1
|
MAZUMDER AVIJIT, KUMAR NAVEEN, DAS SAUMYA. A Comprehensive Review of Gastroesophageal Reflux Disease (GERD) Treatment and its Clinical Perspectives. INTERNATIONAL JOURNAL OF PHARMACEUTICAL SCIENCES AND NANOTECHNOLOGY(IJPSN) 2023; 16:7093-7103. [DOI: 10.37285/ijpsn.2023.16.6.9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Gastroesophageal reflux disease (GERD) occurs by regurgitation of food in the stomach. Aggressive factors increase GERD whereas defensive factors decrease GERD progression. GERD if mild can be put under control by lifestyle modification and giving non-pharmacological treatment methods to patients however if the disease progresses non-pharmacological methods are ineffective. Drugs reduce GERD progression and also maintain the pH of the stomach to a normal level and prevent abnormal acid exposure to the oesophagus. Antacids and alginate protect oesophagus by reducing acidity and increasing viscosity. Proton pump inhibitors and histamine 2 receptor antagonists reduce acid secretion by inhibiting its secretion. Prokinetic agents increase the motility of the stomach and reduce obesity. Metabotropic glutamate receptors, gamma-aminobutyric acid receptor agonists, and cannabinoid receptors are receptor-specific drugs that act on receptors underlying the gastrointestinal tract and alter the function of receptors which increases reflux disease. Combination of antacid and alginate, domperidone and omeprazole, omeprazole and baclofen, aluminum hydroxide, magnesium, and simethicone are frequently given in GERD to expedite the healing rate and reduce acid secretion. Combinations of suitable medications reduce the adverse effects of a single medication and also make it therapeutically more effective than using monotherapy drugs. The pharmacological method is safe and effective and treats GERD completely.
Collapse
|
2
|
Badri H, Gibbard C, Denton D, Satia I, Al-Sheklly B, Dockry RJ, Holt K, McGuiness K, Treadway S, Whorwell P, Houghton L, Lee A, Escott KJ, Lee T, Wilkinson G, Holt A, Canning BJ, Smith JA. A double-blind randomised placebo-controlled trial investigating the effects of lesogaberan on the objective cough frequency and capsaicin evoked coughs in patients with refractory chronic cough. ERJ Open Res 2022; 8:00546-2021. [PMID: 35295236 PMCID: PMC8918934 DOI: 10.1183/23120541.00546-2021] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 01/29/2022] [Indexed: 11/23/2022] Open
Abstract
Objective Baclofen is a centrally acting γ-aminobutyric acid type B (GABAB) receptor agonist which reduces gastro-oesophageal reflux and suppresses the cough reflex; however, central nervous system side-effects limit its use. Lesogaberan is a novel peripherally acting GABAB agonist, but its effects on refractory chronic cough are unknown. Design We performed a single-centre, placebo-controlled, double-blind randomised crossover study in patients with chronic cough, refractory to the treatment of underlying conditions. Patients were randomised to treatment with lesogaberan 120 mg modified release twice daily or matched placebo for 2 weeks and then crossed over to the alternative therapy after a 2-week washout. The primary end-point was 24-h cough frequency measured with an acoustic monitoring system. In addition, cough responses to capsaicin were measured, and gastro-oesophageal reflux assessed by 24-h pH/impedance at screening. Results 22 patients were randomised to receive lesogaberan/placebo or placebo/lesogaberan (female (73%); mean±sd age 63.7±7.2 years; median (interquartile range) cough duration 10.5 (5.8–17.0) years; mean (95% CI) 45 (29–67) reflux events in 24 h; two patients had abnormal oesophageal acid exposure times). Although lesogaberan reduced cough counts by 26% over placebo, this did not reach statistical significance (p=0.12). However, lesogaberan did significantly improve cough responses to capsaicin (p=0.04) and the number of cough bouts (p=0.04) compared with placebo. Lesogaberan was well tolerated in this study. Conclusions Lesogaberan improved cough hypersensitivity and the number of bouts of coughing, but not coughs per hour. This implies a possible role for peripheral GABAB receptors in refractory chronic cough. Lesogaberan, a peripherally acting GABAB agonist, does not reduce 24-h cough frequency in patients with chronic cough despite significantly reducing capsaicin-induced coughinghttps://bit.ly/3uGyPQL
Collapse
|
3
|
GABA B Receptor Chemistry and Pharmacology: Agonists, Antagonists, and Allosteric Modulators. Curr Top Behav Neurosci 2021; 52:81-118. [PMID: 34036555 DOI: 10.1007/7854_2021_232] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The GABAB receptors are metabotropic G protein-coupled receptors (GPCRs) that mediate the actions of the primary inhibitory neurotransmitter, γ-aminobutyric acid (GABA). In the CNS, GABA plays an important role in behavior, learning and memory, cognition, and stress. GABA is also located throughout the gastrointestinal (GI) tract and is involved in the autonomic control of the intestine and esophageal reflex. Consequently, dysregulated GABAB receptor signaling is associated with neurological, mental health, and gastrointestinal disorders; hence, these receptors have been identified as key therapeutic targets and are the focus of multiple drug discovery efforts for indications such as muscle spasticity disorders, schizophrenia, pain, addiction, and gastroesophageal reflex disease (GERD). Numerous agonists, antagonists, and allosteric modulators of the GABAB receptor have been described; however, Lioresal® (Baclofen; β-(4-chlorophenyl)-γ-aminobutyric acid) is the only FDA-approved drug that selectively targets GABAB receptors in clinical use; undesirable side effects, such as sedation, muscle weakness, fatigue, cognitive deficits, seizures, tolerance and potential for abuse, limit their therapeutic use. Here, we review GABAB receptor chemistry and pharmacology, presenting orthosteric agonists, antagonists, and positive and negative allosteric modulators, and highlight the therapeutic potential of targeting GABAB receptor modulation for the treatment of various CNS and peripheral disorders.
Collapse
|
4
|
Bagheri Lankarani K, Sivandzadeh GR, Zare M, Nejati M, Niknam R, Taghavi AR, Ejtehadi F, Naini MA, Moini M, Anbardar MH, Peymani P. A preliminary report on the use of Midodrine in treating refractory gastroesophageal disease: Randomized Double-Blind Controlled Trial. ACTA BIO-MEDICA : ATENEI PARMENSIS 2020; 91:70-78. [PMID: 32191657 PMCID: PMC7569571 DOI: 10.23750/abm.v91i1.8486] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 02/05/2020] [Indexed: 02/08/2023]
Abstract
Background: Gastroesophageal reflux disease (GERD) is a common disease with various clinical presentations. Acid suppression with proton pump inhibitors and lifestyle modification may not lead to satisfactory response in a substantial portion of patients. We investigated the possible effect of midodrine in patients with refractory GERD. Methods: Patients suffering from GERD and were refractory to one-month course of pantoprazole 40mg twice daily entered the study. This was a pilot, randomized, double-blind, and placebo-controlled study. After randomization, one group received Midodrine 5mg before meals for one month, and the other group received placebo for the same period. Meanwhile, pantoprazole was continued 40mg twice daily in both arms. The severity of symptoms was evaluated by the visual scoring system. Quality of life (QoL) in both groups was measured using a standardized version of Quality of Life in Reflux and Dyspepsia questionnaire (QOLRAD). Results: A total of twenty patients were enrolled in this study. There was a significant interaction between the groups and time on all measured scores based on QOLRAD questionnaire. All the markers in the Midodrine group had significant improvement over time, but the placebo group did not show any significant improvement. Both visual severity score and total QoL score in Midodrine arm showed a U shape change during 6 weeks. Conclusions: Midodrine before a meal could be useful in alleviating symptoms and improving QoL in the patients with refractory gastroesophageal disease. (www.actabiomedica.it)
Collapse
Affiliation(s)
| | | | - Marziyeh Zare
- Health Policy Research Center, Institute of Health, Shiraz University of Medical Sciences.
| | | | | | | | | | | | | | | | - Payam Peymani
- Health Policy Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Fars, Iran..
| |
Collapse
|
5
|
Canals M, Poole DP, Veldhuis NA, Schmidt BL, Bunnett NW. G-Protein-Coupled Receptors Are Dynamic Regulators of Digestion and Targets for Digestive Diseases. Gastroenterology 2019; 156:1600-1616. [PMID: 30771352 PMCID: PMC6508858 DOI: 10.1053/j.gastro.2019.01.266] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 12/15/2018] [Accepted: 01/08/2019] [Indexed: 01/11/2023]
Abstract
G-protein-coupled receptors (GPCRs) are the largest family of transmembrane signaling proteins. In the gastrointestinal tract, GPCRs expressed by epithelial cells sense contents of the lumen, and GPCRs expressed by epithelial cells, myocytes, neurons, and immune cells participate in communication among cells. GPCRs control digestion, mediate digestive diseases, and coordinate repair and growth. GPCRs are the target of more than one third of therapeutic drugs, including many drugs used to treat digestive diseases. Recent advances in structural, chemical, and cell biology research have shown that GPCRs are not static binary switches that operate from the plasma membrane to control a defined set of intracellular signals. Rather, GPCRs are dynamic signaling proteins that adopt distinct conformations and subcellular distributions when associated with different ligands and intracellular effectors. An understanding of the dynamic nature of GPCRs has provided insights into the mechanism of activation and signaling of GPCRs and has shown opportunities for drug discovery. We review the allosteric modulation, biased agonism, oligomerization, and compartmentalized signaling of GPCRs that control digestion and digestive diseases. We highlight the implications of these concepts for the development of selective and effective drugs to treat diseases of the gastrointestinal tract.
Collapse
Affiliation(s)
- Meritxell Canals
- Centre for Membrane Proteins and Receptors (COMPARE), School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Daniel P. Poole
- Monash Institute of Pharmaceutical Sciences and Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria, Australia,Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria, Australia
| | - Nicholas A. Veldhuis
- Monash Institute of Pharmaceutical Sciences and Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria, Australia
| | - Brian L. Schmidt
- Bluestone Center for Clinical Research, New York University College of Dentistry, New York, New York
| | - Nigel W. Bunnett
- Monash Institute of Pharmaceutical Sciences and Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria, Australia,Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Victoria, Australia,Columbia University College of Physicians and Surgeons, Columbia University, New York, New York
| |
Collapse
|
6
|
Scarpellini E, Boecxstaens V, Broers C, Vos R, Pauwels A, Tack J. Effect of baclofen on gastric acid pocket in subjects with gastroesophageal reflux disease symptoms. Dis Esophagus 2016; 29:1054-1063. [PMID: 26541138 DOI: 10.1111/dote.12443] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Postprandial gastroesophageal reflux (PGER) in the distal esophagus (DE) is associated with a gastric juice 'acid pocket' (AP). Baclofen reduces AP extension into the DE in healthy volunteers, in part through increased lower esophageal sphincter (LES) pressure. We aimed to verify whether baclofen also affects postprandial AP location and extent in gastroesophageal reflux disease (GERD) patients. Thirteen treatment-naive heartburn-prevalent GERD patients underwent two AP studies, after pretreatment with baclofen 40 mg or placebo 30 minutes preprandially. We performed pH-probe stepwise pull-throughs (PT) (1 cm/min, LES -10 to +5 cm) before and every 30 minutes from 30 minutes before up to 150 minutes after a test meal. After the meal, both after placebo and baclofen, gastric pH significantly dropped at 30, 60, 90 minutes postprandially (P: nadir pHs of 3.9 ± 0.6, 2.3 ± 0.6, 2.1 ± 0.4; B: nadir pHs of 2.5 ± 0.4, 2.8 ± 0.4, 2.5 ± 0.3; all P < 0.05). After placebo, LES pressure decreased at 60, 90 and 120 minutes postprandially (32.7 ± 6.1 vs. 24.5 ± 3.1, 27.3 ± 5.9, 27.3 ± 6.0 mmHg; analysis of variance [ANOVA], P = 0.037), but this was prevented by baclofen (25.4 ± 3.4 vs. 29.4 ± 2, 32.2 ± 1.4, 35.5 ± 1.7 mmHg, ANOVA, P = not significant (NS)). Baclofen did not significantly decrease the postprandial AP extent above the LES but prevented the postprandial increase in transient lower esophageal sphincter relaxations (TLESRs) (preprandial vs. postprandial, placebo: 1.1 ± 0.3 vs. 3.7 ± 0.7, P < 0.05; baclofen: 1.4 ± 0.4 vs. 2 ± 0.5, P = NS). In GERD patients, baclofen significantly increases postprandial LES pressure, prevents the increase TLESRs but, unlike in healthy volunteers, does not affect AP extension into the DE.
Collapse
Affiliation(s)
- E Scarpellini
- TARGID (Translational Research Centre for Gastrointestinal Disorders), University of Leuven, Leuven, Belgium
| | - V Boecxstaens
- TARGID (Translational Research Centre for Gastrointestinal Disorders), University of Leuven, Leuven, Belgium
| | - C Broers
- TARGID (Translational Research Centre for Gastrointestinal Disorders), University of Leuven, Leuven, Belgium
| | - R Vos
- TARGID (Translational Research Centre for Gastrointestinal Disorders), University of Leuven, Leuven, Belgium
| | - A Pauwels
- TARGID (Translational Research Centre for Gastrointestinal Disorders), University of Leuven, Leuven, Belgium
| | - J Tack
- TARGID (Translational Research Centre for Gastrointestinal Disorders), University of Leuven, Leuven, Belgium
| |
Collapse
|
7
|
Doly S, Marullo S. Gatekeepers Controlling GPCR Export and Function. Trends Pharmacol Sci 2016; 36:636-644. [PMID: 26435209 DOI: 10.1016/j.tips.2015.06.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Revised: 06/15/2015] [Accepted: 06/25/2015] [Indexed: 12/17/2022]
Abstract
Regulated export of G protein-coupled receptors (GPCRs) from intracellular stores involves chaperones and escort proteins, which promote their progression to the cell surface, and gatekeepers, which retain them in intracellular compartments. Functional γ-aminobutyric acid (GABA)B receptors, the paradigm of this phenomenon, comprise GB1 and GB2 subunits forming a heterodimer. GB1 is retained in the endoplasmic reticulum (ER) in the absence of GB2. A specific ER-resident gatekeeper, prenylated Rab acceptor family 2 (PRAF2), is involved in GB1 retention and prevents its progression into the biosynthetic pathway. GB1 can be released from PRAF2 only on competitive interaction with GB2. PRAF2 is ubiquitous and belongs to a subgroup of the mammalian Ypt-interacting protein (Yip) family. Several other GPCRs are likely to be regulated by Yip proteins, which might be involved in the pathophysiology of human diseases that are associated with impaired receptor targeting to the cell surface.
Collapse
Affiliation(s)
- Stéphane Doly
- Institut Cochin, INSERM, CNRS, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Stefano Marullo
- Institut Cochin, INSERM, CNRS, Université Paris Descartes, Sorbonne Paris Cité, Paris, France.
| |
Collapse
|
8
|
Scarpellini E, Boecxstaens V, Farré R, Bisschops R, Dewulf D, Gasbarrini A, Pauwels A, Blondeau K, Tack J. Effect of baclofen on the acid pocket at the gastroesophageal junction. Dis Esophagus 2015; 28:488-95. [PMID: 24758736 DOI: 10.1111/dote.12224] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Previous studies established that a pocket of highly acidic gastric juice is present postprandially at the gastroesophageal junction in man. The GABA-B agonist baclofen inhibits postprandial reflux events through its effects on the lower esophageal sphincter (LES). The aim of the current study was to investigate whether baclofen would affect the location and the extent of the postprandial acid pocket in healthy volunteers. Twelve healthy volunteers underwent acid pocket studies on two different occasions, at least 1 week apart. LES position was determined preprandially with pull-through manometry. Dual pH electrode and manometry probe stepwise pull-through (1 cm/minute, LES-10 to +5 cm) was performed at 30-minute intervals for 150 minutes, with administration of placebo or baclofen 40 mg after the first and ingestion of a liquid meal after the second pull-through. After placebo, a significant drop in intragastric gastric pH was present at the gastroesophageal junction after the meal, reflecting the acid pocket, and this was associated with a drop in LES pressure. Baclofen did not affect the presence of the acid pocket, but prevented the postprandial drop in LES pressure, and the extent of the acid pocket above the upper margin of the manometrically located LES was significantly decreased by baclofen (1.6 ± 0.7 vs. 0.3 ± 0.4 cm at 60 minutes, 2.2 ± 0.6 vs. 0.2 ± 0.6 at 90 minutes, and 1.5 ± 0.5 vs. 0.7 ± 0.7 cm at 120 minutes, all P < 0.05). Baclofen does not alter the intragastric acid pocket, but limits its extension into the distal esophagus, probably through an increase in postprandial LES pressure.
Collapse
Affiliation(s)
- E Scarpellini
- Department of Internal Medicine, Division of Gastroenterology, University Hospital Gasthuisberg, Catholic University of Leuven, Leuven, Belgium
| | - V Boecxstaens
- Department of Internal Medicine, Division of Gastroenterology, University Hospital Gasthuisberg, Catholic University of Leuven, Leuven, Belgium
| | - R Farré
- Department of Internal Medicine, Division of Gastroenterology, University Hospital Gasthuisberg, Catholic University of Leuven, Leuven, Belgium
| | - R Bisschops
- Department of Internal Medicine, Division of Gastroenterology, University Hospital Gasthuisberg, Catholic University of Leuven, Leuven, Belgium
| | - D Dewulf
- Department of Internal Medicine, Division of Gastroenterology, University Hospital Gasthuisberg, Catholic University of Leuven, Leuven, Belgium
| | - A Gasbarrini
- Department of Internal Medicine, Division of Gastroenterology, University Hospital Gasthuisberg, Catholic University of Leuven, Leuven, Belgium
| | - A Pauwels
- Department of Internal Medicine, Division of Gastroenterology, University Hospital Gasthuisberg, Catholic University of Leuven, Leuven, Belgium
| | - K Blondeau
- Department of Internal Medicine, Division of Gastroenterology, University Hospital Gasthuisberg, Catholic University of Leuven, Leuven, Belgium
| | - J Tack
- Department of Internal Medicine, Division of Gastroenterology, University Hospital Gasthuisberg, Catholic University of Leuven, Leuven, Belgium
| |
Collapse
|
9
|
Brown KM, Roy KK, Hockerman GH, Doerksen RJ, Colby DA. Activation of the γ-Aminobutyric Acid Type B (GABA(B)) Receptor by Agonists and Positive Allosteric Modulators. J Med Chem 2015; 58:6336-47. [PMID: 25856547 DOI: 10.1021/jm5018913] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Since the discovery of the GABA(B) agonist and muscle relaxant baclofen, there have been substantial advancements in the development of compounds that activate the GABA(B) receptor as agonists or positive allosteric modulators. For the agonists, most of the existing structure-activity data apply to understanding the role of substituents on the backbone of GABA as well as replacing the carboxylic acid and amine groups. In the cases of the positive allosteric modulators, the allosteric binding site(s) and structure-activity relationships are less well-defined; however, multiple classes of molecules have been discovered. The recent report of the X-ray structure of the GABA(B) receptor with bound agonists and antagonists provides new insights for the development of compounds that bind the orthosteric site of this receptor. From a therapeutic perspective, these data have enabled efforts in drug discovery in areas of addiction-related behavior, the treatment of anxiety, and the control of muscle contractility.
Collapse
Affiliation(s)
- Katie M Brown
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University , West Lafayette, Indiana 47907, United States
| | - Kuldeep K Roy
- Department of Biomolecular Sciences, University of Mississippi , University, Mississippi 38677, United States
| | - Gregory H Hockerman
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University , West Lafayette, Indiana 47907, United States
| | - Robert J Doerksen
- Department of Biomolecular Sciences, University of Mississippi , University, Mississippi 38677, United States
| | - David A Colby
- Department of Biomolecular Sciences, University of Mississippi , University, Mississippi 38677, United States
| |
Collapse
|
10
|
|
11
|
Abstract
Acid suppression, with proton pump inhibitors (PPI), is the mainstay of therapy for reflux disease; despite this, symptoms and injury persist in many patients. Novel approaches have focused on (1) augmenting acid suppression with reformulated current PPIs, new PPIs or new acid pump blockers and (2) preventing reflux with reflux inhibitors. Other strategies to reduce reflux, improve gastric emptying or esophageal clearance, protect the mucosa, or reduce esophageal sensitivity are under investigation alone or in combination with PPIs; however, novel approaches face significant challenges. The safety and efficacy of current PPIs hamper demonstration of clinical superiority for new acid suppressants, and the multifactorial etiology of reflux disease means that monotherapy using a non-acid suppressant is unlikely to match PPI therapy while combination therapy will be superior only if susceptible patients can be identified reliably. Advances will come, not from a 'one size fits all' approach but rather from novel pharmaceuticals allied to novel investigations to permit targeted, personalized reflux therapy.
Collapse
Affiliation(s)
- Usha Dutta
- Division of Gastroenterology, Department of Medicine, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4K1, Canada
| | | |
Collapse
|
12
|
Craig OF, Quigley EMM. Current and emerging therapies for the management of functional gastrointestinal disorders. Ther Adv Chronic Dis 2012; 2:87-99. [PMID: 23251744 DOI: 10.1177/2040622310389507] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The functional gastrointestinal disorders are common disorders that are associated with significant quality-of-life impairment and considerable economic burden on the healthcare system. They are frequently associated with a comorbid psychiatric condition; this, together with a striking lack of effective pharmacological therapies, means they represent a considerable therapeutic challenge to the treating physician. In this overview, we examine the evidence to support the use of agents currently used in the management of the more common functional gastrointestinal disorders and review emerging therapies.
Collapse
Affiliation(s)
- Orla F Craig
- Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland and Department of Medicine, Clinical Sciences Building, Cork University Hospital, Cork, Ireland
| | | |
Collapse
|
13
|
Hultin L, Nissen TD, Kakol-Palm D, Lindström E. Colorectal distension-evoked potentials in awake rats: a novel method for studies of visceral sensitivity. Neurogastroenterol Motil 2012; 24:964-e466. [PMID: 22925114 DOI: 10.1111/nmo.12005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
BACKGROUND Quantification of the visceromotor response induced by colorectal distension (CRD) in rodents is commonly used for preclinical studies of visceral pain. The model is well established but does not fully assess the central response to stimulation. The aim of this study was to establish a novel model assessing cerebral evoked potentials (CEPs) in response to CRD in awake rats. METHODS Epidural recording electrodes were chronically implanted in the skull of female Sprague-Dawley rats. Colorectal distension-induced CEPs were recorded using either rapid balloon distensions (100 ms, 20-80 mmHg) or electric stimulation (1 ms, 1-4 mA) using stimulation probes placed in the distal colon. KEY RESULTS Colorectal distension-induced CEPs were separated in three partly temporally overlapping components consisting of five prominent peaks. Peak latencies at 80 mmHg were (P1, N1) 23 ± 1 and 55 ± 4 ms, (N2, P2a, P2b) 91 ± 3, 143 ± 5 and 174 ± 3 ms, and (P3) 297 ± 3 ms. Amplitudes and latencies were, except for the early component, intensity dependent. Intrarectal administration of lidocaine significantly reduced the amplitude of N2 (by 42 ± 6%, P < 0.001) and P2 (by 34 ± 6%, P < 0.001). Electrically induced CEPs were intensity dependent and had similar topography and latencies as the mechanical evoked potentials (P1: 26 ± 2 ms; N1: 61 ± 1 ms; P2: 84 ± 6 ms; N2: 154 ± 6 ms; P3: 326 ± 10 ms), but there were large variations in amplitudes in between repeated electrical stimulations. CONCLUSIONS & INFERENCES Colorectal distension-induced CEPs can be recorded reliably in awake rats and may serve as a surrogate marker of colonic sensation and be a useful parameter in studies of visceral sensitivity.
Collapse
Affiliation(s)
- L Hultin
- Integrative Pharmacology, AstraZeneca R&D, Mölndal, Sweden.
| | | | | | | |
Collapse
|
14
|
Lehmann A, Antonsson M, Aurell-Holmberg A, Blackshaw LA, Brändén L, Elebring T, Jensen J, Kärrberg L, Mattsson JP, Nilsson K, Oja SS, Saransaari P, von Unge S. Different in vitro and in vivo profiles of substituted 3-aminopropylphosphinate and 3-aminopropyl(methyl)phosphinate GABA(B) receptor agonists as inhibitors of transient lower oesophageal sphincter relaxation. Br J Pharmacol 2012; 165:1757-1772. [PMID: 21950457 DOI: 10.1111/j.1476-5381.2011.01682.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND AND PURPOSE Gastro-oesophageal reflux is predominantly caused by transient lower oesophageal sphincter relaxation (TLOSR) and GABA(B) receptor stimulation inhibits TLOSR. Lesogaberan produces fewer CNS side effects than baclofen, which has been attributed to its affinity for the GABA transporter (GAT), the action of which limits stimulation of central GABA(B) receptors. To understand the structure-activity relationship for analogues of lesogaberan (3-aminopropylphosphinic acids), and corresponding 3-aminopropyl(methyl)phosphinic acids, we have compared representatives of these classes in different in vitro and in vivo models. EXPERIMENTAL APPROACH The compounds were characterized in terms of GABA(B) agonism in vitro. Binding to GATs and cellular uptake was done using rat brain membranes and slices respectively. TLOSR was measured in dogs, and CNS side effects were evaluated as hypothermia in mice and rats. KEY RESULTS 3-Aminopropylphosphinic acids inhibited TLOSR with a superior therapeutic index compared to 3-aminopropyl(methyl)phosphinic acids. This difference was most likely due to differential GAT-mediated uptake into brain cells of the former but not latter. In agreement, 3-aminopropyl(methyl)phosphinic acids were much more potent in producing hypothermia in rats even when administered i.c.v. CONCLUSIONS AND IMPLICATIONS An enhanced therapeutic window for 3-aminopropylphosphinic acids compared with 3-aminopropyl(methyl)phosphinic acids with respect to inhibition of TLOSR was observed and is probably mechanistically linked to neural cell uptake of the former but not latter group of compounds. These findings offer a platform for discovery of new GABA(B) receptor agonists for the treatment of reflux disease and other conditions where selective peripheral GABA(B) receptor agonism may afford therapeutic effects.
Collapse
Affiliation(s)
- A Lehmann
- AstraZeneca R&D, Mölndal, SwedenNerve Gut Research Laboratory, Hanson Institute, Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, and University of Adelaide, South Australia, AustraliaDepartment of Paediatrics, Tampere University Hospital, FinlandBrain Research Center, Medical School, University of Tampere, Finland
| | - M Antonsson
- AstraZeneca R&D, Mölndal, SwedenNerve Gut Research Laboratory, Hanson Institute, Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, and University of Adelaide, South Australia, AustraliaDepartment of Paediatrics, Tampere University Hospital, FinlandBrain Research Center, Medical School, University of Tampere, Finland
| | - A Aurell-Holmberg
- AstraZeneca R&D, Mölndal, SwedenNerve Gut Research Laboratory, Hanson Institute, Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, and University of Adelaide, South Australia, AustraliaDepartment of Paediatrics, Tampere University Hospital, FinlandBrain Research Center, Medical School, University of Tampere, Finland
| | - L A Blackshaw
- AstraZeneca R&D, Mölndal, SwedenNerve Gut Research Laboratory, Hanson Institute, Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, and University of Adelaide, South Australia, AustraliaDepartment of Paediatrics, Tampere University Hospital, FinlandBrain Research Center, Medical School, University of Tampere, Finland
| | - L Brändén
- AstraZeneca R&D, Mölndal, SwedenNerve Gut Research Laboratory, Hanson Institute, Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, and University of Adelaide, South Australia, AustraliaDepartment of Paediatrics, Tampere University Hospital, FinlandBrain Research Center, Medical School, University of Tampere, Finland
| | - T Elebring
- AstraZeneca R&D, Mölndal, SwedenNerve Gut Research Laboratory, Hanson Institute, Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, and University of Adelaide, South Australia, AustraliaDepartment of Paediatrics, Tampere University Hospital, FinlandBrain Research Center, Medical School, University of Tampere, Finland
| | - J Jensen
- AstraZeneca R&D, Mölndal, SwedenNerve Gut Research Laboratory, Hanson Institute, Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, and University of Adelaide, South Australia, AustraliaDepartment of Paediatrics, Tampere University Hospital, FinlandBrain Research Center, Medical School, University of Tampere, Finland
| | - L Kärrberg
- AstraZeneca R&D, Mölndal, SwedenNerve Gut Research Laboratory, Hanson Institute, Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, and University of Adelaide, South Australia, AustraliaDepartment of Paediatrics, Tampere University Hospital, FinlandBrain Research Center, Medical School, University of Tampere, Finland
| | - J P Mattsson
- AstraZeneca R&D, Mölndal, SwedenNerve Gut Research Laboratory, Hanson Institute, Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, and University of Adelaide, South Australia, AustraliaDepartment of Paediatrics, Tampere University Hospital, FinlandBrain Research Center, Medical School, University of Tampere, Finland
| | - K Nilsson
- AstraZeneca R&D, Mölndal, SwedenNerve Gut Research Laboratory, Hanson Institute, Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, and University of Adelaide, South Australia, AustraliaDepartment of Paediatrics, Tampere University Hospital, FinlandBrain Research Center, Medical School, University of Tampere, Finland
| | - S S Oja
- AstraZeneca R&D, Mölndal, SwedenNerve Gut Research Laboratory, Hanson Institute, Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, and University of Adelaide, South Australia, AustraliaDepartment of Paediatrics, Tampere University Hospital, FinlandBrain Research Center, Medical School, University of Tampere, Finland
| | - P Saransaari
- AstraZeneca R&D, Mölndal, SwedenNerve Gut Research Laboratory, Hanson Institute, Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, and University of Adelaide, South Australia, AustraliaDepartment of Paediatrics, Tampere University Hospital, FinlandBrain Research Center, Medical School, University of Tampere, Finland
| | - S von Unge
- AstraZeneca R&D, Mölndal, SwedenNerve Gut Research Laboratory, Hanson Institute, Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, and University of Adelaide, South Australia, AustraliaDepartment of Paediatrics, Tampere University Hospital, FinlandBrain Research Center, Medical School, University of Tampere, Finland
| |
Collapse
|
15
|
Chapman MJ, Nguyen NQ, Deane AM. Gastrointestinal dysmotility: clinical consequences and management of the critically ill patient. Gastroenterol Clin North Am 2011; 40:725-39. [PMID: 22100114 DOI: 10.1016/j.gtc.2011.09.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
Abstract
Gastrointestinal dysmotility is a common feature of critical illness, with a number of significant implications that include malnutrition secondary to reduced feed tolerance and absorption, reflux and aspiration resulting in reduced lung function and ventilator-associated pneumonia, bacterial overgrowth and possible translocation causing nosocomial sepsis. Prokinetic agent administration can improve gastric emptying and caloric delivery, but its effect on nutrient absorption and clinical outcomes is, as yet, unclear. Postpyloric delivery of nutrition has not yet been demonstrated to increase caloric intake or improve clinical outcomes.
Collapse
Affiliation(s)
- Marianne J Chapman
- Department of Critical Care Services, Royal Adelaide Hospital, North Terrace, Adelaide, South Australia.
| | | | | |
Collapse
|
16
|
Niazi M, Skrtic S, Ruth M, Holmberg AA. Pharmacokinetic profile of lesogaberan (AZD3355) in healthy subjects: a novel GABA(B)-receptor agonist reflux inhibitor. Drugs R D 2011; 11:77-83. [PMID: 21410297 PMCID: PMC3585951 DOI: 10.2165/11590310-000000000-00000] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Objective: The aim of this study was to evaluate the pharmacokinetic profile of lesogaberan in healthy subjects after single oral and intravenous administration of 14C-labeled lesogaberan and non-14C-labeled lesogaberan. Study Design: This was an open-label, single-center, randomized, two-way crossover, phase I study. Participants: Ten healthy male subjects took part in the study. Intervention: Volunteers were randomized to receive a single dose of either orally dosed (100 mg) or intravenously infused (20 mg) non-14C-labeled lesogaberan, and then orally (100 mg) or intravenously (20 mg) administered 14C-labeled lesogaberan in a crossover design. Treatment periods were separated by a washout period of at least 7 days. Main Outcome Measures Analyses of the rate and route of excretion, dose recovery, area under the plasma concentration versus time curve (AUC), AUC to the last quantifiable concentration, maximal plasma concentration (Cmax), time to Cmax, the apparent elimination half-life, bioavailability, total clearance, renal clearance, fraction of the bioavailable dose excreted unchanged in the urine, cumulative amount of drug excreted unchanged in urine, and the apparent volume of distribution at steady state of lesogaberan. Results: Lesogaberan was rapidly and extensively absorbed from the gastrointestinal tract and Cmax was achieved within 1–2 hours of oral dosing. The terminal half-life of lesogaberan was between 11 and 13 hours. Renal clearance accounted for approximately 22% of total body clearance. Based on the recovery of administered radioactivity, approximately 84% of the dose was excreted into the urine either as the parent compound or as water-soluble metabolite(s). There were no safety concerns raised during the study. Conclusion: Orally administered lesogaberan is rapidly absorbed with high bioavailability and the majority of the dose is excreted by the kidneys either as the parent compound or as metabolites. The major elimination pathway for lesogaberan in man is metabolism.
Collapse
|
17
|
Abstract
In 1950, γ-aminobutyric acid (GABA) was discovered in the brain and in 1967 it was recognized as an inhibitory neurotransmitter. The discovery of the benzodiazepines Librium® (launched in 1960) and Valium® by Sternbach initiated huge research activities resulting in 50 marketed drugs. In 1975, Haefely found that GABA is involved in the actions of benzodiazepines. The baclofen-sensitive, bicuculline-insensitive GABAB receptor was discovered by Bowery in 1980, and the baclofen-insensitive, bicuculline-insensitive GABAC receptor by Johnston in 1984. Barnard & Seeburg reported the cloning of the GABAA receptor in 1987, Cutting the GABAC receptor in 1991 and Bettler the GABAB1a and GABAB1b receptors in 1997. Six groups cloned the GABAB2 receptor in 1998/1999 showing that the GABAB receptor functions as a heterodimer with GABAB1b/GABAB2 mediating postsynaptic inhibition and GABAB1a/GABAB2 mediating presynaptic inhibition. Möhler and McKernan dissected the pharmacology of the benzodiazepine-receptor subtypes. Antagonists and positive allosteric modulators of GABAB receptors were discovered in 1987 and 2001, respectively. GABA transporter inhibitor, tiagabine, was launched in 1996, a GABA aminotransferase inhibitor, vigabatrin, in 1998 and a glutamic acid decarboxylase activator, pregabalin, in 2004. Most recently, brain-penetrating GABAC-receptor antagonists were reported in 2009.
Collapse
|
18
|
Hyland NP, Cryan JF. A Gut Feeling about GABA: Focus on GABA(B) Receptors. Front Pharmacol 2010; 1:124. [PMID: 21833169 PMCID: PMC3153004 DOI: 10.3389/fphar.2010.00124] [Citation(s) in RCA: 118] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2010] [Accepted: 09/07/2010] [Indexed: 12/15/2022] Open
Abstract
γ-Aminobutyric acid (GABA) is the main inhibitory neurotransmitter in the body and hence GABA-mediated neurotransmission regulates many physiological functions, including those in the gastrointestinal (GI) tract. GABA is located throughout the GI tract and is found in enteric nerves as well as in endocrine-like cells, implicating GABA as both a neurotransmitter and an endocrine mediator influencing GI function. GABA mediates its effects via GABA receptors which are either ionotropic GABA(A) or metabotropic GABA(B). The latter which respond to the agonist baclofen have been least characterized, however accumulating data suggest that they play a key role in GI function in health and disease. Like GABA, GABA(B) receptors have been detected throughout the gut of several species in the enteric nervous system, muscle, epithelial layers as well as on endocrine-like cells. Such widespread distribution of this metabotropic GABA receptor is consistent with its significant modulatory role over intestinal motility, gastric emptying, gastric acid secretion, transient lower esophageal sphincter relaxation and visceral sensation of painful colonic stimuli. More intriguing findings, the mechanisms underlying which have yet to be determined, suggest GABA(B) receptors inhibit GI carcinogenesis and tumor growth. Therefore, the diversity of GI functions regulated by GABA(B) receptors makes it a potentially useful target in the treatment of several GI disorders. In light of the development of novel compounds such as peripherally acting GABA(B) receptor agonists, positive allosteric modulators of the GABA(B) receptor and GABA producing enteric bacteria, we review and summarize current knowledge on the function of GABA(B) receptors within the GI tract.
Collapse
Affiliation(s)
- Niall P Hyland
- Alimentary Pharmabiotic Centre and Department of Pharmacology and Therapeutics, University College Cork Cork, Ireland
| | | |
Collapse
|