The effects of a novel metabotropic glutamate receptor 5 antagonist (AZD2066) on transient lower...

12
The effects of a novel metabotropic glutamate receptor 5 antagonist (AZD2066) on transient lower oesophageal sphincter relaxations and reux episodes in healthy volunteers W. O. Rohof*, A. Lei*, D. P. Hirsch*, L. Ny , M. Astrand , M. B. Hansen & G. E. Boeckxstaens* ,*Academic Medical Center, Amsterdam, The Netherlands. AstraZeneca R&D, Mölndal, Sweden. Department of Gastroenterology, University Hospital Leuven, Catholic University of Leuven, Belgium. Correspondence to: Prof. Dr G. E. Boeckxstaens, Translational Research Center for Gastrointestinal Disorders (TARGID), Department of Gastroenterology, University Hospital Leuven, Catholic University of Leuven, Herestraat 49, 3000 Leuven, Belgium. E-mail: guy.boeckxstaens@med. kuleuven.be Publication data Submitted 10 June 2011 First decision 3 August 2011 Resubmitted 10 March 2012 Accepted 10 March 2012 EV Pub Online 2 April 2012 SUMMARY Background Selective metabotropic glutamate receptor 5 (mGluR5) antagonists inhibit transient lower oesophageal sphincter relaxations (TLESRs) in animals and acid reux in humans. Aim To assess the effect of single doses of the mGluR5 antagonist AZD2066 on TLESRs and reux in humans. Methods Healthy male volunteers received AZD2066 13 mg and placebo (part A), or AZD2066 2 mg and AZD2066 6 mg and placebo (part B), in a randomised crossover study. Postprandial manometry/pH-impedance measurements were taken after each dose. Results A total of 13 individuals completed part A of the study and 19 individuals completed part B. There was a signicant reduction in the geometric mean number of TLESRs (27%; P = 0.02) and the geometric mean number of reux episodes (51%; P = 0.01) in subjects receiving AZD2066 13 mg com- pared with placebo. Adverse events in participants receiving AZD2066 13 mg were mostly related to the nervous system [dizziness (3/13); distur- bance in attention (3/13)]. Adverse events were reversible and of mild intensity. There were no serious adverse events. The effects of AZD2066 appeared dose-dependent, with smaller reductions in TLESRs and reux episodes (relative to placebo) and fewer adverse events observed for AZD2066 2 mg and AZD2066 6 mg compared with AZD2066 13 mg. Conclusion The mGluR5-mediated inhibition of TLESRs may be a useful approach for inhibiting gastro-oesophageal reux. Aliment Pharmacol Ther 2012; 35: 12311242 ª 2012 Blackwell Publishing Ltd 1231 doi:10.1111/j.1365-2036.2012.05081.x Alimentary Pharmacology and Therapeutics

Transcript of The effects of a novel metabotropic glutamate receptor 5 antagonist (AZD2066) on transient lower...

The effects of a novel metabotropic glutamate receptor 5antagonist (AZD2066) on transient lower oesophagealsphincter relaxations and reflux episodes in healthy volunteersW. O. Rohof*, A. Lei*, D. P. Hirsch*, L. Ny†, M. Astrand†, M. B. Hansen† & G. E. Boeckxstaens*,‡

*Academic Medical Center,Amsterdam, The Netherlands.†AstraZeneca R&D, Mölndal, Sweden.‡Department of Gastroenterology,University Hospital Leuven, CatholicUniversity of Leuven, Belgium.

Correspondence to:Prof. Dr G. E. Boeckxstaens,Translational Research Center forGastrointestinal Disorders (TARGID),Department of Gastroenterology,University Hospital Leuven, CatholicUniversity of Leuven, Herestraat 49,3000 Leuven, Belgium.E-mail: [email protected]

Publication dataSubmitted 10 June 2011First decision 3 August 2011Resubmitted 10 March 2012Accepted 10 March 2012EV Pub Online 2 April 2012

SUMMARY

BackgroundSelective metabotropic glutamate receptor 5 (mGluR5) antagonists inhibittransient lower oesophageal sphincter relaxations (TLESRs) in animals andacid reflux in humans.

AimTo assess the effect of single doses of the mGluR5 antagonist AZD2066 onTLESRs and reflux in humans.

MethodsHealthy male volunteers received AZD2066 13 mg and placebo (part A), orAZD2066 2 mg and AZD2066 6 mg and placebo (part B), in a randomisedcrossover study. Postprandial manometry/pH-impedance measurementswere taken after each dose.

ResultsA total of 13 individuals completed part A of the study and 19 individualscompleted part B. There was a significant reduction in the geometric meannumber of TLESRs (27%; P = 0.02) and the geometric mean number ofreflux episodes (51%; P = 0.01) in subjects receiving AZD2066 13 mg com-pared with placebo. Adverse events in participants receiving AZD206613 mg were mostly related to the nervous system [dizziness (3/13); distur-bance in attention (3/13)]. Adverse events were reversible and of mildintensity. There were no serious adverse events. The effects of AZD2066appeared dose-dependent, with smaller reductions in TLESRs and refluxepisodes (relative to placebo) and fewer adverse events observed forAZD2066 2 mg and AZD2066 6 mg compared with AZD2066 13 mg.

ConclusionThe mGluR5-mediated inhibition of TLESRs may be a useful approach forinhibiting gastro-oesophageal reflux.

Aliment Pharmacol Ther 2012; 35: 1231–1242

ª 2012 Blackwell Publishing Ltd 1231

doi:10.1111/j.1365-2036.2012.05081.x

Alimentary Pharmacology and Therapeutics

INTRODUCTIONGastro-oesophageal reflux disease (GERD) is a chronicdisease caused by reflux of gastric contents into theoesophagus.1, 2 In Western countries, approximately 10–20% of adults are thought to be affected by GERD symp-toms, with lower rates of around 5% reported in Asianpopulations.3, 4

The mainstay of GERD treatment is acid suppression,most commonly achieved with the use of proton pumpinhibitors (PPIs).5, 6 However, there is mounting evi-dence that reflux symptoms persist in a high proportion(20–30%) of patients despite PPI therapy,7–10 and thatthey are associated with decreased psychological andphysical well-being.11

Transient lower oesophageal sphincter relaxations(TLESRs) are considered to be a major mechanism forgenerating reflux in healthy individuals and in patientswith GERD.12, 13 Pharmacological inhibition of TLESRsis a promising new therapeutic approach because it hasthe potential to reduce most types of reflux. This is par-ticularly relevant for patients who have troublesomereflux symptoms despite acid suppression from PPI use,because weakly acidic and weakly alkaline refluxes arethought to play a role in symptom generation in thisgroup.14

The TLESRs are triggered in response to the activationof stretch receptors in the stomach,15 and are thought tobe mediated by a vago-vagal reflex pathway.16, 17 Thegamma-aminobutyric acid type B (GABAB) receptor actsat several points along this pathway, and recent clinicalstudies demonstrate that inhibition of TLESRs using theGABAB-receptor agonist lesogaberan reduces the numberof reflux episodes and symptoms.18, 19

Inhibition of TLESRs may also be achieved via otherpathways. In particular, metabotropic glutamate receptor5 (mGluR5) expressed in the soma of gastric vagal affer-ents appears to play an important role in triggeringTLESRs.20 Selective antagonists of mGluR5 have beenshown to inhibit TLESRs in animal studies,21, 22 and toreduce acid reflux and to improve clinical symptoms inpatients with GERD.23 However, the effect of mGluR5inhibition on the number of reflux episodes has not beenassessed in humans in relation to TLESR frequency.

The AZD2066 is a novel chemical substance that hasbeen pharmacologically characterised as a selective,non-competitive antagonist of mGluR5, and is beingevaluated for the treatment of neuropathic pain, depres-sion and GERD. The primary objective of this pharma-codynamic study was to measure the effect of AZD2066on the frequency of TLESRs in healthy volunteers.

Secondary objectives included assessing the effect ofAZD2066 on the frequency of TLESRs, number and typeof reflux episodes, oesophageal acid exposure, loweroesophageal sphincter (LES) pressure and number ofswallows, and to determine its pharmacokinetic charac-teristics and safety and tolerability profile.

METHODS

Study participantsTo be enrolled, study participants were required to behealthy men, aged 18–45 years, with a body mass index(BMI = weight/height2) of 19–30 kg/m2. Full details ofthe inclusion/exclusion criteria implemented during thestudy are presented in supplementary Table S1. At the21-day pre-entry visit, each subject had a medical exami-nation which included a full laboratory screening, aphysical examination (general appearance, cardiovascu-lar, lungs, abdomen, skin, head and neck, lymph nodes,thyroid, musculoskeletal/extremities and reflexes), a 12-lead resting ECG recording, blood pressure measurementand a pregnancy test for women. Tests for Hepatitis Band C, HIV and drug abuse were also performed at thepre-entry visit, and participants also underwent a struc-tured psychiatric interview using the Mini InternationalNeuropsychiatric Interview. Participants had to haveclinically normal electrocardiogram, pulse and bloodpressure measurements and laboratory values at the pre-entry visit, and normal laboratory values before adminis-tering the first dose at the second visit.

This study was conducted in accordance with the Dec-laration of Helsinki and the International Conference onHarmonisation Guidelines for Good Clinical Practice,and was approved by the Institutional Review Boardassociated with the study centre (Department of Gastro-enterology, Academic Medical Centre, Amsterdam, TheNetherlands). Participants provided written informedconsent prior to the study. This study is registered on apublicly accessible website (ClinicalTrials.gov Identifier:NCT00813306).

Study design and study drugsThis was a double-blind, randomised, single-centre, phase1, crossover study (2008–2009) conducted in two parts,each involving a different set of participants. Part Aassessed the impact of AZD2066 on gastro-oesophagealsphincter function and reflux variables using a singledose (13 mg) considered to have an acceptable safety andtolerability profile, based on an interim analysis of a mul-tiple ascending dose study (study code: DO475C00002).

1232 Aliment Pharmacol Ther 2012; 35: 1231-1242

ª 2012 Blackwell Publishing Ltd

W. O. Rohof et al.

Each participant received 130 mL of 0.1 mg/mL oralAZD2066 (13 mg) solution and 130 mL of oral placebosolution, each on different days separated by a 7- to 28-day washout period (Figure 1, study part A).

Part B of the study explored the dose–response rela-tionship for AZD2066 using two lower single doses (2 and6 mg). Each participant received 60 mL of 0.1 mg/mLoral AZD2066 (6 mg) solution, 20 mL of 0.1 mg/mLoral AZD2066 (2 mg) solution made up to 60 mL withoral placebo solution and 60 mL oral placebo solutioneach on different days. A washout period of 7–28 dayswas initially planned for both parts of the study. How-ever, in part A of the study, AZD2066 (5–6 nmol atCmax) was detected in the plasma of two subjects in theplacebo-treated group after treatment with AZD2066 anda washout period of 8 days. The washout period betweeneach treatment in part B of the study was thus increasedto 10- to 28-days (Figure 1, study part B).

In both parts of the study, the first dose (AZD2066 orplacebo) was administered � 21 days after the pre-entryvisit. Food and fluids were not allowed after midnight onthe night before dose administration. Participantsreceived a standardised breakfast [700 kcal, of which219 kcal (31.3%) were from fat] 30 min after administra-tion of AZD2066 or placebo, which was to be consumedwithin 15 min. Oesophageal manometric and pH-imped-ance measurements were collected from participants(while in a semi-recumbent position) for 3 h 45 min, sothat 3 h of data collection occurred in the postprandial

period. Participants also had a follow-up visit 5–7 daysafter the last study period.

Assignment and blindingThe principal investigator (GB) assigned individuals tounique enrolment numbers, determined subject eligibilityand assigned eligible participants to unique randomisa-tion numbers. A randomised dosing regimen allocationlist was generated by AstraZeneca R&D Mölndal using avalidated computer program, and the randomisation wasperformed within blocks of consecutive patient numbers.The study was double-blinded so that both investigatorsand participants were unaware of who received whichdosing regimen. Treatment codes were not broken untilthe evaluability of the data from each participant hadbeen established and documented.

Pharmacodynamic assessmentsManometric recordings of the pharynx, oesophagus, LESand stomach were obtained using a perfused 12-channelsilicone rubber assembly (DentSleeve International Ltd,Mississauga, ON, Canada). The manometric catheter waspositioned with the proximal border of the sleeve 1 cmabove the LES. Manometric readings from dosing untilthe end of the 3-h postprandial period were used toassess the following.

(i) Number of TLESRs, defined according to previ-ously described criteria,24 and their time of onset

Pre-entryvisit

Pre-entryvisit

Follow-up

Study period 2

Study period 2 Study period 3

Study period 1

10–28 days 10–28 days 5–7 days

5–7 days7–28 days

Study design part A

Study design part B

≤21 days

≤21 days

Study period 1

Follow-up

AZD2066 13 mgor

placebo

AZD2066 2 mgor

AZD2066 6 mgor

placebo

AZD2066 2 mgor

AZD2066 6 mgor

placebo

AZD2066 2 mgor

AZD2066 6 mgor

placebo

AZD2066 13 mgor

placebo

Figure 1 | Randomised, crossover design used for parts A and B of the study.

Aliment Pharmacol Ther 2012; 35: 1231-1242 1233

ª 2012 Blackwell Publishing Ltd

Effects of AZD2066 on TLESRs and reflux

(primary outcome in relation to AZD2066 13 mg; sec-ondary outcome in relation to AZD2066 2 mg andAZD2066 6 mg).(ii) LES pressure (recorded every 15 min and

expressed as the mean difference between the end expi-ration LES pressure and the end expiration intragastricpressure over 1 min). Mean LES pressure during the 3-hpostprandial period was calculated from measurementstaken 15 min after dose administration and then every15 min until 3 h after the meal (3 h 45 min in total)(secondary outcome).(iii) Number of swallows (defined as a fast increase in

pressure in the pharyngeal channel, clearly distinguish-able from baseline activity) (secondary outcome).

For the impedance-pH readings, a catheter, compris-ing several electrode pairs measuring intraluminalimpedance, and an antimony pH sensor were used. Thecatheter was positioned so that the pH sensor waslocated 5 cm above the LES. Impedance-pH readingsfrom dosing until the end of the 3-h postprandial periodwere used to assess the following:

(i) Number of reflux episodes, defined as a periodwhen impedance decreased to below 50% of baseline(liquid episode) or increased to above 150% of baseline(gas episode), propagating aborally from the most distalchannel (secondary outcome).(ii) Number of reflux episodes that were acidic (intra-

oesophageal pH <4 [or a drop of � 1 pH unit if pH isalready <4] lasting more than 5 s), weakly acidic (intra-oesophageal pH 4.0–6.5 lasting more than 5 s) andweakly alkaline (intra-oesophageal pH � 6.5 lastingmore than 5 s) (secondary outcome).(iii) Percentage of time with an oesophageal pH <4

(secondary outcome).

Pharmacokinetic, safety and tolerability assessmentsBlood samples (2 mL) were collected at frequent inter-vals for 12 h after each dose of AZD2066 to determinethe pharmacokinetic profile (secondary outcome). PlasmaAZD2066 concentrations were determined by usingliquid chromatography and mass spectrometry (quantifi-cation limit: 1.00 nmol⁄L). Adverse events occurring inthe first 24 h after dose administration were consideredto be during active dosing.

Statistical analysisFor part A of the study, it was determined that completedata from a minimum of 10 subjects would be required

to detect a true effect of a 50% reduction in the numberof TLESRs (primary variable). This was based on using aone-sided t-test at a significance threshold of 5%, with90% power and a within subject standard deviation of0.5 for the difference (placebo versus AZD2066) in thelog number of TLESRs. The null hypothesis (H0) wasthat the difference in the log number of TLESRs is � 0.This was tested against the alternative hypothesis (H1)that the difference is >0. It was determined that at least14 subjects would need to be enrolled to ensure that aminimum of 10 subjects would complete the study. Theresults of part A of the study revealed the true effect ofAZD2066 on TLESR frequency to be lower than antici-pated (i.e. there was a 27% reduction in the number ofTLESRs). Based on detecting a true effect of a 27%reduction in the number of TLESRs, it was calculatedthat at least 18 participants would need to complete partB of the study to provide adequate statistical powerbased on the same parameters used in part A. It wasdetermined that at least 22 subjects would need to beenrolled to ensure this level of completion.

Pharmacodynamic and pharmacokinetic variables wereanalysed for all participants not affected by major proto-col violations (per protocol data). Major protocol viola-tions were defined as those that would have led toconcerns over the validity of the analysis and included:entering the study despite not meeting study entry crite-ria; continuing the study despite meeting criteria forbeing withdrawn; receiving the incorrect treatment ordose; taking prohibited concomitant medications duringthe study and severe non compliance with the study pro-tocol. All participants who received at least one dose ofAZD2066 or placebo and for whom post dose informa-tion was available were included in the safety analysis.

Geometric means were used to compare treatmenteffects where possible because this method of calculatingthe mean is less biased by extreme values than the arith-metic mean. Mean total differences in the effect ofAZD2066 and placebo could only be estimated as geo-metric mean ratios if data were able to be log trans-formed i.e. if the value of the variable being assessed was� 1 for every patient. Values for LES pressure and thenumber of TLESRs, reflux episodes and swallows thatoccurred during the 0–3 h postprandial period were ableto be log transformed in the analysis. Analyses for log-transformable data were performed using analysis of var-iance (ANOVA) with treatment, period and sequence asfixed effects and participant’s ID as a random effect.Carry-over effects were not specifically analysed because

1234 Aliment Pharmacol Ther 2012; 35: 1231-1242

ª 2012 Blackwell Publishing Ltd

W. O. Rohof et al.

they cannot be distinguished from treatment, sequenceand period effects in 2 9 2 crossover studies,25 and onlythree of these factors can be assessed at the same time inthe ANOVA model (i.e. to estimate treatment, sequenceand period effects it must be assumed that the carry-overeffect is zero). However, in line with recommendations,the washout period was based on the pharmacokineticsprofile of AZD2066 and sequence and period effectswere accounted for as a proxy for carry-over effects (i.e.carry-over effects are unlikely if sequence and periodeffects are not detected).25

Confidence intervals (CIs) for the true mean were cal-culated in the logarithmic scale based on the mean squareerror obtained in the ANOVA. The limits were transformedback to the original scale to give a CI for the ratio of geo-metric means between dose regimens. The number ofacidic, weakly acidic, pure liquid and mixed gas/liquidreflux episodes that occurred in the 0–3 h postprandialperiod, and values for the% time with oesophageal pH <4,were not able to be log transformed (i.e. not all values ineach dataset were � 1). These variables were thus com-pared between dose regimens in terms of arithmetic meandifferences for which 95% CIs were calculated using Stu-dent’s t-distribution.

RESULTS

Participant flow and follow-upA total of 16 participants enrolled in part A of the study,of whom three did not meet the inclusion, exclusionand/or screening criteria (described above) at the pre-entry visit. The remaining 13 participants were rando-mised to the dose regimens and all 13 completed part A.Of 24 participants enrolled in part B, two did not meetthe inclusion, exclusion and/or screening criteria the pre-entry visit. The remaining 22 participants were rando-mised to the dose regimens. Two discontinued after thefirst treatment visit in part B (included in the safetyanalysis only): one because of severe noncompliance andone because of involvement in the conduct of the study.Another participant discontinued before administrationof the first dose because of a fainting episode. Impedancedata for one patient (required to calculate the number ofTLESRs and reflux episodes) were excluded from theAZD2066 6 mg analysis owing to poor quality tracings.

Participants’ characteristics at baselineAll 13 participants that completed part A of the studywere white men. The mean age of the group was22.1 years (range, 19–27 years). The mean BMI was 22.4

[Standard deviation (s.d.): 2.1] and the median BMI was22.6 (range, 19.2–26.6). Of the 19 participants who com-pleted part B of the study, 18 were white men and onewas a black man. The mean age of the group was25.5 years (range, 18–32 years). The mean BMI was 24.2(s.d.: 2.1) and the median BMI was 24.1 (range, 20.7–28.7). All participants were judged as being healthy atstudy enrolment based on normal results from the physi-cal examination and investigations.

AnalysisPharmacodynamic results: effect of AZD2066 onTLESRs. There was substantial variation between partici-pants in terms of the number of TLESRs that occurredin both part A (Figure 2a) and part B (Figure 3a) of thestudy. A significant 27% reduction was observed in thegeometric mean number of postprandial TLESRs that

AZD2066 13 mg

AZD2066 13 mg (n = 13)

Time period relative to food intake (h)

Mea

n nu

mbe

r of

TLE

SR

sN

umbe

r of

TLE

SR

s

Placebo

30(a)

(b)

25

20

15

10

5

0

14

12

10

2

4

6

8

0–0.5 to 0.0 0.0 to 1.0 1.0 to 2.0 2.0 to 3.0

Placebo (n = 13)

Figure 2 | Number of TLESRs with 13 mg AZD2066 orplacebo (a) presented as individual patient data for the0–3 h after the standardised meal consumed 45 minafter dose administration (b) presented as geometricmeans for individual time periods before and after thestandardised meal consumed 45 min after doseadministration. Error bars show 95% confidenceintervals.

Aliment Pharmacol Ther 2012; 35: 1231-1242 1235

ª 2012 Blackwell Publishing Ltd

Effects of AZD2066 on TLESRs and reflux

occurred when participants received AZD2066 13 mgcompared with when they received placebo (Table 1). Atthe individual time points, the greatest reductions in thenumber of TLESRs were observed 0–1 h and 1–2 h afterfood intake for AZD2066 13 mg relative to placebo(Figure 2b). In part B of the study, no significant reductionin the mean number of TLESRs was observed during the0- to 3-h postprandial period when participants receivedAZD20662 mg(Table 2)orAZD20666 mg(Table 3) com-pared with when they received placebo, and this wasreflected across all of the individual time points(Figure3b).

Pharmacodynamic results: effect of AZD2066 on refluxepisodes. There was also substantial variation in thetotal number of reflux episodes that occurred duringboth parts of the study (Figure 4a and 5a). There was asignificant 51% reduction in the geometric mean numberof reflux episodes that occurred during the 0- to 3-hpostprandial period when participants received AZD206613 mg compared with when they received placebo(Table 1). The 13 mg dose of AZD2066 appeared toreduce the number of reflux episodes relative to placeboboth before and after food intake, with the greatesteffects observed at the 1 to 2- and 2- to 3-h postprandialtime points (Figure 4b).

The initial analysis of data from part B of the studyshowed a significant 39% reduction in the geometricmean number of reflux episodes during the 0- to 3-hpostprandial period for AZD2066 6 mg compared withplacebo (geometric mean ratio, 0.61; 95% CI, 0.41–0.90;P = 0.01). However, closer inspection revealed thatthis effect was mainly driven by two high responders.

AZD2066 6 mg AZD2066 2 mg

AZD2066 2 mg (n = 19)AZD2066 6 mg (n = 18)

Time period relative to food intake (h)

Mea

n nu

mbe

r of

TLE

SR

sN

umbe

r of

TLE

SR

s

Placebo

40(a)

(b)

35

30

25

20

15

10

5

0

14

12

10

2

4

6

8

0–0.5 to 0.0 0.0 to 1.0 1.0 to 2.0 2.0 to 3.0

Placebo (n = 19)

Figure 3 | Number of reflux episodes with 13 mgAZD2066 or placebo (a) presented as individualpatient data for the 0–3 h after the standardised mealconsumed 45 min after dose administration (b)presented as geometric means for individual timeperiods before and after the standardised mealconsumed 45 min after dose administration. Error barsshow 95% confidence intervals.

Table 1 | Pharmacodynamic effects of AZD2066 13 mg compared with placebo 0–3 h after food intake (study part A)

Geometric mean PlaceboAZD206613 mg

Geometric meanratio (95% CI) P-value

Number of TLESRs (n = 13) 19.0 14.0 0.73 (0.53, 1.00) 0.02Number of reflux episodes (n = 13) 13.0 6.2 0.49 (0.29, 0.85) 0.01LES pressure (n = 13) 9.8 10.0 1.05 (0.83, 1.32) 0.33Number of swallows (n = 13) 233.0 242.0 0.92 (0.77, 1.10) 0.34

Arithmetic meanArithmetic meandifference (95% CI) P-value

Number of acid reflux episodes (n = 13) 8.3 5.3 �3.05 (�5.60, �0.5) 0.01Number of weakly acidic reflux episodes (n = 13) 4.1 3.5 �0.57 (�3.97, 2.83) 0.36Pure liquid reflux episodes (n = 13) 8.7 6.6 �2.06 (�6.30, 2.18) 0.15Mixed gas/liquid reflux episodes (n = 13) 4.4 2.4 �1.98 (�4.82, 0.86) 0.08% time with oesophageal pH <4 (n = 13) 2.5 3.7 �1.20 (�2.59, 0.27) 0.10

CI, confidence interval; LES, lower oesophageal sphincter; TLESR, transient lower oesophageal sphincter.

1236 Aliment Pharmacol Ther 2012; 35: 1231-1242

ª 2012 Blackwell Publishing Ltd

W. O. Rohof et al.

Exclusion of these outliers changed the estimated meanreduction in reflux episodes for AZD2066 6 mg to anonsignificant decrease of 19% compared with placebo(Table 2). Compared with when they received placebo,participants did not have a significant reduction in thegeometric mean number of postprandial reflux episodeswhen they received AZD2066 2 mg (Table 3). The meannumber of reflux episodes that occurred at the individualtime points for AZD2066 6 mg, AZD2066 2 mg and pla-cebo are presented in Figure 5b.

Pharmacodynamic results: effect of AZD2066 on differ-ent types of reflux. In part A of the study, the signifi-cantly lower geometric mean number of reflux episodesthat occurred when participants received AZD206613 mg compared with when they received placebo waslargely accounted for by a statistically significant redu-

ction in the mean number of acid reflux episodes(Table 1). The mean number of acid reflux episodes wasnot significantly reduced when participants receivedAZD2066 6 mg (Table 2) or AZD2066 2 mg (Table 3)compared with when they received placebo, althoughAZD2066 6 mg reduced the mean number of acid refluxepisodes to a larger extent relative to placebo than didAZD2066 2 mg. None of the AZD2066 doses in part Aor B of the study significantly reduced the mean numberof weakly acidic reflux episodes (Tables 2 and 3). Analy-ses were not performed for weakly alkaline reflux epi-sodes owing to their infrequent nature.

In part A of the study, the mean numbers of pureliquid and mixed gas/liquid reflux episodes occurringduring the postprandial period were lower whenparticipants received AZD2066 13 mg than when theyreceived placebo (Table 1), although these effects were

Table 2 | Pharmacodynamic effects of AZD2066 6 mg compared with placebo 0–3 h after food intake (study part B)

Geometric mean PlaceboAZD20666 mg

Geometric meanratio (95% CI) P-value

Number of TLESRs (n = 18) 16.0 16.0 0.98 (0.82, 1.17) 0.41Number of reflux episodes (n = 16) 13.0 7.9 0.81 (0.60, 1.10) 0.08LES pressure (n = 19) 12.0 12.0 0.96 (0.79, 1.18) 0.65Number of swallows (n = 19) 212.0 206.0 0.97 (0.82, 1.16) 0.74

Arithmetic meanArithmetic meandifference (95% CI) P-value

Number of acid reflux episodes (n = 16) 7.8 6.1 �1.74 (�4.45, 0.96) 0.10Number of weakly acidic reflux episodes (n = 16) 3.3 3.5 0.18 (�1.94, 2.30) 0.57Pure liquid reflux episodes (n = 16) 5.2 6.3 1.09 (�1.38, 3.55) 0.81Mixed gas/liquid reflux episodes (n = 16) 6.1 3.4 �2.63 (�5.03, �0.23) 0.02% time with oesophageal pH <4 (n = 18) 3.8 6.3 2.54 (�0.84, 5.92) 0.14

CI, confidence interval; LES, lower oesophageal sphincter; TLESR, transient lower oesophageal sphincter.

Table 3 | Pharmacodynamic effects of AZD2066 2 mg compared with placebo 0–3 h after food intake (study part B)

Geometric mean Placebo AZD2066 2 mgGeometric meanratio (95% CI) P-value

Number of TLESRs (n = 19) 16.0 16.0 0.99 (0.83, 1.18) 0.45Number of reflux episodes (n = 19) 13.0 11.0 0.87 (0.61, 1.26) 0.23LES pressure (n = 19) 12.0 14.0 1.15 (0.94, 1.40) 0.08Number of swallows (n = 19) 212.0 225.0 1.06 (0.89, 1.25) 0.50

Arithmetic meanArithmetic meandifference (95% CI) P-value

Number of acid reflux episodes (n = 19) 7.8 7.7 �0.12 (�2.64, 2.41) 0.46Number of weakly acidic reflux episode s (n = 19) 3.3 3.7 0.37 (�1.61, 2.36) 0.65Pure liquid reflux episodes (n = 19) 5.2 7.4 2.26 (�0.04, 4.57) 0.97Mixed gas/liquid reflux episodes (n = 19) 6.1 4.0 �2.06 (�4.32, 0.19) 0.04% time with oesophageal pH <4 (n = 19) 3.8 5.1 1.30 (�1.94, 4.55) 0.42

CI, confidence interval; LES, lower oesophageal sphincter; TLESR, transient lower oesophageal sphincter.

Aliment Pharmacol Ther 2012; 35: 1231-1242 1237

ª 2012 Blackwell Publishing Ltd

Effects of AZD2066 on TLESRs and reflux

not statistically significant. In part B of the study, themean number of mixed gas/liquid reflux episodes thatoccurred in participants receiving AZD2066 6 mg(Table 2) and AZD2066 2 mg (Table 3) was significantlylower than in those receiving placebo. Pure gas refluxepisodes were not analysed owing to the infrequent nat-ure of these events.

Pharmacodynamic results: relationship between TLESRsand reflux episodes. When patients received AZD206613 mg in part A of the study, 43% of TLESRs were tem-porally related to reflux episodes, compared with 56% ofTLESRs when they received placebo, although this differ-ence was not statistically significant. In part B, the pro-portion of TLESRs temporally related to reflux episodes

was 56% for AZD2066 2 mg and 48% for AZD20666 mg, compared with 55% for placebo.

In part A of the study, the proportion of reflux epi-sodes temporally related to TLESRs was 65% when par-ticipants received AZD2066 13 mg compared with 68%when they received placebo. In part B, 73% and 72% ofreflux episodes were temporally related to TLESRs whenparticipants received AZD2066 2 mg and AZD20666 mg, respectively, compared with 80% when theyreceived placebo. The proportion of acid reflux episodestemporally related to TLESRs was 82.4% when patientsreceived AZD2066 13 mg compared with 86.4% whenthey received placebo, while the proportion of weaklyacidic reflux episodes related to TLESRs was 61.1% forAZD2066 13 mg and 83.7% for placebo.

AZD2066 13 mg

AZD2066 13 mg (n = 13)

Time period relative to food intake (h)

Mea

n nu

mbe

r of

ref

lux

epis

odes

Num

ber

of r

eflu

x ep

isod

es

Placebo

30(a)

(b)

25

20

15

10

5

0

10

9

7

1

2

3

4

5

6

8

0

–1–0.5 to 0.0 0.0 to 1.0 1.0 to 2.0 2.0 to 3.0

Placebo (n = 13)

Figure 4 | Number of TLESRs with 2 mg AZD2066,6 mg AZD2066 or placebo (a) presented as individualpatient data for the 0–3 h after the standardised mealconsumed 45 min after dose administration (b)presented as geometric means for individual timeperiods before and after the standardised mealconsumed 45 min after dose administration. Error barsshow 95% confidence intervals.

AZD2066 6 mg AZD2066 2 mg

AZD2066 2 mg (n = 19)AZD2066 6 mg (n = 16)

Time period relative to food intake (h)

Mea

n nu

mbe

r re

flux

epis

odes

Num

ber

of r

eflu

x ep

isod

es

Placebo

40(a)

(b)

35

30

25

20

15

10

5

0

12

10

2

4

6

8

0–0.5 to 0.0 0.0 to 1.0 1.0 to 2.0 2.0 to 3.0

Placebo (n = 19)

Figure 5 | Number of reflux episodes with 2 mgAZD2066, 6 mg AZD2066 or placebo (a) presentedas individual patient data for the 0–3 h after thestandardised meal consumed 45 min after doseadministration (b) presented as geometric means forindividual time periods before and after thestandardised meal consumed 45 min after doseadministration. Error bars show 95% confidenceintervals.

1238 Aliment Pharmacol Ther 2012; 35: 1231-1242

ª 2012 Blackwell Publishing Ltd

W. O. Rohof et al.

Pharmacodynamic results: effect of AZD2066 onoesophageal pH. In part A of the study, time withoesophageal pH <4 was reduced when participantsreceived AZD2066 13 mg compared with when theyreceived placebo, although this effect was not significant(Table 1). In part B of the study, no effect relative toplacebo was observed for AZD2066 6 mg (Table 2) andAZD2066 2 mg (Table 3) on time with oesophagealpH <4.

Pharmacodynamic results: effect of AZD2066 on LESpressure and swallowing. There were no significant dif-ferences relative to placebo in the mean LES pressure orin the mean number of swallows for any of the dose reg-imens investigated (Tables 1–3).

Sequence, period and carry-over effects. For the refluxvariables assessed, no significant sequence or periodeffects were detected, thus indicating an absence ofcarry-over effects. A significant sequence effect wasdetected for AZD2066 13 mg in relation to TLESRs inpart A of the study (P = 0.02), indicating that sequence,period and/or carry-over effects may have occurred.However, the sequence effect was accounted for in theANOVA model used to calculate the statistical significanceof the treatment effect. Furthermore, when results ofonly the first treatment period were analysed, thusreducing the sample size [(AZD2066 13 mg: n = 7; pla-cebo: n = 6) but eliminating potential sequence, periodand/or carryover effects, the impact of AZD2066 13 mg

on TLESRs was still significant (GMR, 0.49; 95% CI,0.28–0.88); P = 0.02].

Pharmacokinetic results. The plasma concentration ofAZD2066 decreased at a similar rate over time for allthree doses (Figure 6). All doses were absorbed rapidly,with median time to maximum concentration values of0.8 h (95% CI, 0.3–1.5) for AZD2066 13 mg, 0.5 h (95%CI, 0.3– 2.8) for AZD2066 6 mg and 0.5 h (95% CI,0.3–6.0) for AZD2066 2 mg.

Safety and tolerability assessments. The adverse eventsexperienced during the study are reported in Table 4. Alladverse events were mild in intensity and reversible. Noserious adverse events were reported during the active

AZD2066 2 mg (n = 19)AZD2066 6 mg (n = 19)AZD2066 13 mg (n = 13)

1250

1000

750

500

250

00 2 4 6

Time after dose (h)

Arit

hmet

ic m

ean

plas

ma

conc

entr

atio

n (n

mol

/L)

8 10 12

Figure 6 | Mean plasma concentration of AZD2066during the 12 h after taking 2, 6 or 13 mg doses of thedrug.

Table 4 | Number of participants with one or more adverse events during active dosing

Part A Part B

Placebo(n = 13)

AZD2066 13 mg(n = 13)

Placebo(n = 20)

AZD2066 6 mg(n = 19)

AZD2066 2 mg(n = 20)

Any adverse event 3 8 5 5 4Serious adverse event 0 0 0 0 0Discontinuation due to an adverse event 0 0 0 0 0Headache 0 1 3 3 3Dizziness 0 3 0 1 1Disturbance in attention 0 3 0 0 0Somnolence 1 1 0 0 0Fatigue 1 1 1 0 1Feeling cold 0 1 0 0 0Increase in blood Bilirubin 1 1 0 0 0Epistaxis 0 0 0 1 0Oropharyngeal pain 0 0 1 0 0Flank pain 0 0 1 0 0Haematuria 0 0 1 0 0Erection increased 0 1 0 0 0

Aliment Pharmacol Ther 2012; 35: 1231-1242 1239

ª 2012 Blackwell Publishing Ltd

Effects of AZD2066 on TLESRs and reflux

dosing periods, and there were no discontinuations dueto adverse events.

In part A of the study, eight of the 13 participantsreported an adverse event when they received AZD206613 mg compared with three when they received placebo.Most of the adverse events reported when participantswere receiving AZD2066 13 mg were related to the ner-vous system, the most common being dizziness (3/13)and disturbance in attention (3/13). Only one nervoussystem disorder (somnolence) was reported when partici-pants were receiving placebo. In part B of the study, thenumber of participants reporting adverse events wassimilar for AZD2066 2 mg (5/20) and AZD2066 6 mg(4/19) compared with placebo (5/20). Headache was themost common adverse event reported during dosing withAZD2066 2 mg and AZD2066 6 mg (3/20 and 3/19respectively), and the same number of headaches wasreported when these participants were taking placebo(3/20).

DISCUSSIONIn this randomised, double-blind, crossover study con-ducted in healthy volunteers, a single oral 13 mg dose ofAZD2066 was found to reduce the numbers of TLESRs,reflux episodes and acid reflux episodes significantlycompared with placebo.

The overall frequency of TLESRs was greater duringthe 3-h postprandial period than in the preprandial per-iod, and differences in the number of TLESRs and refluxepisodes between AZD2066 and placebo were generallymore pronounced during the postprandial period. Theseobservations are consistent with previous findings thatTLESRs can be activated by food intake via stretchreceptors in the stomach.15

A dose-dependent effect was seen with AZD2066: onlythe AZD2066 13 mg dose significantly reduced the geo-metric mean number of TLESRs (27%) and geometricmean number of reflux episodes (51%) compared withplacebo, and there was a trend towards a greater effectfor AZD2066 6 mg than for AZD2066 2 mg. A possibleexception to the dose-dependent effects of AZD2066 wasthe counterintuitive observation of a significant reductionrelative to placebo in mixed gas/liquid reflux when par-ticipants received AZD2066 2 mg and AZD2066 6 mg,but not when they received the higher AZD2066 13 mgdose. However, a similar reduction in the absolute num-ber of mixed gas/liquid reflux episodes relative to pla-cebo was observed for AZD2066 13 mg and AZD20666 mg, both of which reduced mixed gas/liquid reflux toa greater extent that AZD2066 2 mg. The lack of a sta-

tistically significant effect for AZD2066 13 mg in relationto mixed gas/liquid reflux may have been because of thesmaller sample size used in part A of the study(AZD2066 13 mg n = 13) vs. part B (AZD2066 2 mg:n = 19; AZD2066 6 mg: n = 16).

This is not the first study to show that pharmacologi-cal inhibition of TLESRs reduces the number of refluxepisodes in humans. Activation of inhibitory GABAB

receptors using baclofen, a drug indicated for spasticity,has been shown to reduce the number of TLESRs andreflux episodes in healthy volunteers and in patients withGERD.26–28 In more recent studies, inhibition of TLESRsusing the novel GABAB agonist lesogaberan reduced thenumber of reflux episodes in healthy men and inpatients with symptomatic GERD despite PPI treat-ment.18, 19 The present study is, however, the first todemonstrate that the reduced frequency of reflux epi-sodes previously shown to be associated with inhibitionof mGluR5 in humans is related to the ability to reducethe number of TLESRs.

Interestingly, AZD2066 13 mg reduced the geometricmean number of reflux episodes in this study by 51%,despite a reduction in the geometric mean number ofTLESRs of only 27%. Indeed, the number of TLESRstemporally related to reflux episodes was reduced(although not significantly) in participants receivingAZD2066 13 mg compared with those receiving placebo(43% vs. 56%). Lesogaberan has also been shown toreduce the number of reflux episodes to a greater extentthan the number of TLESRs. In the case of lesogaberan,this difference may be partially explained by a concomi-tant increase in mean LES pressure, which has beenshown to reduce reflux via a mechanism that is unre-lated to TLESRs.13 However, AZD2066 was not found toincrease LES pressure in the current study, and themechanism by which AZD2066 reduces the number ofTLESRs generating reflux remains unclear. One possibil-ity is that some TLESRs were not detected in the currentstudy. Although this is consistent with the observationthat only 65–80% of reflux episodes were temporallyrelated to TLESRs, it is difficult to envisage why a smal-ler proportion of TLESRs would be detected in partici-pants receiving AZD2066 13 mg than in those receivingplacebo. It is also possible that inhibition of mGluR5may influence both the quantity and quality of TLESRs.Thus, in the presence of AZD2066, LES relaxations mayoccur that meet the criteria for a TLESR but do notgenerate reflux with sufficient properties to meet thedefinition of a reflux episode (i.e. a drop in impedanceto <50% of baseline). Other risk factors for acid reflux,

1240 Aliment Pharmacol Ther 2012; 35: 1231-1242

ª 2012 Blackwell Publishing Ltd

W. O. Rohof et al.

such as the position of the acid pocket,29 may also beaffected by blocking mGluR5, although to our knowledgeno studies have investigated this.

Another interesting observation from this study wasthat acid reflux, and not weakly acidic or weakly alkalinereflux, was significantly reduced in participants receivingAZD2066 compared with those receiving placebo. A sim-ilarly disproportionate effect on acid reflux has been seenfor both lesogaberan and baclofen.18, 27 It has been pos-tulated that this may be due to the greater effect of thesedrugs on liquid and mixed liquid/gas reflux comparedwith pure gas reflux, the former being more often associ-ated with acid reflux.18 In this study, AZD2066 13 mgreduced liquid and mixed liquid/gas reflux more thanpure gas reflux. However, pure gas reflux made up a verysmall proportion (~6%) of the total number of refluxepisodes observed in this study. It therefore seems unli-kely that the inability of AZD2066 to inhibit pure gasreflux could account for its lack of effect on weaklyacidic reflux, which accounted for approximately onethird of the total number of reflux episodes. There issome evidence to suggest that GABAB agonists may alsoalter the amount and distribution of acid in the stom-ach.29, 30, 31 Given that GABAB and mGluR5 receptorsare both involved in regulating TLESRs, it is conceivablethat mGluR5 may also help regulate digestive compo-nents in the stomach, such as acid. It should also benoted that the proportion of weakly acidic reflux epi-sodes that were unrelated to TLESRs increased from13.6% when patients received placebo to 38.9% whenpatients received AZD2066 13 mg. The lack of effect ofAZD2066 13 mg on weakly acidic reflux may thus beattributed to an increase in the proportion of theseevents that occur independently of TLESRs, although themechanism behind this is unclear.

In conclusion, the novel mGluR5 antagonistAZD2066 reduces the number of postprandial reflux

episodes that occur in healthy individuals, and thiseffect is correlated with a reduction in the frequency ofTLESRs. In addition, AZD2066 appeared to have anacceptable safety and tolerability profile in this study.However, it should be noted that the GABAB-receptoragonist lesogaberan was previously shown to reduce thegeometric mean number of TLESRs in healthy volun-teers to a greater extent (36%) than AZD2066 did inthe current study.19 Lesogaberan has since been discon-tinued owing to a lack of evidence for a clinically sig-nificant effect as an add-on to PPI therapy in patientswith reflux symptoms despite PPI treatment. Neverthe-less, the results of the current study reinforce theimportance of TLESRs in generating reflux and provideanother potential avenue for the development of noveltherapeutic agents for GERD (ClinicalTrials.gov Identi-fier: NCT00813306).

ACKNOWLEDGEMENTSDeclaration of personal interests: L. Ny, M. Astrandand M. B Hansen are/were employees of AstraZenecaR&D, Mölndal, Sweden. Declaration of fundinginterests: This study was funded by AstraZeneca R&D,Mölndal, Sweden. Writing support was provided by DrMichael Molloy-Bland of Oxford PharmaGenesis Ltdand was funded by AstraZeneca R&D, Mölndal,Sweden.

SUPPORTING INFORMATIONAdditional Supporting Information may be found in theonline version of this article:Table S1. Study inclusion and exclusion criteria.Please note: Wiley-Blackwell are not responsible for

the content or functionality of any supporting materialssupplied by the authors. Any queries (other than missingmaterial) should be directed to the corresponding authorfor the article.

REFERENCES1. Armstrong D. Systematic review:

persistence and severity ingastroesophageal reflux disease.Aliment Pharmacol Ther 2008; 28:841–53.

2. Vakil N, Veldhuyzen van Zanten S,Kahrilas P, et al. The Montrealdefinition and classification ofgastro-esophageal reflux disease(GERD) – a global evidence-based

consensus. Am J Gastroenterol 2006;101: 1900–20.

3. Dent J, El-Serag HB, Wallander MA,et al. Epidemiology of gastro-oesophageal reflux disease: asystematic review. Gut 2005; 54:710–17.

4. Wong BCY, Kinoshita Y. Systematicreview on epidemiology ofgastroesophageal reflux disease in Asia.

Clin Gastroenterol Hepatol 2006; 4: 398–407.

5. Dent J. Review article: from 1906 to2006 – a century of major evolution ofunderstanding of gastro-oesophagealreflux disease. Aliment Pharmacol Ther2006; 24: 1269–81.

6. van Pinxteren B, Numans ME, BonisPA, et al. Short-term treatment withproton pump inhibitors, H2-receptor

Aliment Pharmacol Ther 2012; 35: 1231-1242 1241

ª 2012 Blackwell Publishing Ltd

Effects of AZD2066 on TLESRs and reflux

antagonists and prokinetics for gastro-oesophageal reflux disease-likesymptoms and endoscopy negativereflux disease. Cochrane Database SystRev 2001; 4: CD002095.

7. Armstrong D, Pare P, Pericak D, et al.Symptom relief in gastroesophagealreflux disease: a randomized, controlledcomparison of pantoprazole andnizatidine in a mixed patientpopulation with erosive esophagitis orendoscopy-negative reflux disease. Am JGastroenterol 2001; 96: 2849–57.

8. Fass R. Proton-pump inhibitor therapyin patients with gastro-oesophagealreflux disease: putative mechanisms offailure. Drugs 2007; 67: 1521–30.

9. Hatlebakk JG, Hyggen A, Madsen PH,et al. Heartburn treatment in primarycare: randomised, double blind studyfor 8 weeks. BMJ 1999; 319: 550–3.

10. Meineche-Schmidt V, Hauschildt JuhlH, Ostergaard JE, et al. Costs andefficacy of three different esomeprazoletreatment strategies for long-termmanagement of gastro-oesophagealreflux symptoms in primary care.Aliment Pharmacol Ther 2004; 19: 907–15.

11. El-Serag H, Becher A, Jones R.Systematic review: persistent refluxsymptoms on proton pump inhibitortherapy in primary care andcommunity studies. Aliment PharmacolTher 2010; 32: 720–37.

12. Dent J, Dodds WJ, Friedman RH, et al.Mechanism of gastroesophageal refluxin recumbent asymptomatic humansubjects. J Clin Invest 1980; 65:256–67.

13. Dodds WJ, Dent J, Hogan WJ, et al.Mechanisms of gastroesophageal refluxin patients with reflux esophagitis. NEngl J Med 1982; 307: 1547–52.

14. Boeckxstaens GE, Smout A. Systematicreview: role of acid, weakly acidic andweakly alkaline reflux ingastroesophageal reflux disease. AlimentPharmacol Ther 2010; 32: 334–43.

15. Penagini R, Carmagnola S, Cantu P,et al. Mechanoreceptors of theproximal stomach: role in triggeringtransient lower esophageal sphincterrelaxation. Gastroenterology 2004; 126:49–56.

16. Blackshaw LA. Receptors andtransmission in the brain-gut axis:potential for novel therapies. IV. GABA(B) receptors in the brain-gastroesophageal axis. Am J PhysiolGastrointest Liver Physiol 2001; 281:G311–5.

17. Hirsch DP, Tytgat GN, BoeckxstaensGE. Transient lower oesophagealsphincter relaxations – apharmacological target for gastro-oesophageal reflux disease? AlimentPharmacol Ther 2002; 16: 17–26.

18. Boeckxstaens GE, Beaumont H,Mertens V, et al. Effects of lesogaberanon reflux and lower esophagealsphincter function in patients withgastroesophageal reflux disease.Gastroenterology 2010; 139: 409–17.

19. Boeckxstaens GE, Rydholm H, Lei A,et al. Effect of lesogaberan, a novelGABA(B)-receptor agonist, on transientlower esophageal sphincter relaxationsin male subjects. Aliment PharmacolTher 2010; 31: 1208–17.

20. Young RL, Page AJ, O’Donnell TA,et al. Peripheral versus centralmodulation of gastric vagal pathwaysby metabotropic glutamate receptor 5.Am J Physiol Gastrointest Liver Physiol2007; 292: G501–11.

21. Frisby CL, Mattsson JP, Jensen JM,et al. Inhibition of transient loweresophageal sphincter relaxation andgastroesophageal reflux by metabotropicglutamate receptor ligands.Gastroenterology 2005; 129: 995–1004.

22. Jensen J, Lehmann A, Uvebrant A,et al. Transient lower esophagealsphincter relaxations in dogs areinhibited by a metabotropic glutamatereceptor 5 antagonist. Eur J Pharmacol2005; 519: 154–7.

23. Keywood C, Wakefield M, Tack J. Aproof of concept study evaluating theeffect of ADX10059, a metabotropicglutamate receptor-5 negative allostericmodulator, on acid exposure andsymptoms in gastro-esophageal refluxdisease. Gut 2009; 58: 1192–9.

24. Holloway RH, Penagini R, Ireland AC.Criteria for objective definition oftransient lower esophageal sphincterrelaxation. Am J Physiol 1995; 268:G128–33.

25. Senn S. Cross-over Trials in ClinicalResearch. 2nd edn. Chichester: Wiley,2002.

26. Lidums I, Lehmann A, Checklin H,et al. Control of transient loweresophageal sphincter relaxations andreflux by the GABA(B) agonist baclofenin normal subjects. Gastroenterology2000; 118: 7–13.

27. Vela MF, Tutuian R, Katz PO, et al.Baclofen decreases acid and non-acidpost-prandial gastro-oesophageal refluxmeasured by combined multichannelintraluminal impedance and pH.Aliment Pharmacol Ther 2003; 17: 243–51.

28. Zhang Q, Lehmann A, Rigda R, et al.Control of transient lower oesophagealsphincter relaxations and reflux by theGABA(B) agonist baclofen in patientswith gastro-oesophageal reflux disease.Gut 2002; 50: 19–24.

29. Beaumont H, Bennink RJ, de Jong J,et al. The position of the acid pocket asa major risk factor for acidic reflux inhealthy subjects and patients withGORD. Gut 2010; 59: 441–51.

30. del Genio G, del Genio A, Brusciano L,et al. Laparoscopic cardioplasty toavoid esophageal resection in patientnot responsive to Heller myotomy. AnnThorac Surg 2007; 83: 2235–8.

31. Del Tacca M, Blandizzi C, BernardiniMC. Central GABAA excitatory andGABAB inhibitory receptors regulategastric acid secretion in rats. Eur JPharmacol 1990; 177: 189–94.

1242 Aliment Pharmacol Ther 2012; 35: 1231-1242

ª 2012 Blackwell Publishing Ltd

W. O. Rohof et al.