1
|
Boehm T, Jilma B. Are leukotrienes really the world's best bronchoconstrictors and at least 100 to 1000 times more potent than histamine? Drug Discov Today 2025; 30:104349. [PMID: 40180311 DOI: 10.1016/j.drudis.2025.104349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 03/20/2025] [Accepted: 03/27/2025] [Indexed: 04/05/2025]
Abstract
It has been stated numerous times that leukotrienes are 100 to 1000 times more potent compared with histamine, but is this statement correct? Can we really compare a charged mono-cation with lipoid amphiphiles in their ability to penetrate an epithelial cell layer after inhalation challenge? In this review we question the shift in clinical and drug development attention from histamine towards leukotriene receptor antagonists for the treatment of chronic asthma and acute asthma exacerbations. The presented data indicate that histamine very likely plays a much more important role than previously assumed. It is time to rethink mediator involvement during bronchospasm and shift attention back to histamine.
Collapse
Affiliation(s)
- Thomas Boehm
- Department of Clinical Pharmacology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria.
| | - Bernd Jilma
- Department of Clinical Pharmacology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| |
Collapse
|
2
|
Wallin J, Forsberg A, Svenningsson P. Effects of Montelukast on Neuroinflammation in Parkinson's Disease: An Open Label Safety and Tolerability Trial with CSF Markers and [ 11C]PBR28 PET. Mov Disord 2025; 40:739-744. [PMID: 39912596 PMCID: PMC12006882 DOI: 10.1002/mds.30144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 01/18/2025] [Accepted: 01/23/2025] [Indexed: 02/07/2025] Open
Abstract
BACKGROUND Dysregulated leukotriene signaling is proposed to be involved in pathogenesis of Parkinson's disease (PD). OBJECTIVE The objective was to examine the safety and tolerability of montelukast, a cysteinyl-leukotriene receptor1 and GPR17 antagonist, in patients with PD. Secondary outcomes were target engagement, effects on PD signs/symptoms, and central neuroinflammation. METHODS Fifteen PD patients were recruited to a 12-week open-label trial of 20 mg bi-daily montelukast treatment. Patients underwent ratings with the Movement Disorder Society Unified Parkinson Disease Rating Scale (MDS-UPDRS), the Montreal Cognitive Assessment (MoCA), Beck's Depression Inventory (BDI), Parkinson's Disease Questionnaire-39 (PDQ-39), [11C]PBR28-PET, and lumbar punctures before and during montelukast treatment. RESULTS All patients completed the study. Three patients reported loose stool. No serious adverse events related to treatment were reported. MDS-UPDRS-Total scores improved by 6.9 points. Very low levels of montelukast were detected in all cerebrospinal fluid (CSF) samples and resulted in a reduction in inflammation/metabolism markers. [11C]PBR28 binding was lowered in high, but not mixed, affinity binders after montelukast. CONCLUSIONS Montelukast crosses the blood-brain barrier at very low levels and is well tolerated and safe in PD patients. Preliminary effects on neuroinflammation and clinical scores motivate a future randomized controlled trial (RCT) in PD. © 2025 The Author(s). Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Johan Wallin
- Center for Neurology, Region StockholmStockholmSweden
- Department of Clinical NeuroscienceKarolinska InstitutetStockholmSweden
| | - Anton Forsberg
- Centre for Psychiatry Research, Karolinska InstitutetStockholmSweden
| | - Per Svenningsson
- Center for Neurology, Region StockholmStockholmSweden
- Department of Clinical NeuroscienceKarolinska InstitutetStockholmSweden
| |
Collapse
|
3
|
Raffaele S, Nguyen N, Milanese M, Mannella FC, Boccazzi M, Frumento G, Bonanno G, Abbracchio MP, Bonifacino T, Fumagalli M. Montelukast improves disease outcome in SOD1 G93A female mice by counteracting oligodendrocyte dysfunction and aberrant glial reactivity. Br J Pharmacol 2024; 181:3303-3326. [PMID: 38751168 DOI: 10.1111/bph.16408] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 02/19/2024] [Accepted: 03/08/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND AND PURPOSE Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder characterized by progressive motor neuron (MN) loss and consequent muscle atrophy, for which no effective therapies are available. Recent findings reveal that disease progression is fuelled by early aberrant neuroinflammation and the loss of oligodendrocytes with neuroprotective and remyelinating properties. On this basis, pharmacological interventions capable of restoring a pro-regenerative local milieu and re-establish proper oligodendrocyte functions may be beneficial. EXPERIMENTAL APPROACH Here, we evaluated the in vivo therapeutic effects of montelukast (MTK), an antagonist of the oligodendroglial G protein-coupled receptor 17 (GPR17) and of cysteinyl-leukotriene receptor 1 (CysLT1R) receptors on microglia and astrocytes, in the SOD1G93A ALS mouse model. We chronically treated SOD1G93A mice with MTK, starting from the early symptomatic disease stage. Disease progression was assessed by behavioural and immunohistochemical approaches. KEY RESULTS Oral MTK treatment significantly extended survival probability, delayed body weight loss and ameliorated motor functionalityonly in female SOD1G93A mice. Noteworthy, MTK significantly restored oligodendrocyte maturation and induced significant changes in the reactive phenotype and morphological features of microglia/macrophages and astrocytes in the spinal cord of female SOD1G93A mice, suggesting enhanced pro-regenerative functions. Importantly, concomitant MN preservation has been detected after MTK administration. No beneficial effects were observed in male mice, highlighting a sex-based difference in the protective activity of MTK. CONCLUSIONS AND IMPLICATIONS Our results provide the first preclinical evidence indicating that repurposing of MTK, a safe and marketed anti-asthmatic drug, may be a promising sex-specific strategy for personalized ALS treatment.
Collapse
Affiliation(s)
- Stefano Raffaele
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
| | - Nhung Nguyen
- Department of Pharmacy, Unit of Pharmacology and Toxicology, Università degli Studi di Genova, Genoa, Italy
| | - Marco Milanese
- Department of Pharmacy, Unit of Pharmacology and Toxicology, Università degli Studi di Genova, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Francesca C Mannella
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
| | - Marta Boccazzi
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
| | - Giulia Frumento
- Department of Pharmacy, Unit of Pharmacology and Toxicology, Università degli Studi di Genova, Genoa, Italy
| | - Giambattista Bonanno
- Department of Pharmacy, Unit of Pharmacology and Toxicology, Università degli Studi di Genova, Genoa, Italy
| | - Maria P Abbracchio
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmaceutical Sciences, Università degli Studi di Milano, Milan, Italy
| | - Tiziana Bonifacino
- Department of Pharmacy, Unit of Pharmacology and Toxicology, Università degli Studi di Genova, Genoa, Italy
- Inter-University Center for the Promotion of the 3R Principles in Teaching and Research (Centro 3R), Pisa, Italy
| | - Marta Fumagalli
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
4
|
Mulgaonkar N, Wang H, Zhang J, Roundy CM, Tang W, Chaki SP, Pauvolid-Corrêa A, Hamer GL, Fernando S. Montelukast and Telmisartan as Inhibitors of SARS-CoV-2 Omicron Variant. Pharmaceutics 2023; 15:1891. [PMID: 37514075 PMCID: PMC10385313 DOI: 10.3390/pharmaceutics15071891] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 06/29/2023] [Accepted: 06/30/2023] [Indexed: 07/30/2023] Open
Abstract
Earlier studies with montelukast (M) and telmisartan (T) have revealed their potential antiviral properties against SARS-CoV-2 wild-type (WT) but have not assessed their efficacy against emerging Variants of Concern (VOCs) such as Omicron. Our research fills this gap by investigating these drugs' impact on VOCs, a topic that current scientific literature has largely overlooked. We employed computational methodologies, including molecular mechanics and machine learning tools, to identify drugs that could potentially disrupt the SARS-CoV-2 spike RBD-ACE2 protein interaction. This led to the identification of two FDA-approved small molecule drugs, M and T, conventionally used for treating asthma and hypertension, respectively. Our study presents an additional potential use for these drugs as antivirals. Our results show that both M and T can inhibit not only the WT SARS-CoV-2 but also, in the case of M, the Omicron variant, without reaching cytotoxic concentrations. This novel finding fills an existing gap in the literature and introduces the possibility of repurposing these drugs for SARS-CoV-2 VOCs, an essential step in responding to the evolving global pandemic.
Collapse
Affiliation(s)
- Nirmitee Mulgaonkar
- Biological and Agricultural Engineering Department, Texas A&M University, College Station, TX 77843, USA
| | - Haoqi Wang
- Biological and Agricultural Engineering Department, Texas A&M University, College Station, TX 77843, USA
| | - Junrui Zhang
- Biological and Agricultural Engineering Department, Texas A&M University, College Station, TX 77843, USA
| | | | - Wendy Tang
- Department of Entomology, Texas A&M University, College Station, TX 77843, USA
| | - Sankar Prasad Chaki
- Texas A&M Global Health Research Complex, Division of Research, Texas A&M University, College Station, TX 77843, USA
| | - Alex Pauvolid-Corrêa
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843, USA
| | - Gabriel L Hamer
- Department of Entomology, Texas A&M University, College Station, TX 77843, USA
| | - Sandun Fernando
- Biological and Agricultural Engineering Department, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
5
|
Tassan Mazzocco M, Murtaj V, Martins D, Schellino R, Coliva A, Toninelli E, Vercelli A, Turkheimer F, Belloli S, Moresco RM. Exploring the neuroprotective effects of montelukast on brain inflammation and metabolism in a rat model of quinolinic acid-induced striatal neurotoxicity. J Neuroinflammation 2023; 20:34. [PMID: 36782185 PMCID: PMC9923670 DOI: 10.1186/s12974-023-02714-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 01/31/2023] [Indexed: 02/15/2023] Open
Abstract
BACKGROUND One intrastriatal administration of quinolinic acid (QA) in rats induces a lesion with features resembling those observed in Huntington's disease. Our aim is to evaluate the effects of the cysteinyl leukotriene receptor antagonist montelukast (MLK), which exhibited neuroprotection in different preclinical models of neurodegeneration, on QA-induced neuroinflammation and regional metabolic functions. METHODS The right and left striatum of Sprague Dawley and athymic nude rats were injected with QA and vehicle (VEH), respectively. Starting from the day before QA injection, animals were treated with 1 or 10 mg/kg of MLK or VEH for 14 days. At 14 and 30 days post-lesion, animals were monitored with magnetic resonance imaging (MRI) and positron emission tomography (PET) using [18F]-VC701, a translocator protein (TSPO)-specific radiotracer. Striatal neuroinflammatory response was measured post-mortem in rats treated with 1 mg/kg of MLK by immunofluorescence. Rats treated with 10 mg/kg of MLK also underwent a [18F]-FDG PET study at baseline and 4 months after lesion. [18F]-FDG PET data were then used to assess metabolic connectivity between brain regions by applying a covariance analysis method. RESULTS MLK treatment was not able to reduce the QA-induced increase in striatal TSPO PET signal and MRI lesion volume, where we only detected a trend towards reduction in animals treated with 10 mg/kg of MLK. Post-mortem immunofluorescence analysis revealed that MLK attenuated the increase in striatal markers of astrogliosis and activated microglia in the lesioned hemisphere. We also found a significant increase in a marker of anti-inflammatory activity (MannR) and a trend towards reduction in a marker of pro-inflammatory activity (iNOS) in the lesioned striatum of MLK-compared to VEH-treated rats. [18F]-FDG uptake was significantly reduced in the striatum and ipsilesional cortical regions of VEH-treated rats at 4 months after lesion. MLK administration preserved glucose metabolism in these cortical regions, but not in the striatum. Finally, MLK was able to counteract changes in metabolic connectivity and measures of network topology induced by QA, in both lesioned and non-lesioned hemispheres. CONCLUSIONS Overall, MLK treatment produced a significant neuroprotective effect by reducing neuroinflammation assessed by immunofluorescence and preserving regional brain metabolism and metabolic connectivity from QA-induced neurotoxicity in cortical and subcortical regions.
Collapse
Affiliation(s)
- Margherita Tassan Mazzocco
- PhD Program in Neuroscience, Medicine and Surgery Department, University of Milano-Bicocca, Milan, Italy
- Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), Milan, Italy
| | - Valentina Murtaj
- PhD Program in Neuroscience, Medicine and Surgery Department, University of Milano-Bicocca, Milan, Italy
- Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), Milan, Italy
| | - Daniel Martins
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Roberta Schellino
- Department of Neuroscience "Rita Levi Montalcini" and Neuroscience Institute Cavalieri Ottolenghi, University of Turin, Turin, Italy
| | - Angela Coliva
- Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), Milan, Italy
| | - Elisa Toninelli
- Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), Milan, Italy
| | - Alessandro Vercelli
- Department of Neuroscience "Rita Levi Montalcini" and Neuroscience Institute Cavalieri Ottolenghi, University of Turin, Turin, Italy
| | - Federico Turkheimer
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Sara Belloli
- Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), Milan, Italy
- Institute of Molecular Bioimaging and Physiology (IBFM), CNR, Milan, Italy
| | - Rosa Maria Moresco
- Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), Milan, Italy.
- Institute of Molecular Bioimaging and Physiology (IBFM), CNR, Milan, Italy.
- Technomed Foundation and Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy.
| |
Collapse
|
6
|
Marques CF, Marques MM, Justino GC. Leukotrienes vs. Montelukast—Activity, Metabolism, and Toxicity Hints for Repurposing. Pharmaceuticals (Basel) 2022; 15:ph15091039. [PMID: 36145259 PMCID: PMC9505853 DOI: 10.3390/ph15091039] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/17/2022] [Accepted: 08/19/2022] [Indexed: 11/16/2022] Open
Abstract
Increasing environmental distress is associated with a growing asthma incidence; no treatments are available but montelukast (MTK)—an antagonist of the cysteinyl leukotrienes receptor 1—is widely used in the management of symptoms among adults and children. Recently, new molecular targets have been identified and MTK has been proposed for repurposing in other therapeutic applications, with several ongoing clinical trials. The proposed applications include neuroinflammation control, which could be explored in some neurodegenerative disorders, such as Alzheimer’s and Parkinson’s diseases (AD and PD). However, this drug has been associated with an increasing number of reported neuropsychiatric adverse drug reactions (ADRs). Besides, and despite being on the market since 1998, MTK metabolism is still poorly understood and the mechanisms underlying neuropsychiatric ADRs remain unknown. We review the role of MTK as a modulator of leukotriene pathways and systematize the current knowledge about MTK metabolism. Known toxic effects of MTK are discussed, and repurposing applications are presented comprehensively, with a focus on AD and PD.
Collapse
Affiliation(s)
- Cátia F. Marques
- Centro de Química Estrutural, Institute of Molecular Sciences, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal
| | - Maria Matilde Marques
- Centro de Química Estrutural, Institute of Molecular Sciences, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal
- Departamento de Engenharia Química, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal
| | - Gonçalo C. Justino
- Centro de Química Estrutural, Institute of Molecular Sciences, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal
- Correspondence:
| |
Collapse
|
7
|
Gallegos EM, Reed TD, Mathes FA, Guevara NV, Neau DB, Huang W, Newcomer ME, Gilbert NC. Helical remodeling augments 5-lipoxygenase activity in the synthesis of pro-inflammatory mediators. J Biol Chem 2022; 298:102282. [PMID: 35863431 PMCID: PMC9418500 DOI: 10.1016/j.jbc.2022.102282] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 07/13/2022] [Accepted: 07/15/2022] [Indexed: 11/29/2022] Open
Abstract
The synthesis of pro-inflammatory leukotrienes implicated in asthma, allergic rhinitis, and atherosclerosis is initiated by the enzyme 5-lipoxygenase (5-LOX). The crystal structure of human Stable-5-LOX revealed a conformation where the catalytic iron was inaccessible to bulk solvent as two aromatic residues on a conserved helix-α2 (Hα2) plugged the substrate access portal. Whether 5-LOX can also adopt a more open conformation has not been resolved. Here, we present a new conformation of 5-LOX where Hα2 adopts an elongated conformation equivalent to that described in other animal lipoxygenase structures. Our observation of the sigmoidal kinetic behavior of 5-LOX, which is indicative of positive cooperativity, is consistent with a substrate-induced conformational change that shifts the ensemble of enzyme populations to favor the catalytically competent state. Strategic point mutations along Hα2 designed to unlock the closed conformation and elongate Hα2 resulted in improved kinetic parameters, altered limited-proteolysis data, and a drastic reduction in the length of the lag phase yielding the most active Stable-5-LOX to date. Structural predictions by AlphaFold2 of these variants statistically favor an elongated Hα2 and reinforce a model in which improved kinetic parameters correlate with a more readily adopted, open conformation. Taken together, these data provide valuable insights into the synthesis of leukotrienes.
Collapse
Affiliation(s)
- Eden M Gallegos
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, USA
| | - Tanner D Reed
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, USA
| | - Forge A Mathes
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, USA
| | - Nelson V Guevara
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, USA
| | - David B Neau
- Cornell University, Northeastern Collaborative Access Team, Argonne National Laboratory, Argonne, IL, USA
| | - Wei Huang
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH, USA
| | - Marcia E Newcomer
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, USA
| | - Nathaniel C Gilbert
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, USA.
| |
Collapse
|
8
|
Sokolowska M, Rovati GE, Diamant Z, Untersmayr E, Schwarze J, Lukasik Z, Sava F, Angelina A, Palomares O, Akdis C, O'Mahony L, Jesenak M, Pfaar O, Torres MJ, Sanak M, Dahlén S, Woszczek G. Effects of non-steroidal anti-inflammatory drugs and other eicosanoid pathway modifiers on antiviral and allergic responses: EAACI task force on eicosanoids consensus report in times of COVID-19. Allergy 2022; 77:2337-2354. [PMID: 35174512 PMCID: PMC9111413 DOI: 10.1111/all.15258] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 01/25/2022] [Accepted: 02/11/2022] [Indexed: 11/29/2022]
Abstract
Non‐steroidal anti‐inflammatory drugs (NSAIDs) and other eicosanoid pathway modifiers are among the most ubiquitously used medications in the general population. Their broad anti‐inflammatory, antipyretic, and analgesic effects are applied against symptoms of respiratory infections, including SARS‐CoV‐2, as well as in other acute and chronic inflammatory diseases that often coexist with allergy and asthma. However, the current pandemic of COVID‐19 also revealed the gaps in our understanding of their mechanism of action, selectivity, and interactions not only during viral infections and inflammation, but also in asthma exacerbations, uncontrolled allergic inflammation, and NSAIDs‐exacerbated respiratory disease (NERD). In this context, the consensus report summarizes currently available knowledge, novel discoveries, and controversies regarding the use of NSAIDs in COVID‐19, and the role of NSAIDs in asthma and viral asthma exacerbations. We also describe here novel mechanisms of action of leukotriene receptor antagonists (LTRAs), outline how to predict responses to LTRA therapy and discuss a potential role of LTRA therapy in COVID‐19 treatment. Moreover, we discuss interactions of novel T2 biologicals and other eicosanoid pathway modifiers on the horizon, such as prostaglandin D2 antagonists and cannabinoids, with eicosanoid pathways, in context of viral infections and exacerbations of asthma and allergic diseases. Finally, we identify and summarize the major knowledge gaps and unmet needs in current eicosanoid research.
Collapse
Affiliation(s)
- Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
- Christine Kühne ‐ Center for Allergy Research and Education (CK‐CARE) Davos Switzerland
| | - G Enrico Rovati
- Department of Pharmaceutical Sciences Section of Pharmacology and Biosciences University of Milan Milano Italy
| | - Zuzana Diamant
- Department of Respiratory Medicine and Allergology Skane University Hospital Lund Sweden
- Department Microbiology Immunology and Transplantation Ku Leuven, Catholic University of Leuven Belgium
- Department of Respiratory Medicine First Faculty of Medicine Charles University and Thomayer Hospital Prague Czech Republic
| | - Eva Untersmayr
- Institute of Pathophysiology and Allergy Research Center for Pathophysiology, Infectiology and Immunology Medical University of Vienna Vienna Austria
| | - Jürgen Schwarze
- Child Life and Health and Centre for Inflammation Research The University of Edinburgh Edinburgh UK
| | - Zuzanna Lukasik
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
- VIB Center for Inflammation Research Ghent University Ghent Belgium
| | - Florentina Sava
- London North Genomic Laboratory Hub Great Ormond Street Hospital for Children NHS Foundation Trust London UK
| | - Alba Angelina
- Department of Biochemistry and Molecular Biology School of Chemistry Complutense University Madrid Spain
| | - Oscar Palomares
- Department of Biochemistry and Molecular Biology School of Chemistry Complutense University Madrid Spain
| | - Cezmi Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
- Christine Kühne ‐ Center for Allergy Research and Education (CK‐CARE) Davos Switzerland
| | - Liam O'Mahony
- Departments of Medicine and Microbiology APC Microbiome IrelandUniversity College Cork Cork Ireland
| | - Milos Jesenak
- Department of Pulmonology and Phthisiology Department of Allergology and Clinical Immunology Department of Pediatrics Jessenius Faculty of Medicine in Martin Comenius University in BratislavaUniversity Teaching Hospital in Martin Slovakia
| | - Oliver Pfaar
- Department of Otorhinolaryngology, Head and Neck Surgery Section of Rhinology and Allergy University Hospital MarburgPhilipps‐Universität Marburg Marburg Germany
| | - María José Torres
- Allergy Unit Málaga Regional University Hospital‐IBIMA‐UMA Málaga Spain
| | - Marek Sanak
- Department of Medicine Jagiellonian University Medical College Krakow Poland
| | - Sven‐Erik Dahlén
- Institute of Environmental Medicine and the Centre for Allergy Research, Karolinska Institute, and the Department of Respiratory Medicine Karolinska University Hospital Stockholm Sweden
| | - Grzegorz Woszczek
- Asthma UK Centre in Allergic Mechanisms of Asthma School of Immunology and Microbial Sciences King's College London London UK
| |
Collapse
|
9
|
Trained immunity in type 2 immune responses. Mucosal Immunol 2022; 15:1158-1169. [PMID: 36065058 PMCID: PMC9705254 DOI: 10.1038/s41385-022-00557-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 07/27/2022] [Accepted: 08/08/2022] [Indexed: 02/04/2023]
Abstract
Immunological memory of innate immune cells, also termed "trained immunity", allows for cross-protection against distinct pathogens, but may also drive chronic inflammation. Recent studies have shown that memory responses associated with type 2 immunity do not solely rely on adaptive immune cells, such as T- and B cells, but also involve the innate immune system and epithelial cells. Memory responses have been described for monocytes, macrophages and airway epithelial cells of asthmatic patients as well as for macrophages and group 2 innate lymphoid cells (ILC2) from allergen-sensitized or helminth-infected mice. The metabolic and epigenetic mechanisms that mediate allergen- or helminth-induced reprogramming of innate immune cells are only beginning to be uncovered. Trained immunity has been implicated in helminth-driven immune regulation and allergen-specific immunotherapy, suggesting its exploitation in future therapies. Here, we discuss recent advances and key remaining questions regarding the mechanisms and functions of trained type 2 immunity in infection and inflammation.
Collapse
|
10
|
Durdagi S, Avsar T, Orhan MD, Serhatli M, Balcioglu BK, Ozturk HU, Kayabolen A, Cetin Y, Aydinlik S, Bagci-Onder T, Tekin S, Demirci H, Guzel M, Akdemir A, Calis S, Oktay L, Tolu I, Butun YE, Erdemoglu E, Olkan A, Tokay N, Işık Ş, Ozcan A, Acar E, Buyukkilic S, Yumak Y. The neutralization effect of montelukaston SARS-CoV-2 is shown by multiscale in silicosimulations and combined in vitro studies. Mol Ther 2021; 30:963-974. [PMID: 34678509 PMCID: PMC8524809 DOI: 10.1016/j.ymthe.2021.10.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 08/31/2021] [Accepted: 10/15/2021] [Indexed: 12/22/2022] Open
Abstract
Small molecule inhibitors have previously been investigated in different studies as possible therapeutics in the treatment of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). In the current drug repurposing study, we identified the leukotriene (D4) receptor antagonist montelukast as a novel agent that simultaneously targets two important drug targets of SARS-CoV-2. We initially demonstrated the dual inhibition profile of montelukast through multiscale molecular modeling studies. Next, we characterized its effect on both targets by different in vitro experiments including the enzyme (main protease) inhibition-based assay, surface plasmon resonance (SPR) spectroscopy, pseudovirus neutralization on HEK293T/hACE2+TMPRSS2, and virus neutralization assay using xCELLigence MP real-time cell analyzer. Our integrated in silico and in vitro results confirmed the dual potential effect of montelukast both on the main protease enzyme inhibition and virus entry into the host cell (spike/ACE2). The virus neutralization assay results showed that SARS-CoV-2 virus activity was delayed with montelukast for 20 h on the infected cells. The rapid use of new small molecules in the pandemic is very important today. Montelukast, whose pharmacokinetic and pharmacodynamic properties are very well characterized and has been widely used in the treatment of asthma since 1998, should urgently be completed in clinical phase studies and, if its effect is proved in clinical phase studies, it should be used against coronavirus disease 2019 (COVID-19).
Collapse
Affiliation(s)
- Serdar Durdagi
- Department of Biophysics, Computational Biology and Molecular Simulations Laboratory, School of Medicine, Bahçeşehir University, Istanbul, Turkey.
| | - Timucin Avsar
- Department of Medical Biology, School of Medicine, Bahçeşehir University, Istanbul, Turkey
| | - Muge Didem Orhan
- Department of Medical Biology, School of Medicine, Bahçeşehir University, Istanbul, Turkey
| | - Muge Serhatli
- The Scientific and Technological Research Council of Turkey (TÜBİTAK) Marmara Research Center (MAM), Genetic Engineering and Biotechnology Institute, 41470 Gebze, Kocaeli
| | - Bertan Koray Balcioglu
- The Scientific and Technological Research Council of Turkey (TÜBİTAK) Marmara Research Center (MAM), Genetic Engineering and Biotechnology Institute, 41470 Gebze, Kocaeli
| | - Hasan Umit Ozturk
- The Scientific and Technological Research Council of Turkey (TÜBİTAK) Marmara Research Center (MAM), Genetic Engineering and Biotechnology Institute, 41470 Gebze, Kocaeli
| | - Alisan Kayabolen
- Brain Cancer Research and Therapy Laboratory, Koç University School of Medicine, 34450 Istanbul, Turkey
| | - Yuksel Cetin
- The Scientific and Technological Research Council of Turkey (TÜBİTAK) Marmara Research Center (MAM), Genetic Engineering and Biotechnology Institute, 41470 Gebze, Kocaeli
| | - Seyma Aydinlik
- The Scientific and Technological Research Council of Turkey (TÜBİTAK) Marmara Research Center (MAM), Genetic Engineering and Biotechnology Institute, 41470 Gebze, Kocaeli
| | - Tugba Bagci-Onder
- Brain Cancer Research and Therapy Laboratory, Koç University School of Medicine, 34450 Istanbul, Turkey; Koç University Research Center for Translational Medicine, 34450 Istanbul, Turkey
| | - Saban Tekin
- The Scientific and Technological Research Council of Turkey (TÜBİTAK) Marmara Research Center (MAM), Genetic Engineering and Biotechnology Institute, 41470 Gebze, Kocaeli; Department of Basic Sciences, Division of Medical Biology, Faculty of Medicine, University of Health Sciences, Istanbul, Turkey
| | - Hasan Demirci
- Department of Molecular Biology and Genetics, Koç University, 34450 Istanbul, Turkey
| | - Mustafa Guzel
- Department of Medical Pharmacology, International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Atilla Akdemir
- Department of Pharmacology, Computer-aided Drug Discovery Laboratory, Faculty of Pharmacy, Bezmialem Vakif University, Istanbul, Turkey
| | - Seyma Calis
- Department of Medical Biology, School of Medicine, Bahçeşehir University, Istanbul, Turkey; Department of Molecular Biology-Genetics and Biotechnology, Istanbul Technical University, 34485 Istanbul, Turkey
| | - Lalehan Oktay
- Department of Biophysics, Computational Biology and Molecular Simulations Laboratory, School of Medicine, Bahçeşehir University, Istanbul, Turkey
| | - Ilayda Tolu
- Department of Biophysics, Computational Biology and Molecular Simulations Laboratory, School of Medicine, Bahçeşehir University, Istanbul, Turkey
| | - Yasar Enes Butun
- Department of Medical Pharmacology, International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Ece Erdemoglu
- Department of Biophysics, Computational Biology and Molecular Simulations Laboratory, School of Medicine, Bahçeşehir University, Istanbul, Turkey; Faculty of Medicine, Mersin University, Mersin, Turkey
| | - Alpsu Olkan
- Department of Biophysics, Computational Biology and Molecular Simulations Laboratory, School of Medicine, Bahçeşehir University, Istanbul, Turkey
| | - Nurettin Tokay
- The Scientific and Technological Research Council of Turkey (TÜBİTAK) Marmara Research Center (MAM), Genetic Engineering and Biotechnology Institute, 41470 Gebze, Kocaeli
| | - Şeyma Işık
- The Scientific and Technological Research Council of Turkey (TÜBİTAK) Marmara Research Center (MAM), Genetic Engineering and Biotechnology Institute, 41470 Gebze, Kocaeli
| | - Aysenur Ozcan
- Department of Biophysics, Computational Biology and Molecular Simulations Laboratory, School of Medicine, Bahçeşehir University, Istanbul, Turkey; Faculty of Medicine, Istanbul Medeniyet University, Istanbul, Turkey
| | - Elif Acar
- Department of Biophysics, Computational Biology and Molecular Simulations Laboratory, School of Medicine, Bahçeşehir University, Istanbul, Turkey; Faculty of Medicine, Istanbul Medeniyet University, Istanbul, Turkey
| | - Sehriban Buyukkilic
- Department of Biophysics, Computational Biology and Molecular Simulations Laboratory, School of Medicine, Bahçeşehir University, Istanbul, Turkey; Faculty of Science, Necmettin Erbakan University, Konya, Turkey
| | - Yesim Yumak
- Department of Biophysics, Computational Biology and Molecular Simulations Laboratory, School of Medicine, Bahçeşehir University, Istanbul, Turkey; Faculty of Science and Letters, Tokat Gaziosmanpaşa University, Tokat, Turkey
| |
Collapse
|
11
|
Potential Effects of Leukotriene Receptor Antagonist Montelukast in Treatment of Neuroinflammation in Parkinson's Disease. Int J Mol Sci 2021; 22:ijms22115606. [PMID: 34070609 PMCID: PMC8198163 DOI: 10.3390/ijms22115606] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/16/2021] [Accepted: 05/21/2021] [Indexed: 12/11/2022] Open
Abstract
Parkinson’s disease (PD) is a neurodegenerative disorder where misfolded alpha-synuclein-enriched aggregates called Lewy bodies are central in pathogenesis. No neuroprotective or disease-modifying treatments are currently available. Parkinson’s disease is considered a multifactorial disease and evidence from multiple patient studies and animal models has shown a significant immune component during the course of the disease, highlighting immunomodulation as a potential treatment strategy. The immune changes occur centrally, involving microglia and astrocytes but also peripherally with changes to the innate and adaptive immune system. Here, we review current understanding of different components of the PD immune response with a particular emphasis on the leukotriene pathway. We will also describe evidence of montelukast, a leukotriene receptor antagonist, as a possible anti-inflammatory treatment for PD.
Collapse
|
12
|
Matsuda M, Tabuchi Y, Nishimura K, Nakamura Y, Sekioka T, Kadode M, Kawabata K, Nabe T. Increased expression of CysLT 2 receptors in the lung of asthmatic mice and role in allergic responses. Prostaglandins Leukot Essent Fatty Acids 2018; 131:24-31. [PMID: 29628047 DOI: 10.1016/j.plefa.2018.03.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 03/13/2018] [Accepted: 03/21/2018] [Indexed: 12/23/2022]
Abstract
Compared with CysLT1 receptors, the functional role of CysLT2 receptors in asthma has not been clarified. The purpose of this study was to determine 1) whether CysLT2 receptors are expressed in the lung of mice and if expression increases in asthmatic mice, and 2) whether CysLT2 receptors are involved in allergic leukocyte infiltration into the lung and in the development of airway remodeling in asthmatic mice. BALB/c mice were sensitized with ovalbumin (OVA) + Al(OH)3, and intratracheally challenged with OVA 4 times. Lung tissue was isolated before and after the 4th OVA challenge for detection of CysLT2 receptors by immunohistochemistry and flow cytometry. The effect of a CysLT2 receptor antagonist BayCysLT2RA on multiple antigen challenge-induced leukocyte infiltration into the lung and the development of airway remodeling was evaluated. Even in non-challenged mice, CysLT2 receptors were expressed in bronchial smooth muscle. After multiple challenges, expression was also observed in leukocytes infiltrating into alveolar spaces. CysLT2R+ leukocytes included alveolar macrophages, conventional dendritic cells, and eosinophils. BayCysLT2RA significantly inhibited multiple antigen challenge-induced increases in eosinophils and mononuclear cells in the lung. The development of airway remodeling was tended to be suppressed by CysLT2 receptor antagonist. In conclusion, CysLT2 receptors were constitutively expressed in the lung, and expression was strengthened in asthmatic mice. Activation of CysLT2 receptors was functionally involved in allergic leukocyte infiltration into the lung. The CysLT2 receptor can be a molecular target for the development of new pharmacotherapies for asthma.
Collapse
Affiliation(s)
- Masaya Matsuda
- Department of Immunopharmacology, Faculty of Pharmaceutical Sciences, Setsunan University, Osaka, Japan
| | - Yuki Tabuchi
- Department of Immunopharmacology, Faculty of Pharmaceutical Sciences, Setsunan University, Osaka, Japan
| | - Kazuma Nishimura
- Department of Immunopharmacology, Faculty of Pharmaceutical Sciences, Setsunan University, Osaka, Japan
| | - Yuri Nakamura
- Department of Immunopharmacology, Faculty of Pharmaceutical Sciences, Setsunan University, Osaka, Japan
| | - Tomohiko Sekioka
- Minase Research Institute, Ono Pharmaceutical Co., Ltd, Osaka, Japan
| | - Michiaki Kadode
- Minase Research Institute, Ono Pharmaceutical Co., Ltd, Osaka, Japan
| | - Kazuhito Kawabata
- Minase Research Institute, Ono Pharmaceutical Co., Ltd, Osaka, Japan
| | - Takeshi Nabe
- Department of Immunopharmacology, Faculty of Pharmaceutical Sciences, Setsunan University, Osaka, Japan.
| |
Collapse
|
13
|
Leukotriene E 4 induces airflow obstruction and mast cell activation through the cysteinyl leukotriene type 1 receptor. J Allergy Clin Immunol 2018. [PMID: 29518425 DOI: 10.1016/j.jaci.2018.02.024] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND Leukotriene (LT) E4 is the final active metabolite among the cysteinyl leukotrienes (CysLTs). Animal studies have identified a distinct LTE4 receptor, suggesting that current cysteinyl leukotriene type 1 (CysLT1) receptor antagonists can provide incomplete inhibition of CysLT responses. OBJECTIVE We tested this hypothesis by assessing the influence of the CysLT1 antagonist montelukast on responses induced by means of inhalation of LTE4 in asthmatic patients. METHODS Fourteen patients with mild intermittent asthma and 2 patients with aspirin-exacerbated respiratory disease received 20 mg of montelukast twice daily and placebo for 5 to 7 days in a randomized, double-blind, crossover study (NCT01841164). The PD20 value was determined at the end of each treatment period based on an increasing dose challenge. Measurements included lipid mediators in urine and sputum cells 4 hours after LTE4 challenge. RESULTS Montelukast completely blocked LTE4-induced bronchoconstriction. Despite tolerating an at least 10 times higher dose of LTE4 after montelukast, there was no difference in the percentage of eosinophils in sputum. Urinary excretion of all major lipid mediators increased after LTE4 inhalation. Montelukast blocked release of the mast cell product prostaglandin (PG) D2, as well as release of PGF2α and thromboxane (Tx) A2, but not increased excretion of PGE2 and its metabolites or isoprostanes. CONCLUSION LTE4 induces airflow obstruction and mast cell activation through the CysLT1 receptor.
Collapse
|
14
|
Korenblat P, Kerwin E, Leshchenko I, Yen K, Holweg CTJ, Anzures-Cabrera J, Martin C, Putnam WS, Governale L, Olsson J, Matthews JG. Efficacy and safety of lebrikizumab in adult patients with mild-to-moderate asthma not receiving inhaled corticosteroids. Respir Med 2017; 134:143-149. [PMID: 29413502 DOI: 10.1016/j.rmed.2017.12.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 10/27/2017] [Accepted: 12/10/2017] [Indexed: 10/18/2022]
Abstract
BACKGROUND Asthma is a heterogeneous and complex disease in both its clinical course and response to treatment. IL-13 is central to Type 2 inflammation and contributes to many features of asthma. In a previous Phase 2 study, lebrikizumab, an anti-IL-13 monoclonal antibody, did not significantly improve FEV1 in mild-to-moderate asthma patients not receiving ICS therapy. This Phase 3 study was designed to further assess the efficacy and safety of lebrikizumab in adult patients with mild-to-moderate asthma treated with daily short-acting β2-agonist therapy alone. METHODS Adult patients with mild-to-moderate asthma were randomised to receive lebrikizumab 125 mg subcutaneously (SC), placebo SC, or montelukast 10 mg orally for 12 weeks, with an 8-week follow-up period. The primary efficacy endpoint was absolute change in pre-bronchodilator FEV1 from baseline at Week 12. FINDINGS A total of 310 patients were randomised and dosed in the study. The mean absolute change in FEV1 from baseline at Week 12 was higher in the lebrikizumab-treated arm compared with placebo (150 mL versus 67 mL); however, this improvement did not achieve statistical significance (overall adjusted difference of 83 mL [95% CI: -3, 170]; p = .06). Montelukast did not improve FEV1 as compared with placebo. Lebrikizumab was generally safe and well tolerated during the study. INTERPRETATION Lebrikizumab did not significantly improve FEV1 in mild-to-moderate asthma patients at a dose expected to inhibit the IL-13 pathway. Inhibiting IL-13 in this patient population was not sufficient to improve lung function. These data support the findings of a previous trial of lebrikizumab in patients not receiving ICS. CLINICAL TRIALS REGISTRY NUMBER This trial was registered under NCT02104674 at http://www.clinicaltrials.gov.
Collapse
Affiliation(s)
| | - Edwin Kerwin
- Clinical Research Institute of Southern Oregon PC, Medford, OR, USA
| | | | - Karl Yen
- Genentech, Inc., South San Francisco, CA, USA
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Arnold DH, Bowman N, Reiss TF, Hartert TV, Seger DL. Adverse events are rare after single-dose montelukast exposures in children. Clin Toxicol (Phila) 2017. [PMID: 28639856 DOI: 10.1080/15563650.2017.1337123] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
STUDY OBJECTIVE Montelukast sodium is a leukotriene-receptor antagonist approved as a controller medication for chronic asthma and allergic rhinitis in children and adults. We sought to characterize adverse events associated with single montelukast exposures in children ages 5-17 years and to determine whether adverse events were dose related for all-dose and for ultra-high-dose (≥50 mg) exposures. METHODS This is a retrospective analysis of data from the National Poison Data System for exposures that included montelukast in individuals aged 5-17 years for calendar years 2000-2016. Filters were applied to identify exposure events in which montelukast was the primary exposure and for which the exact or lowest-possible ingested dose was recorded. Characteristics of adverse events were examined using descriptive statistics and multivariable logistic models were used to examine whether associations of montelukast and adverse events were dose related. RESULTS During the 17-year study period, there were 17,069 montelukast exposures available for analyses. Patients were median [interquartile range] age 7 (5, 9) years, and 10,907 (64%) male gender. Abdominal pain was the most common adverse event (0.23%). There were 618 ultra-high-dose exposures (≥50 mg). These patients had median age 6 (5, 8) years, and 347 (56%) male gender. Abdominal pain was the most common adverse event (1.46%). Increasing ingested dose was associated with abdominal pain (adjusted odds ratio, 1.01, 95% confidence interval 1.01, 1.02) after adjustment for age and gender. No serious or life-threatening events were reported. CONCLUSIONS Single-dose exposures of montelukast up to 445 mg are rarely associated with any adverse events and are not associated with serious or life-threatening adverse events in children aged 5-17 years.
Collapse
Affiliation(s)
- Donald H Arnold
- a Departments of Pediatrics and Emergency Medicine , Vanderbilt University School of Medicine , Nashville , TN , USA.,b Center for Asthma Research , Vanderbilt University School of Medicine , Nashville , TN , USA
| | - Nena Bowman
- c Tennessee Poison Center , Nashville , TN , USA.,d Department of Pharmacy , Vanderbilt University Medical Center , Nashville , TN , USA
| | | | - Tina V Hartert
- b Center for Asthma Research , Vanderbilt University School of Medicine , Nashville , TN , USA.,f Department of Medicine , Vanderbilt University School of Medicine , Nashville , TN , USA
| | - Donna L Seger
- c Tennessee Poison Center , Nashville , TN , USA.,g Departments of Medicine and Emergency Medicine , Vanderbilt University School of Medicine , Nashville , TN , USA
| |
Collapse
|
16
|
Proschak E, Heitel P, Kalinowsky L, Merk D. Opportunities and Challenges for Fatty Acid Mimetics in Drug Discovery. J Med Chem 2017; 60:5235-5266. [PMID: 28252961 DOI: 10.1021/acs.jmedchem.6b01287] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Fatty acids beyond their role as an endogenous energy source and storage are increasingly considered as signaling molecules regulating various physiological effects in metabolism and inflammation. Accordingly, the molecular targets involved in formation and physiological activities of fatty acids hold significant therapeutic potential. A number of these fatty acid targets are addressed by some of the oldest and most widely used drugs such as cyclooxygenase inhibiting NSAIDs, whereas others remain unexploited. Compounds orthosterically binding to proteins that endogenously bind fatty acids are considered as fatty acid mimetics. On the basis of their structural resemblance, fatty acid mimetics constitute a family of bioactive compounds showing specific binding thermodynamics and following similar pharmacokinetic mechanisms. This perspective systematically evaluates targets for fatty acid mimetics, investigates their common structural characteristics, and highlights demands in their discovery and design. In summary, fatty acid mimetics share particularly favorable characteristics justifying the conclusion that their therapeutic potential vastly outweighs the challenges in their design.
Collapse
Affiliation(s)
- Ewgenij Proschak
- Institute of Pharmaceutical Chemistry, Goethe-University Frankfurt , Max-von-Laue-Straße 9, 60438 Frankfurt, Germany
| | - Pascal Heitel
- Institute of Pharmaceutical Chemistry, Goethe-University Frankfurt , Max-von-Laue-Straße 9, 60438 Frankfurt, Germany
| | - Lena Kalinowsky
- Institute of Pharmaceutical Chemistry, Goethe-University Frankfurt , Max-von-Laue-Straße 9, 60438 Frankfurt, Germany
| | - Daniel Merk
- Institute of Pharmaceutical Chemistry, Goethe-University Frankfurt , Max-von-Laue-Straße 9, 60438 Frankfurt, Germany
| |
Collapse
|
17
|
Thompson MD, Capra V, Clunes MT, Rovati GE, Stankova J, Maj MC, Duffy DL. Cysteinyl Leukotrienes Pathway Genes, Atopic Asthma and Drug Response: From Population Isolates to Large Genome-Wide Association Studies. Front Pharmacol 2016; 7:299. [PMID: 27990118 PMCID: PMC5131607 DOI: 10.3389/fphar.2016.00299] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 08/24/2016] [Indexed: 02/05/2023] Open
Abstract
Genetic variants associated with asthma pathogenesis and altered response to drug therapy are discussed. Many studies implicate polymorphisms in genes encoding the enzymes responsible for leukotriene synthesis and intracellular signaling through activation of seven transmembrane domain receptors, such as the cysteinyl leukotriene 1 (CYSLTR1) and 2 (CYSLTR2) receptors. The leukotrienes are polyunsaturated lipoxygenated eicosatetraenoic acids that exhibit a wide range of pharmacological and physiological actions. Of the three enzymes involved in the formation of the leukotrienes, arachidonate 5 lipoxygenase 5 (ALOX5), leukotriene C4 synthase (LTC4S), and leukotriene hydrolase (LTA4H) are all polymorphic. These polymorphisms often result in variable production of the CysLTs (LTC4, LTD4, and LTE4) and LTB4. Variable number tandem repeat sequences located in the Sp1-binding motif within the promotor region of the ALOX5 gene are associated with leukotriene burden and bronchoconstriction independent of asthma risk. A 444A > C SNP polymorphism in the LTC4S gene, encoding an enzyme required for the formation of a glutathione adduct at the C-6 position of the arachidonic acid backbone, is associated with severe asthma and altered response to the CYSLTR1 receptor antagonist zafirlukast. Genetic variability in the CysLT pathway may contribute additively or synergistically to altered drug responses. The 601 A > G variant of the CYSLTR2 gene, encoding the Met201Val CYSLTR2 receptor variant, is associated with atopic asthma in the general European population, where it is present at a frequency of ∼2.6%. The variant was originally found in the founder population of Tristan da Cunha, a remote island in the South Atlantic, in which the prevalence of atopy is approximately 45% and the prevalence of asthma is 36%. In vitro work showed that the atopy-associated Met201Val variant was inactivating with respect to ligand binding, Ca2+ flux and inositol phosphate generation. In addition, the CYSLTR1 gene, located at Xq13-21.1, has been associated with atopic asthma. The activating Gly300Ser CYSLTR1 variant is discussed. In addition to genetic loci, risk for asthma may be influenced by environmental factors such as smoking. The contribution of CysLT pathway gene sequence variants to atopic asthma is discussed in the context of other genes and environmental influences known to influence asthma.
Collapse
Affiliation(s)
- Miles D Thompson
- Biochemical Genetics and Metabolomics Laboratory, Department of Pediatrics, University of California, San Diego, La JollaCA, USA; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ONCanada
| | - Valerie Capra
- Department of Health Sciences, San Paolo Hospital, Università degli Studi di Milano Milano, Italy
| | - Mark T Clunes
- Department of Physiology/Neuroscience, School of Medicine, Saint George's University Saint George's, Grenada
| | - G E Rovati
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano Milano, Italy
| | - Jana Stankova
- Division of Immunology and Allergy, Department of Pediatrics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke QC, Canada
| | - Mary C Maj
- Department of Biochemistry, School of Medicine, Saint George's University Saint George's, Grenada
| | - David L Duffy
- QIMR Berghofer Medical Research Institute, Herston QLD, Australia
| |
Collapse
|
18
|
Pyasi K, Tufvesson E, Moitra S. Evaluating the role of leukotriene-modifying drugs in asthma management: Are their benefits 'losing in translation'? Pulm Pharmacol Ther 2016; 41:52-59. [PMID: 27651322 DOI: 10.1016/j.pupt.2016.09.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 09/13/2016] [Accepted: 09/16/2016] [Indexed: 02/06/2023]
Abstract
Leukotrienes (LTs) initiate a cascade of reactions that cause bronchoconstriction and inflammation in asthma. LT-modifying drugs have been proved very effective to reduce inflammation and associated exacerbation however despite some illustrious clinical trials the usage of these drugs remains overlooked because the evidence to support their utility in asthma management has been mixed and varied between studies. Although, there are plenty of evidences which suggest that the leukotriene-modifying drugs provide consistent improvement even after just the first oral dose and reduce asthma exacerbations, the beneficial effect of these drugs has remained sparse and widely debated. And these beneficial effects are often overlooked because most of the clinical studies include a mixed population of asthmatics who do not respond to LT-modifiers equally. Therefore, in the present era of personalized medicine, it is important to properly stratify the patients and non-invasive measurements of biomarkers may warrant the possibility to characterize biological/pathological pathway to direct treatment to those who will benefit from it. Endotyping based on individual's leukotriene levels should probably ascertain a subgroup of patients that would clearly benefit from the treatment even though the trial fails to show overall significance. In this article, we have methodically evaluated contemporary literature describing the efficacy of LT-modifying drugs in the management of asthma and highlighted the importance of phenotyping the asthmatics for better treatment outcomes.
Collapse
Affiliation(s)
- Kanchan Pyasi
- Molecular Respiratory Research Laboratory, Chest Research Foundation, Pune, India
| | - Ellen Tufvesson
- Department of Respiratory Medicine and Allergology, Lund University, Lund, Sweden
| | - Subhabrata Moitra
- Department of Respiratory Medicine and Allergology, Lund University, Lund, Sweden; Department of Pneumology, Allergy and Asthma Research Centre, Kolkata, India.
| |
Collapse
|
19
|
Kittana N, Hattab S, Ziyadeh-Isleem A, Jaradat N, Zaid AN. Montelukast, current indications and prospective future applications. Expert Rev Respir Med 2016; 10:943-56. [PMID: 27485393 DOI: 10.1080/17476348.2016.1207533] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Montelukast is recommended for the treatment of asthma, exercise -induced bronchospasm and allergic rhinitis. Several trials demonstrated potential therapeutic effects in other respiratory conditions, and different animal-model-based studies explored potential pharmacological actions in non-respiratory conditions. AREAS COVERED Clinical investigations on the pharmacotherapeutic effects of montelukast, in addition to in-vivo studies on animal models of non-respiratory diseases. The data discussed in this review were mainly obtained from clinical randomized trials, real-life studies, and studies based on animal models as approve of concept. As a condition, all of the discussed articles were published in journals cited by Pubmed. Expert commentary: The current clinical data are in favor of montelukast use in the management of chronic asthma as an add-on or alternative therapy to the inhaled corticosteroids. Further clinical trials are required to confirm the effectiveness and feasibility of montelukast for the treatment of conditions other than the current clinical indications.
Collapse
Affiliation(s)
- Naim Kittana
- a Division of Pharmacology and Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences , An-Najah National University , Nablus , Palestine
| | - Suhaib Hattab
- a Division of Pharmacology and Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences , An-Najah National University , Nablus , Palestine
| | - Azza Ziyadeh-Isleem
- a Division of Pharmacology and Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences , An-Najah National University , Nablus , Palestine
| | - Nidal Jaradat
- b Division of Pharmaceutical Chemistry and Technology, Department of Pharmacy, Faculty of Medicine and Health Sciences , An-Najah National University , Nablus , Palestine
| | - Abdel-Naser Zaid
- b Division of Pharmaceutical Chemistry and Technology, Department of Pharmacy, Faculty of Medicine and Health Sciences , An-Najah National University , Nablus , Palestine
| |
Collapse
|
20
|
Reynolds AL, Alvarez Y, Sasore T, Waghorne N, Butler CT, Kilty C, Smith AJ, McVicar C, Wong VHY, Galvin O, Merrigan S, Osman J, Grebnev G, Sjölander A, Stitt AW, Kennedy BN. Phenotype-based Discovery of 2-[(E)-2-(Quinolin-2-yl)vinyl]phenol as a Novel Regulator of Ocular Angiogenesis. J Biol Chem 2016; 291:7242-55. [PMID: 26846851 DOI: 10.1074/jbc.m115.710665] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Indexed: 12/21/2022] Open
Abstract
Retinal angiogenesis is tightly regulated to meet oxygenation and nutritional requirements. In diseases such as proliferative diabetic retinopathy and neovascular age-related macular degeneration, uncontrolled angiogenesis can lead to blindness. Our goal is to better understand the molecular processes controlling retinal angiogenesis and discover novel drugs that inhibit retinal neovascularization. Phenotype-based chemical screens were performed using the ChemBridge Diverset(TM)library and inhibition of hyaloid vessel angiogenesis in Tg(fli1:EGFP) zebrafish. 2-[(E)-2-(Quinolin-2-yl)vinyl]phenol, (quininib) robustly inhibits developmental angiogenesis at 4-10 μmin zebrafish and significantly inhibits angiogenic tubule formation in HMEC-1 cells, angiogenic sprouting in aortic ring explants, and retinal revascularization in oxygen-induced retinopathy mice. Quininib is well tolerated in zebrafish, human cell lines, and murine eyes. Profiling screens of 153 angiogenic and inflammatory targets revealed that quininib does not directly target VEGF receptors but antagonizes cysteinyl leukotriene receptors 1 and 2 (CysLT1-2) at micromolar IC50values. In summary, quininib is a novel anti-angiogenic small-molecule CysLT receptor antagonist. Quininib inhibits angiogenesis in a range of cell and tissue systems, revealing novel physiological roles for CysLT signaling. Quininib has potential as a novel therapeutic agent to treat ocular neovascular pathologies and may complement current anti-VEGF biological agents.
Collapse
Affiliation(s)
- Alison L Reynolds
- From the University College Dublin School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Yolanda Alvarez
- From the University College Dublin School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Temitope Sasore
- From the University College Dublin School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Nora Waghorne
- From the University College Dublin School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Clare T Butler
- From the University College Dublin School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Claire Kilty
- From the University College Dublin School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Andrew J Smith
- From the University College Dublin School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Carmel McVicar
- the Centre for Experimental Medicine, Queen's University Belfast, Wellcome-Wolfson Building, 97 Lisburn Road, Belfast, BT9 7BL, United Kingdom, and
| | - Vickie H Y Wong
- the Centre for Experimental Medicine, Queen's University Belfast, Wellcome-Wolfson Building, 97 Lisburn Road, Belfast, BT9 7BL, United Kingdom, and
| | - Orla Galvin
- From the University College Dublin School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Stephanie Merrigan
- From the University College Dublin School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Janina Osman
- the Division of Cell and Experimental Pathology, Department of Translational Medicine, Lund University, Skåne University Hospital, 20502 Malmö, Sweden
| | - Gleb Grebnev
- From the University College Dublin School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Anita Sjölander
- the Division of Cell and Experimental Pathology, Department of Translational Medicine, Lund University, Skåne University Hospital, 20502 Malmö, Sweden
| | - Alan W Stitt
- the Centre for Experimental Medicine, Queen's University Belfast, Wellcome-Wolfson Building, 97 Lisburn Road, Belfast, BT9 7BL, United Kingdom, and
| | - Breandán N Kennedy
- From the University College Dublin School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland,
| |
Collapse
|
21
|
Cardoso JDO, Oliveira RV, Lu JBL, Desta Z. In Vitro Metabolism of Montelukast by Cytochrome P450s and UDP-Glucuronosyltransferases. Drug Metab Dispos 2015; 43:1905-16. [PMID: 26374173 PMCID: PMC4658491 DOI: 10.1124/dmd.115.065763] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Accepted: 09/14/2015] [Indexed: 11/22/2022] Open
Abstract
Montelukast has been recommended as a selective in vitro and in vivo probe of cytochrome P450 (P450) CYP2C8 activity, but its selectivity toward this enzyme remains unclear. We performed detailed characterization of montelukast metabolism in vitro using human liver microsomes (HLMs), expressed P450s, and uridine 5'-diphospho-glucuronosyltransferases (UGTs). Kinetic and inhibition experiments performed at therapeutically relevant concentrations reveal that CYP2C8 and CYP2C9 are the principal enzymes responsible for montelukast 36-hydroxylation to 1,2-diol. CYP3A4 was the main catalyst of montelukast sulfoxidation and stereoselective 21-hydroxylation, and multiple P450s participated in montelukast 25-hydroxylation. We confirmed direct glucuronidation of montelukast to an acyl-glucuronide. We also identified a novel peak that appears consistent with an ether-glucuronide. Kinetic analysis in HLMs and experiments in expressed UGTs indicate that both metabolites were exclusively formed by UGT1A3. Comparison of in vitro intrinsic clearance in HLMs suggest that direct glucuronidation may play a greater role in the overall metabolism of montelukast than does P450-mediated oxidation, but the in vivo contribution of UGT1A3 needs further testing. In conclusion, our in vitro findings provide new insight toward montelukast metabolism. The utility of montelukast as a probe of CYP2C8 activity may be compromised owing to involvement of multiple P450s and UGT1A3 in its metabolism.
Collapse
|
22
|
Marschallinger J, Schäffner I, Klein B, Gelfert R, Rivera FJ, Illes S, Grassner L, Janssen M, Rotheneichner P, Schmuckermair C, Coras R, Boccazzi M, Chishty M, Lagler FB, Renic M, Bauer HC, Singewald N, Blümcke I, Bogdahn U, Couillard-Despres S, Lie DC, Abbracchio MP, Aigner L. Structural and functional rejuvenation of the aged brain by an approved anti-asthmatic drug. Nat Commun 2015; 6:8466. [PMID: 26506265 PMCID: PMC4639806 DOI: 10.1038/ncomms9466] [Citation(s) in RCA: 135] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 08/24/2015] [Indexed: 01/19/2023] Open
Abstract
As human life expectancy has improved rapidly in industrialized societies, age-related cognitive impairment presents an increasing challenge. Targeting histopathological processes that correlate with age-related cognitive declines, such as neuroinflammation, low levels of neurogenesis, disrupted blood–brain barrier and altered neuronal activity, might lead to structural and functional rejuvenation of the aged brain. Here we show that a 6-week treatment of young (4 months) and old (20 months) rats with montelukast, a marketed anti-asthmatic drug antagonizing leukotriene receptors, reduces neuroinflammation, elevates hippocampal neurogenesis and improves learning and memory in old animals. By using gene knockdown and knockout approaches, we demonstrate that the effect is mediated through inhibition of the GPR17 receptor. This work illustrates that inhibition of leukotriene receptor signalling might represent a safe and druggable target to restore cognitive functions in old individuals and paves the way for future clinical translation of leukotriene receptor inhibition for the treatment of dementias. The leukotriene receptor antagonist montelukast is an anti-asthmatic drug. Here, the authors show that montelukast reduces neuroinflammation, promotes hippocampal neurogenesis and restores learning and memory in old rats suffering from ageing-associated cognitive dysfunction.
Collapse
Affiliation(s)
- Julia Marschallinger
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, 5020 Salzburg, Austria.,Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, 5020 Salzburg, Austria
| | - Iris Schäffner
- Institute of Biochemistry, Emil Fischer Center, Friedrich-Alexander-University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Barbara Klein
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, 5020 Salzburg, Austria.,Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, 5020 Salzburg, Austria
| | - Renate Gelfert
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, 5020 Salzburg, Austria.,Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, 5020 Salzburg, Austria
| | - Francisco J Rivera
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, 5020 Salzburg, Austria.,Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, 5020 Salzburg, Austria
| | - Sebastian Illes
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, 5020 Salzburg, Austria.,Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, 5020 Salzburg, Austria
| | - Lukas Grassner
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, 5020 Salzburg, Austria.,Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, 5020 Salzburg, Austria.,Center for Spinal Cord Injuries, BG Trauma Center Murnau, 82418 Murnau am Staffelsee, Germany
| | - Maximilian Janssen
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, 5020 Salzburg, Austria.,Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, 5020 Salzburg, Austria
| | - Peter Rotheneichner
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, 5020 Salzburg, Austria.,Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, 5020 Salzburg, Austria.,Institute of Experimental Neuroregeneration, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Claudia Schmuckermair
- Department of Pharmacology and Toxicology, Institute of Pharmacy and CMBI, Leopold-Franzens-University of Innsbruck, 6020 Innsbruck, Austria
| | - Roland Coras
- Department of Neuropathology, University Hospital Erlangen, 91054 Erlangen, Germany
| | - Marta Boccazzi
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy
| | | | - Florian B Lagler
- Department for Paediatrics, Institute for Inborn Errors of Metabolism, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Marija Renic
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
| | - Hans-Christian Bauer
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, 5020 Salzburg, Austria.,Institute of Tendon and Bone Regeneration, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Nicolas Singewald
- Department of Pharmacology and Toxicology, Institute of Pharmacy and CMBI, Leopold-Franzens-University of Innsbruck, 6020 Innsbruck, Austria
| | - Ingmar Blümcke
- Department of Neuropathology, University Hospital Erlangen, 91054 Erlangen, Germany
| | - Ulrich Bogdahn
- Department of Neurology, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Sebastien Couillard-Despres
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, 5020 Salzburg, Austria.,Institute of Experimental Neuroregeneration, Paracelsus Medical University, 5020 Salzburg, Austria
| | - D Chichung Lie
- Institute of Biochemistry, Emil Fischer Center, Friedrich-Alexander-University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Maria P Abbracchio
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy
| | - Ludwig Aigner
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, 5020 Salzburg, Austria.,Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, 5020 Salzburg, Austria
| |
Collapse
|
23
|
Sekioka T, Kadode M, Fujii M, Kawabata K, Abe T, Horiba M, Kohno S, Nabe T. Expression of CysLT2 receptors in asthma lung, and their possible role in bronchoconstriction. Allergol Int 2015; 64:351-8. [PMID: 26433531 DOI: 10.1016/j.alit.2015.04.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 04/13/2015] [Accepted: 04/14/2015] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The expression and functional role of CysLT2 receptors in asthma have not been clarified. In this study, we evaluated CysLT2 receptors expression, and effects of CysLT2-and CysLT1/2-receptor antagonists on antigen-induced bronchoconstriction using isolated lung tissues from both asthma and non-asthma subjects. METHODS CysLT1 and CysLT2 receptors expression in asthma and non-asthma lung tissue preparations was examined in immunohistochemistry experiments, and their functional roles in antigen-induced bronchoconstriction were assessed using ONO-6950, a dual CysLT1/2-receptor antagonist, montelukast, a CysLT1 receptor antagonist, and BayCysLT2RA, a CysLT2 receptor-specific antagonist. RESULTS CysLT1 receptors were expressed on the bronchial smooth muscle and epithelium, and on alveolar leukocytes in 5 in 5 non-asthma subjects and 2 in 2 asthma subjects. On the other hand, although degrees of CysLT2 receptors expression were variable among the 5 non-asthma subjects, the expression in the asthma lung was detected on bronchial smooth muscle, epithelium and alveolar leukocytes in 2 in 2 asthma subjects. In the non-asthma specimens, antagonism of CysLT2 receptors did not affect antigen-induced bronchial contractions, even after pretreatment with the CysLT1-receptor specific antagonist, montelukast. However, in the bronchus isolated from one of the 2 asthma subjects, antagonism of CysLT2 receptors suppressed contractions, and dual antagonism of CysLT1 and CysLT2 receptors resulted in additive inhibitory effect on anaphylactic contractions. CONCLUSIONS CysLT2 receptors were expressed in lung specimens isolated from asthma subjects. Activation of CysLT2 receptors may contribute to antigen-induced bronchoconstriction in certain asthma population.
Collapse
Affiliation(s)
- Tomohiko Sekioka
- Discovery Research Laboratories II, Department of Biology & Pharmacology, Ono Pharmaceutical Co., Ltd., Osaka, Japan; Department of Pharmacology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Michiaki Kadode
- Discovery Research Laboratories II, Department of Biology & Pharmacology, Ono Pharmaceutical Co., Ltd., Osaka, Japan; Department of Pharmacology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Masanori Fujii
- Department of Pharmacology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Kazuhito Kawabata
- Discovery Research Laboratories II, Department of Biology & Pharmacology, Ono Pharmaceutical Co., Ltd., Osaka, Japan
| | - Takashi Abe
- Department of Pneumology, Ogaki Municipal Hospital, Gifu, Japan
| | - Michiaki Horiba
- Department of Pneumology, Ogaki Municipal Hospital, Gifu, Japan
| | - Shigekatsu Kohno
- Department of Pharmacology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Takeshi Nabe
- Laboratory of Immunopharmacology, Faculty of Pharmaceutical Sciences, Setsunan University, Osaka, Japan; Department of Pharmacology, Kyoto Pharmaceutical University, Kyoto, Japan.
| |
Collapse
|
24
|
Platt M. Pharmacotherapy for allergic rhinitis. Int Forum Allergy Rhinol 2015; 4 Suppl 2:S35-40. [PMID: 25182353 DOI: 10.1002/alr.21381] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2014] [Revised: 07/01/2014] [Accepted: 07/02/2014] [Indexed: 12/19/2022]
Abstract
BACKGROUND Pharmacotherapy for allergic rhinitis is a mainstay of treatment for patients with mild to severe nasal allergy symptoms. A wide array of medical treatment options is available for both episodic relief and prevention of symptoms. Treatment regimens can be tailored to individual patients based on nasal symptoms, severity, and associated atopic disorders. The purposes of this review are to identify available pharmacotherapies for allergic rhinitis, to discuss the benefits and limitations of each treatment option, and to help guide practitioners in providing optimal medical treatment for patients with allergic rhinitis. METHODS A comprehensive review of pharmacotherapies for allergic rhinitis was performed using a PubMed search. Secondary sources within indexed studies were also compiled to review current medication options for patients with allergic rhinitis. The benefits and limitations of each class of allergy medication were reviewed to provide information on selecting the optimal treatment regimen for patients with allergic rhinitis. RESULTS Pharmacotherapies for allergic rhinitis that are currently used in clinical practice include antihistamines, corticosteroids, leukotriene modifiers, mast cell stabilizers, expectorants, and decongestants. Symptoms of nasal congestion, itching, sneezing, and rhinorrhea can be targeted with specific therapies that modulate the acute-phase or late-phase allergic reactions. Associated atopic disorders, including conjunctivitis and asthma, can help guide medication selection. CONCLUSION Pharmacotherapies for allergic rhinitis offer numerous options that are safe, effective, and readily available to target specific nasal symptoms. Symptom-based selection of allergy medications can result in optimal treatment for patients with allergic rhinitis.
Collapse
Affiliation(s)
- Michael Platt
- Department of Otolaryngology-Head and Neck Surgery, Boston University School of Medicine, Boston, MA
| |
Collapse
|
25
|
Thompson MD, Cole DEC, Capra V, Siminovitch KA, Rovati GE, Burnham WM, Rana BK. Pharmacogenetics of the G protein-coupled receptors. Methods Mol Biol 2014; 1175:189-242. [PMID: 25150871 DOI: 10.1007/978-1-4939-0956-8_9] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Pharmacogenetics investigates the influence of genetic variants on physiological phenotypes related to drug response and disease, while pharmacogenomics takes a genome-wide approach to advancing this knowledge. Both play an important role in identifying responders and nonresponders to medication, avoiding adverse drug reactions, and optimizing drug dose for the individual. G protein-coupled receptors (GPCRs) are the primary target of therapeutic drugs and have been the focus of these studies. With the advance of genomic technologies, there has been a substantial increase in the inventory of naturally occurring rare and common GPCR variants. These variants include single-nucleotide polymorphisms and insertion or deletions that have potential to alter GPCR expression of function. In vivo and in vitro studies have determined functional roles for many GPCR variants, but genetic association studies that define the physiological impact of the majority of these common variants are still limited. Despite the breadth of pharmacogenetic data available, GPCR variants have not been included in drug labeling and are only occasionally considered in optimizing clinical use of GPCR-targeted agents. In this chapter, pharmacogenetic and genomic studies on GPCR variants are reviewed with respect to a subset of GPCR systems, including the adrenergic, calcium sensing, cysteinyl leukotriene, cannabinoid CB1 and CB2 receptors, and the de-orphanized receptors such as GPR55. The nature of the disruption to receptor function is discussed with respect to regulation of gene expression, expression on the cell surface (affected by receptor trafficking, dimerization, desensitization/downregulation), or perturbation of receptor function (altered ligand binding, G protein coupling, constitutive activity). The large body of experimental data generated on structure and function relationships and receptor-ligand interactions are being harnessed for the in silico functional prediction of naturally occurring GPCR variants. We provide information on online resources dedicated to GPCRs and present applications of publically available computational tools for pharmacogenetic studies of GPCRs. As the breadth of GPCR pharmacogenomic data becomes clearer, the opportunity for routine assessment of GPCR variants to predict disease risk, drug response, and potential adverse drug effects will become possible.
Collapse
Affiliation(s)
- Miles D Thompson
- Department of Pharmacology and Toxicology, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, ON, Canada, M5S 1A8,
| | | | | | | | | | | | | |
Collapse
|
26
|
Long EK, Hellberg K, Foncea R, Hertzel AV, Suttles J, Bernlohr DA. Fatty acids induce leukotriene C4 synthesis in macrophages in a fatty acid binding protein-dependent manner. Biochim Biophys Acta Mol Cell Biol Lipids 2013; 1831:1199-207. [PMID: 23583845 DOI: 10.1016/j.bbalip.2013.04.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Revised: 04/02/2013] [Accepted: 04/05/2013] [Indexed: 12/30/2022]
Abstract
Obesity results in increased macrophage recruitment to adipose tissue that promotes a chronic low-grade inflammatory state linked to increased fatty acid efflux from adipocytes. Activated macrophages produce a variety of pro-inflammatory lipids such as leukotriene C4 (LTC4) and 5-, 12-, and 15-hydroxyeicosatetraenoic acid (HETE) suggesting the hypothesis that fatty acids may stimulate eicosanoid synthesis. To assess if eicosanoid production increases with obesity, adipose tissue of leptin deficient ob/ob mice was analyzed. In ob/ob mice, LTC4 and 12-HETE levels increased in the visceral (but not subcutaneous) adipose depot while the 5-HETE levels decreased and 15-HETE abundance was unchanged. Since macrophages produce the majority of inflammatory molecules in adipose tissue, treatment of RAW264.7 or primary peritoneal macrophages with free fatty acids led to increased secretion of LTC4 and 5-HETE, but not 12- or 15-HETE. Fatty acid binding proteins (FABPs) facilitate the intracellular trafficking of fatty acids and other hydrophobic ligands and in vitro stabilize the LTC4 precursor leukotriene A4 (LTA4) from non-enzymatic hydrolysis. Consistent with a role for FABPs in LTC4 synthesis, treatment of macrophages with HTS01037, a specific FABP inhibitor, resulted in a marked decrease in both basal and fatty acid-stimulated LTC4 secretion but no change in 5-HETE production or 5-lipoxygenase expression. These results indicate that the products of adipocyte lipolysis may stimulate the 5-lipoxygenase pathway leading to FABP-dependent production of LTC4 and contribute to the insulin resistant state.
Collapse
Affiliation(s)
- Eric K Long
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455 USA
| | | | | | | | | | | |
Collapse
|
27
|
Zaid AN, Natour S, Qaddomi A, Abu Ghoush A. Formulation and in vitro and in vivo evaluation of film-coated montelukast sodium tablets using Opadry® yellow 20A82938 on an industrial scale. DRUG DESIGN DEVELOPMENT AND THERAPY 2013; 7:83-91. [PMID: 23430138 PMCID: PMC3573806 DOI: 10.2147/dddt.s37369] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Indexed: 11/23/2022]
Abstract
Purpose: The aim of this study was to formulate stable film-coated montelukast sodium (MS) tablets using Opadry® yellow 20A82938 (Montikast® tablets) and to evaluate their in vitro and in vivo release profile. Methods: MS core tablets were manufactured using a direct compression method. Opadry yellow 20A82938 aqueous coating dispersion was used as the film-coating material. Dissolution of the film-coated tablets was tested in 900 mL of 0.5% sodium lauryl sulfate solution and the bioequivalence of the tablets was tested by comparing them with a reference formulation – Singulair® tablets. In vitro–in vivo correlation was evaluated. The stability of the obtained film-coated tablets was evaluated according to International Conference on Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use guidelines. Results: The efficiency of the film coating was determined by subjecting the coated tablets to gastric pH and drug release was analyzed using high-performance liquid chromatography. The coated tablets had no obvious defects. MS release met the study criterion of not less than 80% dissolved after 30 minutes in 0.5% sodium lauryl sulfate solution. Statistical comparison of the main pharmacokinetic parameters clearly indicated no significant difference between test and reference in any of the calculated pharmacokinetic parameters. Level A correlation between in vitro drug release and in vivo absorption was found to be satisfactory. Conclusion: These findings suggest that aqueous film coating with Opadry yellow 20A82938 is an easy, reproducible, and economical approach for preparing stable MS film-coated tablets without affecting the drug-release characteristics.
Collapse
Affiliation(s)
- Abdel Naser Zaid
- Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine.
| | | | | | | |
Collapse
|
28
|
Chauhan BF, Ducharme FM, Cochrane Airways Group. Anti-leukotriene agents compared to inhaled corticosteroids in the management of recurrent and/or chronic asthma in adults and children. Cochrane Database Syst Rev 2012; 2012:CD002314. [PMID: 22592685 PMCID: PMC4164381 DOI: 10.1002/14651858.cd002314.pub3] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Anti-leukotrienes (5-lipoxygenase inhibitors and leukotriene receptors antagonists) serve as alternative monotherapy to inhaled corticosteroids (ICS) in the management of recurrent and/or chronic asthma in adults and children. OBJECTIVES To determine the safety and efficacy of anti-leukotrienes compared to inhaled corticosteroids as monotherapy in adults and children with asthma and to provide better insight into the influence of patient and treatment characteristics on the magnitude of effects. SEARCH METHODS We searched MEDLINE (1966 to Dec 2010), EMBASE (1980 to Dec 2010), CINAHL (1982 to Dec 2010), the Cochrane Airways Group trials register, and the Cochrane Central Register of Controlled Trials (Dec 2010), abstract books, and reference lists of review articles and trials. We contacted colleagues and the international headquarters of anti-leukotrienes producers. SELECTION CRITERIA We included randomised trials that compared anti-leukotrienes with inhaled corticosteroids as monotherapy for a minimum period of four weeks in patients with asthma aged two years and older. DATA COLLECTION AND ANALYSIS Two review authors independently assessed the methodological quality of trials and extracted data. The primary outcome was the number of patients with at least one exacerbation requiring systemic corticosteroids. Secondary outcomes included patients with at least one exacerbation requiring hospital admission, lung function tests, indices of chronic asthma control, adverse effects, withdrawal rates and biological inflammatory markers. MAIN RESULTS Sixty-five trials met the inclusion criteria for this review. Fifty-six trials (19 paediatric trials) contributed data (representing total of 10,005 adults and 3,333 children); 21 trials were of high methodological quality; 44 were published in full-text. All trials pertained to patients with mild or moderate persistent asthma. Trial durations varied from four to 52 weeks. The median dose of inhaled corticosteroids was quite homogeneous at 200 µg/day of microfine hydrofluoroalkane-propelled beclomethasone or equivalent (HFA-BDP eq). Patients treated with anti-leukotrienes were more likely to suffer an exacerbation requiring systemic corticosteroids (N = 6077 participants; risk ratio (RR) 1.51, 95% confidence interval (CI) 1.17, 1.96). For every 28 (95% CI 15 to 82) patients treated with anti-leukotrienes instead of inhaled corticosteroids, there was one additional patient with an exacerbation requiring rescue systemic corticosteroids. The magnitude of effect was significantly greater in patients with moderate compared with those with mild airway obstruction (RR 2.03, 95% CI 1.41, 2.91 versus RR 1.25, 95% CI 0.97, 1.61), but was not significantly influenced by age group (children representing 23% of the weight versus adults), anti-leukotriene used, duration of intervention, methodological quality, and funding source. Significant group differences favouring inhaled corticosteroids were noted in most secondary outcomes including patients with at least one exacerbation requiring hospital admission (N = 2715 participants; RR 3.33; 95% CI 1.02 to 10.94), the change from baseline FEV(1) (N = 7128 participants; mean group difference (MD) 110 mL, 95% CI 140 to 80) as well as other lung function parameters, asthma symptoms, nocturnal awakenings, rescue medication use, symptom-free days, the quality of life, parents' and physicians' satisfaction. Anti-leukotriene therapy was associated with increased risk of withdrawals due to poor asthma control (N = 7669 participants; RR 2.56; 95% CI 2.01 to 3.27). For every thirty one (95% CI 22 to 47) patients treated with anti-leukotrienes instead of inhaled corticosteroids, there was one additional withdrawal due to poor control. Risk of side effects was not significantly different between both groups. AUTHORS' CONCLUSIONS As monotherapy, inhaled corticosteroids display superior efficacy to anti-leukotrienes in adults and children with persistent asthma; the superiority is particularly marked in patients with moderate airway obstruction. On the basis of efficacy, the results support the current guidelines' recommendation that inhaled corticosteroids remain the preferred monotherapy.
Collapse
Affiliation(s)
- Bhupendrasinh F Chauhan
- Research Centre, CHU Sainte‐JustineClinical Research Unit on Childhood Asthma3175, Cote Sainte‐CatherineMontrealQCCanada
| | - Francine M Ducharme
- University of MontrealDepartment of PaediatricsMontrealQCCanada
- CHU Sainte‐JustineResearch CentreMontrealCanada
| | | |
Collapse
|
29
|
Blanc FX. [Antileukotrienes in asthma: a nice story, still to be completed]. Rev Mal Respir 2011; 28:701-3. [PMID: 21742231 DOI: 10.1016/j.rmr.2011.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2011] [Accepted: 05/19/2011] [Indexed: 11/17/2022]
|
30
|
Shin YS, Takeda K, Ohnishi H, Jia Y, Shiraishi Y, Cox ML, Fine JS, Rosenblum S, Lundel D, Jenh CH, Manfra DJ, Gelfand EW. Targeting CXCR3 reduces ligand-induced T-cell activation but not development of lung allergic responses. Ann Allergy Asthma Immunol 2011; 107:145-53. [PMID: 21802023 DOI: 10.1016/j.anai.2011.04.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2011] [Revised: 03/29/2011] [Accepted: 04/26/2011] [Indexed: 10/18/2022]
Abstract
BACKGROUND Asthma is a chronic airway inflammatory disease that is associated with a large influx of inflammatory cells. Several chemokines and chemokine receptors play critical roles in the development of allergic airway inflammation. OBJECTIVE Because polarized human T(H)2 cells express a functional CXCR3 chemokine receptor, we evaluated the effects of a selective CXCR3 inhibitor in a mouse model of allergic airway disease. METHODS Ovalbumin-specific CD8(+) T effector cells were generated from OT-1 mice in the presence of interleukin 2. The activity of a CXCR3 inhibitor was examined in vitro by monitoring Ca(2+) influx after receptor ligation. In vivo, the activity was assessed in sensitized and challenged mice by monitoring airway function, inflammatory parameters, including cellular infiltrates and cytokines in the bronchoalveolar lavage fluid. RESULTS Approximately 40% of CD8(+) T effector cells expressed the CXCR3 receptor. In vitro, CXCR3 antagonism reduced Ca(2+) influx after receptor engagement. In contrast, the CXCR3 antagonist had little to no effect on airway function or inflammatory parameters despite adequate exposure levels. CONCLUSIONS CXCR3 antagonism did not prevent allergen-induced airway hyperresponsiveness or airway inflammation in a mouse allergy model despite having activity in in vitro functional assays.
Collapse
Affiliation(s)
- Yoo Seob Shin
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, Colorado 80206, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Kazani S, Sadeh J, Bunga S, Wechsler ME, Israel E. Cysteinyl leukotriene antagonism inhibits bronchoconstriction in response to hypertonic saline inhalation in asthma. Respir Med 2011; 105:667-73. [PMID: 21169002 PMCID: PMC3080101 DOI: 10.1016/j.rmed.2010.11.025] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2010] [Revised: 11/21/2010] [Accepted: 11/23/2010] [Indexed: 11/26/2022]
Abstract
BACKGROUND In asthma, cysteinyl leukotrienes (CysLTs) play varying roles in the bronchomotor response to multiple provocative stimuli. The contribution of CysLTs on the airway's response to hypertonic saline (HS) inhalation in asthma is unknown. Whether polymorphisms in the leukotriene biosynthetic pathway affect the contribution of CysLTs to this response is also unknown. METHODS In a prospective, randomized, double-blind, placebo-controlled cross-over study, mild and moderate asymptomatic asthmatics underwent inhaled 3% HS challenge by doubling the duration of nebulization (0.5, 1, 2, 4, and 8 min) 2 h after one dose of montelukast (a CysLT receptor 1 [CysLTR1] antagonist) or placebo, and after three-week courses. We examined the effect of the leukotriene C(4) synthase (LTC(4)S) polymorphism (A-444C) on the efficacy of montelukast against HS inhalation in an exploratory manner. RESULTS In 37 subjects, 2 h after administration of montelukast, the mean provocative dose of HS required to cause a 20% drop in FEV(1) (HS-PD(20)) increased by 59% (9.17 ml after placebo vs. 14.55 ml after montelukast, p=0.0154). Three weeks of cysLTR1 antagonism increased the HS-PD(20) by 84% (10.97 vs. 20.21 ml, p=0.0002). Three weeks of CysLTR1 antagonism appeared to produce greater effects on blocking bronchial hyper-responsiveness (2 h vs. three-week HS-PD(20) values 14.55 vs. 20.21 ml respectively, p=0.0898). We did not observe an effect of the LTC(4)S polymorphism on the response to CysLTR1 antagonism in this cohort. CONCLUSIONS A significant proportion of HS-induced bronchoconstriction is mediated by release of leukotrienes as evidenced by substantial acute inhibition with a CysLTR1 antagonist. There was a trend toward greater inhibition of bronchial responsiveness with three weeks of therapy as opposed to acute CysLTR1 antagonism. Clinicaltrials.gov registration number NCT00116324.
Collapse
Affiliation(s)
- Shamsah Kazani
- Pulmonary and Critical Care Division, Department of Medicine, Brigham and Women's Hospital, PBB Clinics 3, 75 Francis Street, Boston, MA 02115, USA.
| | | | | | | | | |
Collapse
|
32
|
Zhao R, Shi WZ, Zhang YM, Fang SH, Wei EQ. Montelukast, a cysteinyl leukotriene receptor-1 antagonist, attenuates chronic brain injury after focal cerebral ischaemia in mice and rats. ACTA ACUST UNITED AC 2011; 63:550-7. [PMID: 21401607 DOI: 10.1111/j.2042-7158.2010.01238.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
OBJECTIVES Previously we demonstrated the neuroprotective effect of montelukast, a cysteinyl leukotriene receptor-1 (CysLT(1) ) antagonist, on acute brain injury after focal cerebral ischaemia in mice. In this study, we have determined its effect on chronic brain injury after focal cerebral ischaemia in mice and rats. METHODS After transient focal cerebral ischaemia was induced by middle cerebral artery occlusion, montelukast was intraperitoneally injected in mice or orally administered to rats for five days. Behavioural dysfunction, brain infarct volume, brain atrophy and neuron loss were determined to evaluate brain lesions. KEY FINDINGS Montelukast (0.1 mg/kg) attenuated behavioural dysfunction, brain infarct volume, brain atrophy and neuron loss in mice, which was similar to pranlukast, another CysLT(1) receptor antagonist. Oral montelukast (0.5 mg/kg) was effective in rats and was more effective than edaravone, a free radical scavenger. CONCLUSION Montelukast protected mice and rats against chronic brain injury after focal cerebral ischaemia, supporting the therapeutic potential of CysLT(1) receptor antagonists.
Collapse
Affiliation(s)
- Rui Zhao
- Department of Pharmacology, School of Medicine, Zhejiang University Department of Pharmacy, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou Department of Physiology and Department of Neurobiology, Xuzhou Medical College, Xuzhou, Jiangsu, China
| | | | | | | | | |
Collapse
|
33
|
Philip G, Pedinoff A, Vandormael K, Tymofyeyev Y, Smugar SS, Reiss TF, Korenblat PE. A phase I randomized, placebo-controlled, dose-exploration study of single-dose inhaled montelukast in patients with chronic asthma. J Asthma 2010; 47:1078-84. [PMID: 20936994 DOI: 10.3109/02770903.2010.520100] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND The efficacy of oral montelukast has been well established in asthma and allergic rhinitis in adults and children. The purpose of this study was to evaluate dose-related bronchodilation and tolerability of inhaled montelukast. METHODS Randomized, double-blind, crossover, adaptive-design study comparing single-dose administration of inhaled montelukast versus placebo in patients age 15-65 years with chronic asthma (n = 68). Montelukast was delivered as a witnessed dose through dry powder inhaler at doses of 25, 250, or 1000 μg, and doses of 50, 100, and 500 μg could be used if needed based on a prespecified dose-response algorithm. Each administration was followed by a 4- to 7-day washout period before crossing over to the next treatment. The primary endpoint was the change from baseline in a forced expiratory volume in 1 second (FEV₁) over the first 4 hours after administration, calculated as a time-weighted average (ΔFEV₁ [0-4 hours]). Other endpoints included the onset and duration of bronchodilation and the effect of albuterol when added to inhaled montelukast. RESULTS Over 4 hours postdose, and compared with placebo (least-squares [LS] mean 0.03 L), inhaled montelukast 100 μg (0.13 L; p ≤ .001), 250 μg (0.10 L; p < .01), and 1000 μg (0.12 L; p ≤ .001) had significantly greater ΔFEV₁ (0-4 hours). At 24 hours postdose, inhaled montelukast 100 μg (0.10 L) and 1000 μg (0.09 L) had significantly greater bronchodilation compared with placebo (0.02 L; p < .05 vs. montelukast). Montelukast 1000 μg provided significant bronchodilation versus placebo within 20 minutes of administration (0.03 L vs. -0.05 L), whereas montelukast 100 μg provided significant bronchodilation relative to placebo within 2 hours of dosing (0.09 L vs. 0.01 L). Montelukast (pooled doses) plus albuterol was significantly more effective than montelukast plus placebo for ΔFEV₁ (0-90 minutes) (0.34 L vs. 0.15 L; p = .015). The tolerability of inhaled montelukast was similar to that of placebo. No serious adverse experiences were reported. CONCLUSIONS Inhaled montelukast provided significant bronchodilation compared with placebo as early as 20 minutes after the administration that persisted for 24 hours and provided additive bronchodilation to albuterol.
Collapse
Affiliation(s)
- George Philip
- Merck Research Laboratories, North Wales, PA 19454, USA.
| | | | | | | | | | | | | |
Collapse
|
34
|
Matthew Hutzler J, Linder CD, Melton RJ, Vincent J, Daniels JS. In vitro-in vivo correlation and translation to the clinical outcome for CJ-13,610, a novel inhibitor of 5-lipoxygenase. Drug Metab Dispos 2010; 38:1113-21. [PMID: 20375180 DOI: 10.1124/dmd.110.032706] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
The metabolism of the 5-lipoxygenase inhibitor, 4-(3-(4-(2-methyl-1H-imidazol-1-yl)phenylthio)phenyl)-tetrahydro-2H-pyran-4-carboxamide (CJ-13,610), was investigated in liver microsomes from human and preclinical species in an effort to compare metabolite profiles and evaluate the in vitro-in vivo correlation for metabolic clearance. Overall, the metabolite profile of CJ-13,610 was comparable across the species tested with multiple oxidative metabolites observed, including sulfoxidation. The sulfoxidation kinetics characterized in rat, dog, and human liver microsomes (HLM) indicated a low apparent Michaelis-Menten constant (K(m, app)) of 4 to 5 microM. Results from cDNA-expressed cytochrome P450 (P450) studies indicated that the metabolism in HLM was primarily mediated by CYP3A4 and 3A5. A subsequent in vitro study using ketoconazole as an inhibitor of CJ-13,610 sulfoxidation corroborated the CYP3A4/5-mediated pathway (IC(50) = 7 nM). Assessment of multiple methods for predicting the human pharmacokinetic profile observed with CJ-13,610 after a 30-mg single oral dose indicated that clearance scaled from human liver microsomes yielded a better prediction when coupled with a Vd(ss) term that was scaled from dog [area under the concentration-time curve (AUC) and half-life within 1.3-fold of actual] versus a Vd(ss) term obtained from rat. Single-species allometric scaling of clearance and Vd(ss) from dog pharmacokinetic studies was equally predictive, whereas scaling from rat resulted in underpredictions of both AUC and maximal concentration (C(max)). Results from these studies support the strategy of predicting human pharmacokinetics using human liver microsomal intrinsic clearance data. More importantly, results from the present investigation enabled the selection of alternative drug candidates from the chemical series via in vitro screening, while subsequently eliminating costly routine preclinical in vivo studies.
Collapse
Affiliation(s)
- J Matthew Hutzler
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Global Research and Development, St. Louis Laboratories, St. Louis, Missouri, USA.
| | | | | | | | | |
Collapse
|
35
|
Gemfibrozil Markedly Increases the Plasma Concentrations of Montelukast: A Previously Unrecognized Role for CYP2C8 in the Metabolism of Montelukast. Clin Pharmacol Ther 2010; 88:223-30. [DOI: 10.1038/clpt.2010.73] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
36
|
Nials AT, Uddin S. Mouse models of allergic asthma: acute and chronic allergen challenge. Dis Model Mech 2009; 1:213-20. [PMID: 19093027 DOI: 10.1242/dmm.000323] [Citation(s) in RCA: 299] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Asthma is defined as a chronic inflammatory disease of the airways; however, the underlying physiological and immunological processes are not fully understood. Animal models have been used to elucidate asthma pathophysiology, and to identify and evaluate novel therapeutic targets. Several recent review articles (Epstein, 2004; Lloyd, 2007; Boyce and Austen, 2005; Zosky and Sly, 2007) have discussed the potential value of these models. Allergen challenge models reproduce many features of clinical asthma and have been widely used by investigators; however, the majority involve acute allergen challenge procedures. It is recognised that asthma is a chronic inflammatory disease resulting from continued or intermittent allergen exposure, usually via inhalation, and there has been a recent focus on developing chronic allergen exposure models, predominantly in mice. Here, we review the acute and chronic exposure mouse models, and consider their potential role and impact in the field of asthma research.
Collapse
Affiliation(s)
- Anthony T Nials
- Discovery Biology, Respiratory Centre of Excellence for Drug Discovery, GlaxoSmithKline Medicines Research Centre, Gunnels Wood Road, Stevenage SG1 2NY, UK.
| | | |
Collapse
|
37
|
Stewart RA, Ram B, Hamilton G, Weiner J, Kane KJ. Montelukast as an adjunct to oral and inhaled steroid therapy in chronic nasal polyposis. Otolaryngol Head Neck Surg 2009; 139:682-7. [PMID: 18984264 DOI: 10.1016/j.otohns.2008.07.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2007] [Revised: 03/24/2008] [Accepted: 07/08/2008] [Indexed: 10/21/2022]
Abstract
OBJECTIVE To examine the potential of montelukast, a leukotriene receptor antagonist, as an adjunct to oral and inhaled steroid in subjects with chronic nasal polyps. STUDY DESIGN Prospective, randomized controlled trial. SUBJECTS AND METHODS Thirty-eight consecutive adult patients with bilateral nasal polyps were randomized into two groups. Eighteen subjects were treated with oral prednisolone for 14 days and budenoside nasal spray for 8 weeks. Twenty subjects received similar treatment with additional oral montelukast for 8 weeks. Subjects completed a modified nasal ICSD symptom score at 8 and 12 weeks after beginning treatment and the SF-36 quality of life questionnaire at 12 weeks. RESULTS Symptom scores improved in both groups after treatment. Subjects treated with montelukast reported significantly less headache (P = 0.013), facial pain (P = 0.048) and sneezing (P = 0.03) than controls. Four weeks after completing treatment, no significant differences were recorded. CONCLUSION Montelukast therapy may have clinical benefit as an adjunct to oral and inhaled steroid in chronic nasal polyposis, but effects are not maintained after cessation of treatment.
Collapse
Affiliation(s)
- Rosemary A Stewart
- Department of Otolaryngology, Royal Victorian Eye & Ear Hospital, Melbourne, Australia
| | | | | | | | | |
Collapse
|
38
|
Bisgaard H. Pathophysiology of the cysteinyl leukotrienes and effects of leukotriene receptor antagonists in asthma. Allergy 2008. [DOI: 10.1111/j.1398-9995.2001.00002.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
39
|
Abstract
Common G protein-coupled receptor (GPCR) gene variants that encode receptor proteins with a distinct sequence may alter drug efficacy without always resulting in a disease phenotype. GPCR genetic loci harbor numerous variants, such as DNA insertions or deletions and single-nucleotide polymorphisms that alter GPCR expression and function, thereby contributing to interindividual differences in disease susceptibility/progression and drug responses. In this chapter, these pharmacogenetic phenomena are reviewed with respect to a limited sampling of GPCR systems, including the beta(2)-adrenergic receptors, the cysteinyl leukotriene receptors, and the calcium-sensing receptor. In each example, the nature of the disruption to receptor function that results from each variant is discussed with respect to the regulation of gene expression, expression on cell surface (affected by receptor trafficking, dimerization, desensitization/downregulation), or perturbation of receptor function (by altering ligand binding, G protein coupling, and receptor constitutive activity). Despite the breadth of pharmacogenetic knowledge available, assessment for genetic variants is only occasionally applied to drug development projects involving pharmacogenomics or to optimizing the clinical use of GPCR drugs. The continued effort by the basic science of pharmacogenetics may draw the attention of drug discovery projects and clinicians alike to the utility of personalized pharmacogenomics as a means to optimize novel GPCR drug targets.
Collapse
Affiliation(s)
- Miles D Thompson
- Department of Laboratory Medicine and Pathobiology, Banting Institute, University of Toronto, Toronto, Ontario, Canada
| | | | | |
Collapse
|
40
|
Thompson MD, Takasaki J, Capra V, Rovati GE, Siminovitch KA, Burnham WM, Hudson TJ, Bossé Y, Cole DEC. G-protein-coupled receptors and asthma endophenotypes: the cysteinyl leukotriene system in perspective. Mol Diagn Ther 2007; 10:353-66. [PMID: 17154652 DOI: 10.1007/bf03256212] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Genetic variation in specific G-protein coupled receptors (GPCRs) is associated with a spectrum of respiratory disease predispositions and drug response phenotypes. Although certain GPCR gene variants can be disease-causing through the expression of inactive, overactive, or constitutively active receptor proteins, many more GPCR gene variants confer risk for potentially deleterious endophenotypes. Endophenotypes are traits, such as bronchiole hyperactivity, atopy, and aspirin intolerant asthma, which have a strong genetic component and are risk factors for a variety of more complex outcomes that may include disease states. GPCR genes implicated in asthma endophenotypes include variants of the cysteinyl leukotriene receptors (CYSLTR1 and CYSLTR2), and prostaglandin D2 receptors (PTGDR and CRTH2), thromboxane A2 receptor (TBXA2R), beta2-adrenergic receptor (ADRB2), chemokine receptor 5 (CCR5), and the G protein-coupled receptor associated with asthma (GPRA). This review of the contribution of variability in these genes places the contribution of the cysteinyl leukotriene system to respiratory endophenotypes in perspective. The genetic variant(s) of receptors that are associated with endophenotypes are discussed in the context of the extent to which they contribute to a disease phenotype or altered drug efficacy.
Collapse
Affiliation(s)
- Miles D Thompson
- Department of Laboratory Medicine and Pathobiology, University of Toronto, and Department of Medical Genetics and Microbiology, Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
The ultimate objective of biomedical research is to connect human diseases with the genes that underlie them and drugs that treat them. But this remains a daunting task, and even the most inspired researchers still have to resort to laborious screens of genetic or chemical libraries. What if at least some parts of this screening process could be systematized and centralized? And hits found and hypotheses generated with something resembling an internet search engine? These are the questions the Connectivity Map project set out to answer.
Collapse
Affiliation(s)
- Justin Lamb
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts 02142, USA.
| |
Collapse
|
42
|
Wu Y, Zhou C, Tao J, Li S. Montelukast prevents the decrease of interleukin-10 and inhibits NF-kappaB activation in inflammatory airway of asthmatic guinea pigs. Can J Physiol Pharmacol 2006; 84:531-7. [PMID: 16902598 DOI: 10.1139/y06-003] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Interleukin (IL)-10 is an important immunoregulatory and anti-inflammatory cytokine, whereas nuclear factor-kappaB (NF-kappaB) plays an important role in the pathogenesis of asthma. In the present study, the effects of montelukast on the level of IL-10 and on the activation of NF-kappaB in the inflammatory airway of asthmatic guinea pigs were investigated. Guinea pigs were sensitized by ovalbumin. Pulmonary inflammation was observed by hematoxylin and eosin staining. The eosinophils in broncho-alveolar lavage fluid and blood were separated by density gradient centrifugation and counted under microscope. The level of IL-10 in broncho-alveolar lavage fluid was measured by enzyme-linked immunoadsorbent assay. Activation of NF-kappaB in lung tissues was inspected by immunohistochemistry. Montelukast at medium and high doses prevented the decrease of IL-10 in broncho-alveolar lavage fluid (n = 8, p < 0.01 vs. asthma model group), inhibited the activation of NF-kappaB in lung tissues (n = 8; medium dose, p < 0.05; high dose, p < 0.01; vs. asthma model group). There was a significantly negative correlation between the level of IL-10 and the activation of NF-kappaB in lung tissues (r = -0.488, p < 0.01). Montelukast reduced the severity of airway inflammation and the number of eosinophils in asthmatic guinea pigs. From all these findings we conclude that montelukast can prevent the decrease of IL-10 and inhibit the activation of NF-kappaB in inflammatory airway of asthmatic guinea pigs, which may be the new important mechanisms of montelukast's anti-airway-inflammation effects in asthmatic guinea pigs.
Collapse
Affiliation(s)
- Yuqing Wu
- Department of Pharmacology, Nanjing Medical University, 210029, Hanzhong Road 140, Nanjing, China
| | | | | | | |
Collapse
|
43
|
Dahlén SE. Treatment of asthma with antileukotrienes: first line or last resort therapy? Eur J Pharmacol 2006; 533:40-56. [PMID: 16510137 DOI: 10.1016/j.ejphar.2005.12.070] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/13/2005] [Indexed: 02/04/2023]
Abstract
Twenty five years after the structure elucidation of slow reacting substance of anaphylaxis, antileukotrienes are established as a new therapeutic modality in asthma. The chapter reviews the biochemistry and pharmacology of leukotrienes and antileukotrienes with particular focus on the different usage of antileukotrienes for treatment of asthma and rhinitis in Europe and the US. Further research needs and new areas for leukotriene involvement in respiratory diseases are also discussed.
Collapse
Affiliation(s)
- Sven-Erik Dahlén
- Experimental Asthma and Allergy Research, The National Institute of Environmental Medicine, Karolinska Institute, SE-171 77 Stockholm, Sweden.
| |
Collapse
|
44
|
McKew JC, Foley MA, Thakker P, Behnke ML, Lovering FE, Sum FW, Tam S, Wu K, Shen MWH, Zhang W, Gonzalez M, Liu S, Mahadevan A, Sard H, Khor SP, Clark JD. Inhibition of cytosolic phospholipase A2alpha: hit to lead optimization. J Med Chem 2006; 49:135-58. [PMID: 16392799 DOI: 10.1021/jm0507882] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Compound 1 was previously reported to be a potent inhibitor of cPLA(2)alpha in both artificial monomeric substrate and cell-based assays. However, 1 was inactive in whole blood assays previously used to characterize cyclooxygenase and lipoxygenase inhibitors. The IC(50) of 1 increased dramatically with cell number or lipid/detergent concentration. In an attempt to insert an electrophilic ketone between the indole and benzoic acid moieties, we discovered that increasing the distance between the two moieties gave a compound with activity in the GLU (7-hydroxycoumarinyl-gamma-linolenate) micelle assay, which contains lipid and detergent. Extensive structure-activity relationship work around this lead identified a potent pharmacophore for cPLA(2)alpha inhibition. The IC(50)s between the GLU micelle and rat whole blood assays correlated highly. No correlation was found for other parameters, including lipophilicity or acidity of the required acid functionality. Compounds 25, 39, and 94 emerged as potent, selective inhibitors of cPLA(2)alpha and represent well-validated starting points for further optimization.
Collapse
Affiliation(s)
- John C McKew
- Department of Chemical and Screening Sciences, Wyeth Research, 200 CambridgePark Drive, Cambridge, Massachusetts 02140, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Thompson MD, Burnham WM, Cole DEC. The G protein-coupled receptors: pharmacogenetics and disease. Crit Rev Clin Lab Sci 2005; 42:311-92. [PMID: 16281738 DOI: 10.1080/10408360591001895] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Genetic variation in G-protein coupled receptors (GPCRs) is associated with a wide spectrum of disease phenotypes and predispositions that are of special significance because they are the targets of therapeutic agents. Each variant provides an opportunity to understand receptor function that complements a plethora of available in vitro data elucidating the pharmacology of the GPCRs. For example, discrete portions of the proximal tail of the dopamine D1 receptor have been discovered, in vitro, that may be involved in desensitization, recycling and trafficking. Similar in vitro strategies have been used to elucidate naturally occurring GPCR mutations. Inactive, over-active or constitutively active receptors have been identified by changes in ligand binding, G-protein coupling, receptor desensitization and receptor recycling. Selected examples reviewed include those disorders resulting from mutations in rhodopsin, thyrotropin, luteinizing hormone, vasopressin and angiotensin receptors. By comparison, the recurrent pharmacogenetic variants are more likely to result in an altered predisposition to complex disease in the population. These common variants may affect receptor sequence without intrinsic phenotype change or spontaneous induction of disease and yet result in significant alteration in drug efficacy. These pharmacogenetic phenomena will be reviewed with respect to a limited sampling of GPCR systems including the orexin/hypocretin system, the beta2 adrenergic receptors, the cysteinyl leukotriene receptors and the calcium-sensing receptor. These developments will be discussed with respect to strategies for drug discovery that take into account the potential for the development of drugs targeted at mutated and wild-type proteins.
Collapse
Affiliation(s)
- Miles D Thompson
- Department of Laboratory Medicine and Pathobiology, Banting Institute, University of Toronto, ON, Canada.
| | | | | |
Collapse
|
46
|
Yu GL, Wei EQ, Zhang SH, Xu HM, Chu LS, Zhang WP, Zhang Q, Chen Z, Mei RH, Zhao MH. Montelukast, a Cysteinyl Leukotriene Receptor-1 Antagonist, Dose- and Time-Dependently Protects against Focal Cerebral Ischemia in Mice. Pharmacology 2004; 73:31-40. [PMID: 15452361 DOI: 10.1159/000081072] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2004] [Accepted: 05/27/2004] [Indexed: 12/29/2022]
Abstract
Our previous studies showed that cysteinyl leukotriene receptor-1 (CysLT1) antagonist pranlukast has a neuroprotective effect on cerebral ischemia in rats and mice. However, whether the neuroprotective effect of pranlukast is its special action or a common action of CysLT1 receptor antagonists remains to be clarified. This study was performed to determine whether montelukast, another CysLT1 receptor antagonist, has the neuroprotective effect on focal cerebral ischemia in mice, and to observe its dose- and time-dependent properties. Permanent focal cerebral ischemia was induced by middle cerebral artery occlusion (MCAO). Montelukast was injected intraperitoneally either as multiple doses (once a day for 3 days and 30 min before MCAO) or as a single dose (at 30 min before, 30 min after, or 1 h after MCAO), respectively, and pranlukast and edaravone were used as controls. The neurological deficits, infarct volumes, brain edema, neuron density, and Evans blue extravasation in the brain were determined 24 h after MCAO. Pretreatments with multiple doses or a single dose of montelukast (0.1 and 1.0 mg/kg) before MCAO significantly attenuated all the ischemic insults. Post-treatment with a single dose of montelukast (0.1 and 1.0 mg/kg) at 30 min after MCAO also significantly decreased brain edema and infarct volume, but not neurological deficits. However, post-treatment with a single dose of montelukast at 1 h after MCAO had no significant effect. Pranlukast showed the same effects as montelukast, but edaravone attenuated the ischemic insults only with multiple doses before MCAO. Thus, montelukast has a dose- and time-dependent neuroprotective effect on permanent focal cerebral ischemia in mice, with an effective dose range of 0.1-1.0 mg/kg and a therapeutic window of 30 min. These findings further support the therapeutic potential of CysLT1 receptor antagonists in the treatment of cerebral ischemia at earlier phases.
Collapse
Affiliation(s)
- Guo-Liang Yu
- Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Ferrari F, Mennuni L, Caselli G, Zanelli T, Makovec F. Pharmacological profile of CR3465, a new leukotriene CysLT1 receptor antagonist with broad anti-inflammatory activity. Eur J Pharmacol 2004; 504:223-33. [PMID: 15541426 DOI: 10.1016/j.ejphar.2004.10.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2004] [Revised: 09/30/2004] [Accepted: 10/06/2004] [Indexed: 11/19/2022]
Abstract
CR3465 (L-Tyrosine, N-[(2-quinolinyl)carbonyl]-O-(7-fluoro-2-quinolinylmethyl) sodium salt) is a potent antagonist of [3H]leukotriene D4 ([3H]LTD4) binding to guinea pig lung preparations, its Ki (4.7+/-0.7 nM) being comparable with that of montelukast (5.6+/-0.6 nM). In tracheal strips from standard or ovalbumin-sensitized guinea pigs, CR3465 caused parallel rightward shifts in the concentration-response curves obtained with either LTD4 or antigen (pA(2), 8.74 and 8.15). Intravenous (i.v.) administration of the agent both antagonized (ED50, 9.9+/-1.9 microg/kg) and reverted LTD4 -induced bronchoconstriction of anesthetized guinea pigs. CR3465 reduced inflammatory infiltrates in the bronchoalveolar lavage fluid after antigen challenge of sensitized animals, and proved also active in inhibiting phosphodiesterase 3 (PDE3) and phosphodiesterase 4 (PDE4) activities exhibited by human platelets and neutrophils (IC50, 2.01+/-0.07 and 4.7+/-0.5 microM). In line with properties shown by phosphodiesterase inhibitors, CR3465 reduced the contractile response of guinea pig airways to histamine and decreased N-formyl-Met-Leu-Phe (fMLP)-induced degranulation of human neutrophils (IC50, 13.8 microM). Oral administration (20 mg/kg) of the compound in rats produced a significant (37%) ex vivo inhibition of tumor necrosis factor-alpha (TNF-alpha) release from lipopolysaccharide-stimulated whole blood. Pharmacokinetic data in the rat demonstrated approximately 100% bioavailability of the agent. We conclude that CR3465 represents a potent leukotriene CysLT1 receptor antagonist with enhanced effects, being also useful for counteracting spasmogenic and inflammatory stimuli other than those elicited by cysteinyl-leukotrienes (Cys-LTs).
Collapse
Affiliation(s)
- Flora Ferrari
- Rotta Research Laboratorium S.p.A., Pharmacology and Toxicology Department, Via Valosa di Sopra 7/9-20052 Monza (MI), Italy.
| | | | | | | | | |
Collapse
|
48
|
Bjermer L, Diamant Z. Current and emerging nonsteroidal anti-inflammatory therapies targeting specific mechanisms in asthma and allergy. ACTA ACUST UNITED AC 2004; 3:235-46. [PMID: 15350162 DOI: 10.2165/00151829-200403040-00004] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Today inhaled corticosteroids (ICS) are regarded as the first-line controller anti-inflammatory treatment in the management of asthma. However, there is an increasing awareness of the risk of long-term adverse effects of ICS and that asthma is not only an organ-specific disease but also a systemic and small airway disease. This thinking has called for systemic treatment alternatives to treat asthma targeting more disease-specific mechanisms without influencing normal physiologic functions. Blocking of disease-specific mediators is a mechanism utilized by anti-leukotrienes and anti-immunoglobulin E treatment, each proven to be effective in both asthma and allergic rhinitis.Different cytokine-modifying strategies have been tested in clinical trials with variable results, some disappointing and some encouraging. Anti-interleukin (IL)-5 monoclonal antibody treatment effectively reduces the number of eosinophils locally in the airways and in peripheral blood in asthmatic patients. Unfortunately, this marked effect on eosinophils was not associated with an improvement in bronchial hyperresponsiveness and/or symptoms. Clinical trials with a recombinant soluble IL-4 receptor have been somewhat more successful at improving asthma control and allowing reduction of ICS therapy in asthma. Treatment with recombinant IL-12 had an effect on bronchial hyperresponsiveness and eosinophilic response, but was associated with unacceptable adverse effects. Other interesting cytokine-modulating treatments include those targeting IL-9, IL-10, IL-12 and IL-13.Immune-modulating treatment with bacterial antigens represents another strategy, originating from the hypothesis that some bacterial infections guide the immune system towards a T helper (Th) type 1 immune response. Mycobacterium vaccae, Bacille Calmette-Guerin (BCG) and immunostimulatory DNA sequences have all been tested in clinical trials, with encouraging results. Future asthma and allergy treatment will probably include not only one but also two or more disease-modifying agents administered to the same patient.
Collapse
Affiliation(s)
- Leif Bjermer
- Department of Respiratory Medicine & Allergology, University Hospital, Lund, Sweden.
| | | |
Collapse
|
49
|
Abstract
Vernal keratoconjunctivitis (VKC) is an allergic eye disease that especially affects young boys. The most common symptoms are itching, photophobia, burning, and tearing. The most common signs are giant papillae, superficial keratitis, and conjunctival hyperaemia. Patients with VKC frequently have a family or medical history of atopic diseases, such as asthma, rhinitis, and eczema. However, VKC is not associated with a positive skin test or RAST in 42-47% of patients, confirming that it is not solely an IgE-mediated disease. On the basis of challenge studies as well as immunohistochemical and mediator studies, a Th2-driven mechanism with the involvement of mast cells, eosinophils, and lymphocytes has been suggested. Th2 lymphocytes are responsible for both hyperproduction of IgE (interleukin 4, IL-4) and for differentiation and activation of mast cells (IL-3) and eosinophils (IL-5). Other studies have demonstrated the involvement of neural factors such as substance P and NGF in the pathogenesis of VKC, and the overexpression of oestrogen and progesterone receptors in the conjunctiva of VKC patients has introduced the possible involvement of sex hormones. Thus, the pathogenesis of VKC is probably multifactorial, with the interaction of the immune, nervous, and endocrine systems. The clinical management of VKC requires a swift diagnosis, correct therapy, and evaluation of the prognosis. The diagnosis is generally based on the signs and symptoms of the disease, but in difficult cases can be aided by conjunctival scraping, demonstrating the presence of infiltrating eosinophils. Therapeutic options are many, in most cases topical, and should be chosen on the basis of the severity of the disease. The most effective drugs, steroids, should however be carefully administered, and only for brief periods, to avoid secondary development of glaucoma.A 2% solution of cyclosporine in olive oil or in castor oil should be considered as an alternative. The long-term prognosis of patients is generally good; however 6% of patients develop corneal damage, cataract, or glaucoma.
Collapse
Affiliation(s)
- S Bonini
- Interdisciplinary Center for Biomedical Research (CIR), Laboratory of Ophthalmology, University of Rome, Campus Bio-Medico, Rome, Italy.
| | | | | | | |
Collapse
|
50
|
Minciullo PL, Saija A, Bonanno D, Ferlazzo E, Gangemi S. Montelukast-induced generalized urticaria. Ann Pharmacother 2004; 38:999-1001. [PMID: 15113985 DOI: 10.1345/aph.1d547] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
OBJECTIVE To report a case of generalized urticaria induced by montelukast treatment. CASE SUMMARY A 28-year-old man with allergic rhinitis and moderate persistent asthma developed generalized urticaria 5 days after the initiation of montelukast and inhaled fluticasone. Symptoms disappeared within one day after suspension of both drugs. Two months later, after the resumption of montelukast and fluticasone, the patient developed generalized urticaria and eyelid angioedema, which were successfully treated with intravenous betamethasone, achieving complete remission within hours. After 2 days, the patient resumed inhaled fluticasone only and continued this therapy for several months without any adverse reaction. DISCUSSION We attributed the adverse reaction to montelukast because of the temporal relationship between use of montelukast and urticaria, the absence of other identified causative factors and other explanations for allergic reactions, and the positive dechallenge and rechallenge. The Naranjo probability scale showed a probable relationship between skin manifestations and montelukast treatment. CONCLUSIONS The use of antileukotrienes is increasing in asthma therapy. In cases of generalized urticaria in asthmatic patients undergoing montelukast therapy, physicians should be aware of a potential adverse reaction to this drug.
Collapse
Affiliation(s)
- Paola L Minciullo
- Department of Human Pathology, Division and School of Allergy and Clinical Immunology, University of Messina, 98123 Messina, Italy
| | | | | | | | | |
Collapse
|