Grandstaff-Singleton, D. Miller (coordinators);

University of Minnesota, Minneapolis: M.N. Blumenthal (principal investigator), G. Brottman, J. Hagen (co-principal investigators), A. Decker, D. Lascewski, S. Kelleher (principal clinic coordinators), K. Bachman, C. Quintard, C. Sherry (coordinators);

University of Missouri, Kansas City School of Medicine, Kansas City: G. Salzman (principal investigator), C. Dinakar, D. Pyszczynski (co-principal investigators), P. Haney (principal clinic coordinator);

St. Louis Asthma Clinical Research Center: Washington University, St. Louis: M. Castro (principal investigator), L. Bacharier, K. Sumino (co-investigators), J. Tarsi (principal coordinator), B. Patterson (coordinator);

University of California San Diego: S. Wasserman (principal investigator), J. Ramsdell (co-principal investigator), P. Ferguson, K. Kinninger, T. Greene (clinic coordinators);

Chairman's Office University of Alabama, Birmingham (formerly at Respiratory Hospital, Winnipeg, Man., Canada): W. Bailey and N. Anthonisen (research group chair);

Data Coordinating Center, Johns Hopkins University Center for Clinical Trials, Baltimore: R. Wise (center director), J. Holbrook (deputy director), E. Brown (principal coordinator), D. Amend-Libercci, K. Barry, M. Daniel, A. Lears, G. Leatherman, C. Levine, D. Nowakowski, N. Prusakowski, S. Rayapudi, S. Roettger, A. Thurman, D. Shade, E. Sugar, C. Wei;

Esophageal pH Probe Quality Control Center, Children's Center for Digestive Healthcare Pediatric Gastroenterology, Hepatology, and Nutrition, (formerly at Emory University School of Medicine): B. Gold (center director);

Data and Safety Monitoring Board: S. Lazarus (chair), W. Calhoun, M. Cloutier, B. McWilliams, A. Rogatko, C. Sorkness;

Project Office, American Lung Association, New York: E. Lancet (project officer), N. Edelman (scientific consultant), S. Rappaport;

Project Office, National Heart Lung and Blood Institute: V. Taggart (project officer), G. Weinmann (DSMB secretary, airway branch chief);

ALA Scientific Advisory Committee: E. N. Schachter (chair), L. A. Baggott (vice-chair) W. C. Bailey, A. L. Brannen II, M. Castro, B. W. Christman, A. Chuang, R. M. Donaldson, C. Holloway, T. A. Mahr, J. A. Neubauer, J. M. Samet, E. R. Swenson, D. J. Upson, D. J. Weiss, R. Wise.

The protocol committee for the trial also served as the writing committee. All members had input into the design of the study, acquisition of data, interpretation of statistical analysis of data and critical revision of the manuscript. Drs. Holbrook and Wise have access to the full database and take responsibility for the integrity of the data and of the data analysis.

Supported by the American Lung Association Asthma Clinical Research Center's Infrastructure Award, and NIH/NHLBI U01 HL080433 (Teague) and U01 HL080450 (Holbrook). Lansoprazole and placebo were provided by Takeda Pharmaceuticals, North America, Albuterol HFA was provided GlaxoSmithKline. Sponsors had no role in design, conduct, analysis, or interpretation of the study.

Footnotes

*

Investigators and staff of the American Lung Association Asthma Clinical Research Centers are listed in the appendix.

References

  • 1.Tucci F, Resti M, Fontana R, Novembre E, Lami CA, Vierucci A. Gastroesophageal reflux and bronchial asthma: prevalence and effect of cisapride therapy. J Pediatr Gastroenterol Nutr. 1993 Oct;17(3):265–70. doi: 10.1097/00005176-199310000-00006. [DOI] [PubMed] [Google Scholar]
  • 2.Cinquetti M, Micelli S, Voltolina C, Zoppi G. The pattern of gastroesophageal reflux in asthmatic children. J Asthma. 2002 Apr;39(2):135–42. doi: 10.1081/jas-120002194. [DOI] [PubMed] [Google Scholar]
  • 3.Vandenplas Y, Rudolph CD, Di Lorenzo C, et al. Pediatric gastroesophageal reflux clinical practice guidelines: joint recommendations of the North American Society for Pediatric Gastroenterology, Hepatology, and Nutrition (NASPGHAN) and the European Society for Pediatric Gastroenterology, Hepatology, and Nutrition (ESPGHAN). J Pediatr Gastroenterol Nutr. 2009 Oct;49(4):498–547. doi: 10.1097/MPG.0b013e3181b7f563. [DOI] [PubMed] [Google Scholar]
  • 4.Hancox RJ, Poulton R, Taylor DR, et al. Associations between respiratory symptoms, lung function and gastrooesophageal reflux symptoms in a population-based birth cohort. Respir Res. 2006 Dec 5;7:142. doi: 10.1186/1465-9921-7-142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Andze GO, Brandt ML, St Vil D, Bensoussan AL, Blanchard H. Diagnosis and treatment of gastroesophageal reflux in 500 children with respiratory symptoms: The value of pH monitoring. J Pediatr Surg. 1991 Mar;26(3):295–9. doi: 10.1016/0022-3468(91)90505-n. [DOI] [PubMed] [Google Scholar]
  • 6.El-Serag HB, Gilger M, Kuebeler M, Rabeneck L. Extraesophageal associations of gastroesophageal reflux disease in children without neurologic defects. Gastroenterology. 2001 Dec;121(6):1294–9. doi: 10.1053/gast.2001.29545. [DOI] [PubMed] [Google Scholar]
  • 7.Harding SM, Richter JE. The role of gastroesophageal reflux in chronic cough and asthma. Chest. 1997 May;111(5):1389–402. doi: 10.1378/chest.111.5.1389. [DOI] [PubMed] [Google Scholar]
  • 8.Sheikh S, Stephen T, Howell L, Eid N. Gastroesophageal reflux in infants with wheezing. Pediatr Pulmonol. 1999 Sep;28(3):181–6. doi: 10.1002/(sici)1099-0496(199909)28:3<181::aid-ppul4>3.0.co;2-s. [DOI] [PubMed] [Google Scholar]
  • 9.Sherman PM, Hassall E, Fagundes-Neto U, Gold BD, Kato S, Koletzko S, Orenstein S, Rudolph C, Vakie N, Vandenplas Y. A global, evidence-based consensus on the definition of gastroesophageal reflux disease in the pediatric populations. Am J Gastroenterol. 2009;104(5):1278–1295. doi: 10.1038/ajg.2009.129. [DOI] [PubMed] [Google Scholar]
  • 10.EPR—3. Expert panel report 3: guidelines for the diagnosis and management of asthma (EPR-3 2007). NIH publication no. 08-4051. Department of Health and Human Services; National Institutes of Health; National Heart, Lung, and Blood Institute; National Asthma Education and Prevention Program; Bethesda, MD: U.S.: 2007. [Google Scholar]
  • 11.Kiljander TO, Harding SM, Field SK, et al. Effects of esomeprazole 40 mg twice daily on asthma: a randomized placebo-controlled trial. Am J Respir Crit Care Med. 2006 May 15;173(10):1091–7. doi: 10.1164/rccm.200507-1167OC. [DOI] [PubMed] [Google Scholar]
  • 12.Kiljander TO, Junghard O, Beckman O, Lind T. Effect of esomeprazole 40 mg once or twice daily on asthma: a randomized, placebo-controlled study. Am J Respir Crit Care Med. 2010 May 15;181(10):1042–8. doi: 10.1164/rccm.200910-1537OC. [DOI] [PubMed] [Google Scholar]
  • 13.American Lung Association Asthma Clinical Research Centers. Mastronarde JG, Anthonisen NR, et al. Efficacy of esomeprazole for treatment of poorly controlled asthma. N Engl J Med. 2009 Apr 9;360(15):1487–99. doi: 10.1056/NEJMoa0806290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Khoshoo V, Haydel R. Effect of antireflux treatment on asthma exacerbation in nonatopic children. J Pediatr Gastroenterol Nutr. 2007;44(3):331–335. doi: 10.1097/MPG.0b013e31802fe89c. [DOI] [PubMed] [Google Scholar]
  • 15.Revicki DA, Leidy NK, Brennan-Diemer F, Sorensen S, Togias A. Integrating patient preferences into health outcomes assessment: the multiattribute Asthma Symptom Utility Index. Chest. 1998 Oct;114(4):998–1007. doi: 10.1378/chest.114.4.998. [DOI] [PubMed] [Google Scholar]
  • 16.Nathan RA, Sorkness CA, Kosinski M, Schatz M, Li JT, Marcus P, Murray JJ, Pendergraft TB. Development of the asthma control test: a survey for assessing asthma control. J Allergy Clin Immunol. 2004 Jan;113(1):59–65. doi: 10.1016/j.jaci.2003.09.008. [DOI] [PubMed] [Google Scholar]
  • 17.Liu AH, Zeiger R, Sorkness C, Mahr T, Ostrom N, Burgess S, Rosenzweig JC, Manjunath R. Development and cross-sectional validation of the Childhood Asthma Control Test. J Allergy Clin Immunol. 2007 Apr;119(4):817–25. doi: 10.1016/j.jaci.2006.12.662. [DOI] [PubMed] [Google Scholar]
  • 18.Juniper EF, Guyatt GH, Feeny DH, Ferrie PJ, Griffith LE, Townsend M. Measuring quality of life in the parents of children with asthma. Qual Life Res. 1996 Feb;5(1):27–34. doi: 10.1007/BF00435966. [DOI] [PubMed] [Google Scholar]
  • 19.Crapo RO, Casaburi R, Coates AL, et al. Guidelines for methacholine and exercise challenge testing-1999 This official statement of the American Thoracic Society was adopted by the ATS Board of Directors, July 1999. Am J Respir Crit Care Med. 2000 Jan;161(1):309–29. doi: 10.1164/ajrccm.161.1.ats11-99. [DOI] [PubMed] [Google Scholar]
  • 20.Hankinson JL, Odencrantz JR, Fedan KB. Spirometric reference values from a sample of the general US population. Am J Respir Crit Care Med. 1999 Jan;159(1):179–87. doi: 10.1164/ajrccm.159.1.9712108. [DOI] [PubMed] [Google Scholar]
  • 21.Food and Drug Administration . Postmarket Drug Safety Information for Patients and Providers: FDA Drug Safety Communication [Internet] Food and Drug Administration (US); Rockville (MD): [2011 Sep 8]. c2010 [updated 2011 Mar 3] Available from: http://www.fda.gov/Drugs/DrugSafety/PostmarketDrugSafetyInformationforPatientsandProviders/ucm213206.htm. [Google Scholar]
  • 22.Zeger SL, Liang KY. Longitudinal data analysis for discrete and continuous outcomes. Biometrics. 1986 Mar;42(1):121–30. [PubMed] [Google Scholar]
  • 23.Keene ON, Jones MR, Lane PW, Anderson J. Analysis of exacerbation rates in asthma and chronic obstructive pulmonary disease: example from the TRISTAN study. Pharm Stat. 2007 Apr-Jun;6(2):89–97. doi: 10.1002/pst.250. [DOI] [PubMed] [Google Scholar]
  • 24.Tolia V, Ferry G, Gunasekaran T, Huang B, Keith R, Book L. Efficacy of lansoprazole in the treatment of gastroesophageal reflux disease in children. J Pediatr Gastroenterol Nutr. 2002;35(Suppl 4):S308–18. doi: 10.1097/00005176-200211004-00003. [DOI] [PubMed] [Google Scholar]
  • 25.Khoshoo V, Mohnot S, Haydel R, Jr, Saturno E, Edell D, Kobernick A. Bronchial hyperreactivity in non-atopic children with asthma and reflux: effect of anti-reflux treatment. Pediatr Pulmonol. 2009 Nov;44(11):1070–4. doi: 10.1002/ppul.21094. [DOI] [PubMed] [Google Scholar]
  • 26.Størdal K, Johannesdottir GB, Bentsen BS, et al. Acid suppression does not change respiratory symptoms in children with asthma and gastro-oesophageal reflux disease. Arch Dis Child. 2005;90(9):956–260. doi: 10.1136/adc.2004.068890. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Condino AA, Sondheimer J, Pan Z, Gralla J, Perry D, O'Connor JA. Evaluation of gastroesophageal reflux in pediatric patients with asthma using impedance-pH monitoring. J Pediatr. Aug. 2006;149(2):216–219. doi: 10.1016/j.jpeds.2006.03.022. [DOI] [PubMed] [Google Scholar]
  • 28.Tolia V, Vandenplas Y. Systematic review: the extra-oesophageal symptoms of gastro-oesophageal reflux disease in children. Aliment Pharmacol Ther. 2009 Feb 1;29(3):258–272. doi: 10.1111/j.1365-2036.2008.03879.x. [DOI] [PubMed] [Google Scholar]
  • 29.Blondeau K, Mertens V, Dupont L, et al. The relationship between gastroesophageal reflux and cough in children with chronic unexplained cough using combined impedance-pH-manometry recordings. Pediatr Pulmonol. 2011;46:286–294. doi: 10.1002/ppul.21365. [DOI] [PubMed] [Google Scholar]
  • 30.DiMango E, Holbrook JT, Simpson E, et al. Effects of asymptomatic proximal and distal gastroesophageal reflux on asthma severity. Am J Respir Crit Care Med. 2009 Nov 1;180(9):809–816. doi: 10.1164/rccm.200904-0625OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Orensteim SR, Hassall E, Furmaga-Jablonska W, Atkinson S, Raanan M. Multicenter, double-blind, randomized, placebo-controlled trial assessing the efficacy and safety of proton pump inhibitor lansoprazole in infants with symptoms of gastroesophageal reflux disease. J Pediatr. 2009;154:514–520. doi: 10.1016/j.jpeds.2008.09.054. [DOI] [PubMed] [Google Scholar]
  • 32.Meijvis SC, Cornips MC, Paul Voorn G, et al. Microbial evaluation of proton pump inhibitors and the risk of pneumonia. Eur Respir J. 2011 Apr 18; doi: 10.1183/09031936.00020811. [DOI] [PubMed] [Google Scholar]
  • 33.Eom CS, Jeon CY, Lim JW, Cho EG, Park SM, Lee KS. Use of acid-suppressive drugs and risk of pneumonia: a systematic review and meta-analysis. CMAJ. 2011 Feb 22;183(3):310–319. doi: 10.1503/cmaj.092129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Laheij RJ, Sturkenboom MC, Hassing RJ, Dieleman J, Stricker BH, Jansen JB. Risk of community-acquired pneumonia and use of gastric acid-suppressive drugs. JAMA. 2004 Oct 27;292(16):1955–1960. doi: 10.1001/jama.292.16.1955. [DOI] [PubMed] [Google Scholar]
  • 35.Canani RB, Cirillo P, Roggero P, et al. Therapy with gastric acidity inhibitors increases the risk of acute gastroenteritis and community-acquired pneumonia in children. Pediatrics. 2006 May;117(5):e817–820. doi: 10.1542/peds.2005-1655. [DOI] [PubMed] [Google Scholar]
  • 36.Ohara T, Arakawa T. Lansoprazole decreases peripheral blood monocytes and intercellular adhesion molecule-1-positive mononuclear cells. Dig Dis Sci. 1999 Aug;44(8):1710–5. doi: 10.1023/a:1026604203237. [DOI] [PubMed] [Google Scholar]
  • 37.Yoshida N, Yoshikawa T, Tanaka Y, et al. A new mechanism for anti-inflammatory actions of proton pump inhibitors--inhibitory effects on neutrophil-endothelial cell interactions. Aliment Pharmacol Ther. 2000 Apr;14(Suppl 1):74–81. doi: 10.1046/j.1365-2036.2000.014s1074.x. [DOI] [PubMed] [Google Scholar]
  • 38.Zedtwitz-Liebenstein K, Wenisch C, Patruta S, Parschalk B, Daxböck F, Graninger W. Omeprazole treatment diminishes intra- and extracellular neutrophil reactive oxygen production and bactericidal activity. Crit Care Med. 2002 May;30(5):1118–22. doi: 10.1097/00003246-200205000-00026. [DOI] [PubMed] [Google Scholar]
  • 39.Taylor AM. Pediatric focused safety review proton pump inhibitors esomeprazole, lansoprazole, omeprazole, rabeprazole. [October 5, 2011];Presented to FDA Pediatric Advisory Committee. 2010 Jun 21; http://www.fda.gov/downloads/AdvisoryCommittees/CommitteesMeetingMaterials/PediatricAdvisoryCommittee/UCM216303.pdf.
  • 40. [October 5, 2011];Transcripts of the June 21, 2010 meeting of the Pediatric Advisory Committee. http://www.fda.gov/AdvisoryCommittees/CommitteesMeetingMaterials/PediatricAdvisoryCommittee/ucm201871.htm.
  • 41. [October 5, 2011];Transcripts of the November 5, 2010 meeting of the Gastrointestinal Drugs Advisory Committee. http://www.fda.gov/AdvisoryCommittees/CommitteesMeetingMaterials/Drugs/GastrointestinalDrugsAdvisoryCommittee/ucm195280.htm.

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

01

RESOURCES