1
|
Agache I, Antolin‐Amerigo D, Blay F, Boccabella C, Caruso C, Chanez P, Couto M, Covar R, Doan S, Fauquert J, Gauvreau G, Gherasim A, Klimek L, Lemiere C, Nair P, Ojanguren I, Peden D, Perez‐de‐Llano L, Pfaar O, Rondon C, Rukhazde M, Sastre J, Schulze J, Silva D, Tarlo S, Toppila‐Salmi S, Walusiak‐Skorupa J, Zielen S, Eguiluz‐Gracia I. EAACI position paper on the clinical use of the bronchial allergen challenge: Unmet needs and research priorities. Allergy 2022; 77:1667-1684. [PMID: 34978085 DOI: 10.1111/all.15203] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 12/16/2021] [Accepted: 12/27/2021] [Indexed: 12/22/2022]
Abstract
Allergic asthma (AA) is a common asthma phenotype, and its diagnosis requires both the demonstration of IgE-sensitization to aeroallergens and the causative role of this sensitization as a major driver of asthma symptoms. Therefore, a bronchial allergen challenge (BAC) would be occasionally required to identify AA patients among atopic asthmatics. Nevertheless, BAC is usually considered a research tool only, with existing protocols being tailored to mild asthmatics and research needs (eg long washout period for inhaled corticosteroids). Consequently, existing BAC protocols are not designed to be performed in moderate-to-severe asthmatics or in clinical practice. The correct diagnosis of AA might help select patients for immunomodulatory therapies. Allergen sublingual immunotherapy is now registered and recommended for controlled or partially controlled patients with house dust mite-driven AA and with FEV1 ≥ 70%. Allergen avoidance is costly and difficult to implement for the management of AA, so the proper selection of patients is also beneficial. In this position paper, the EAACI Task Force proposes a methodology for clinical BAC that would need to be validated in future studies. The clinical implementation of BAC could ultimately translate into a better phenotyping of asthmatics in real life, and into a more accurate selection of patients for long-term and costly management pathways.
Collapse
Affiliation(s)
- Ioana Agache
- Faculty of Medicine Transylvania University Brasov Romania
| | - Dario Antolin‐Amerigo
- Servicio de Alergia Hospital Universitario Ramón y Cajal Instituto Ramón y Cajal de Investigación Sanitaria Madrid Spain
| | - Frederic Blay
- ALYATEC Environmental Exposure Chamber Chest Diseases Department Strasbourg University Hospital University of Strasbourg Strasbourg France
| | - Cristina Boccabella
- Department of Cardiovascular and Thoracic Sciences Università Cattolica del Sacro Cuore Fondazione Policlinico Universitario A. Gemelli ‐ IRCCS Rome Italy
| | | | - Pascal Chanez
- Department of Respiratory CIC Nord INSERMINRAE C2VN Aix Marseille University Marseille France
| | - Mariana Couto
- Centro de Alergia Hospital CUF Descobertas Lisboa Portugal
| | - Ronina Covar
- Pediatrics National Jewish Health Denver Colorado USA
| | | | | | - Gail Gauvreau
- Division of Respirology Department of Medicine McMaster University Hamilton Ontario Canada
| | - Alina Gherasim
- ALYATEC Environmental Exposure Chamber Strasbourg France
| | - Ludger Klimek
- Center for Rhinology and Allergology Wiesbaden Germany
| | - Catherine Lemiere
- Research Centre Centre Intégré Universitaire de santé et de services sociaux du Nord‐de‐l'île‐de‐Montréal Montréal Quebec Canada
- Faculty of Medicine Université de Montreal Montreal Quebec Canada
| | - Parameswaran Nair
- Department of Medicine Firestone Institute of Respiratory Health at St. Joseph's Healthcare McMaster University Hamilton Ontario Canada
| | - Iñigo Ojanguren
- Departament de Medicina Servei de Pneumología Hospital Universitari Valld´Hebron Universitat Autònoma de Barcelona (UAB) Institut de Recerca (VHIR) CIBER de Enfermedades Respiratorias (CIBERES) Barcelona Spain
| | - David Peden
- Division of Pediatric Allergy and Immunology Center for Environmental Medicine, Asthma and Lung Biology The School of Medicine The University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Luis Perez‐de‐Llano
- Department of Respiratory Medicine University Hospital Lucus Augusti Lugo Spain
| | - Oliver Pfaar
- Section of Rhinology and Allergy Department of Otorhinolaryngology, Head and Neck Surgery University Hospital Marburg Philipps‐Universität Marburg Marburg Germany
| | - Carmen Rondon
- Allergy Unit Hospital Regional Universitario de Malaga Instituto de Investigacion Biomedica de Malaga (IBIMA) Malaga Spain
| | - Maia Rukhazde
- Center of Allergy & Immunology Teaching University Geomedi LLC Tbilisi Georgia
| | - Joaquin Sastre
- Allergy Unit Hospital Universitario Fundación Jiménez Díaz Center for Biomedical Network of Respiratory Diseases (CIBERES) Instituto de Salud Carlos III (ISCIII) Madrid Spain
| | - Johannes Schulze
- Department for Children and Adolescents, Division of Allergology Pulmonology and Cystic Fibrosis Goethe‐University Hospital Frankfurt am Main Germany
| | - Diana Silva
- Basic and Clinical Immunology Unit Department of Pathology Faculty of Medicine University of Porto and Serviço de Imunoalergologia Centro Hospitalar São João, EPE Porto Portugal
| | - Susan Tarlo
- Respiratory Division Department of Medicine University Health Network, Toronto Western Hospital University of Toronto Department of Medicine, and Dalla Lana Department of Public Health Toronto Ontario Canada
| | - Sanna Toppila‐Salmi
- Haartman Institute, Medicum, Skin and Allergy Hospital Hospital District of Helsinki and Uusimaa Helsinki University Hospital and University of Helsinki Helsinki Finland
| | - Jolanta Walusiak‐Skorupa
- Department of Occupational Diseases and Environmental Health Nofer Institute of Occupational Medicine Łódź Poland
| | - Stefan Zielen
- Department for Children and Adolescents, Division of Allergology Pulmonology and Cystic Fibrosis Goethe‐University Hospital Frankfurt am Main Germany
| | - Ibon Eguiluz‐Gracia
- Allergy Unit Hospital Regional Universitario de Malaga Instituto de Investigacion Biomedica de Malaga (IBIMA) Malaga Spain
| |
Collapse
|
2
|
Spyridaki I, Taka S, Skevaki C, Trochoutsou A, Papadopoulos NG. In Vitro Effects of 5-Lipoxygenase Pathway Inhibition on Rhinovirus-Associated Bronchial Epithelial Inflammation. Pulm Ther 2021; 7:237-249. [PMID: 33847974 PMCID: PMC8137792 DOI: 10.1007/s41030-021-00152-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 03/02/2021] [Indexed: 11/29/2022] Open
Abstract
Introduction The leukotriene pathway may be implicated in the induction of virus-induced inflammation. Respiratory epithelial cells may express low levels of 5-lipoxygenase (5-LO) and release leukotrienes (LTs) C4, D4, and E4, upon exposure to viruses or other stimuli. Enhanced expression of 5-LO pathway proteins after rhinovirus (RV) infection has previously been described. We hypothesized that anti-leukotriene treatment of epithelial cells, with or without exposure to RV-infected peripheral blood mononuclear cells (PBMCs)-conditioned media, may inhibit RV-induced up-regulation of inflammatory cytokines. Methods PBMCs from a healthy donor were exposed to RV1B and supernatants were harvested at 48 h post infection. BEAS-2B cells were infected with RV, with or without conditioning with the PBMC supernatant. Treatment with anti-LT agents was performed either on both PBMCs and BEAS-2B or at the bronchial epithelial level only, with varying concentrations of montelukast (CysLT receptor antagonist) or MK-886 [FLAP(5-lipoxygenase-activating-protein) inhibitor]. Evaluation of the inflammatory cytokines IL-8, RANTES, IL-11, IL-6, and IP-10 was performed using ELISA. Results Our results show that anti-LT treatment of RV-infected bronchial epithelial cells suppresses epithelial RV-mediated cytokine production, independent of conditioning. Conclusions This observation may represent an indirect mode of action of the anti-leukotrienes in virus-induced asthma. Supplementary Information The online version contains supplementary material available at 10.1007/s41030-021-00152-x.
Collapse
Affiliation(s)
- Irini Spyridaki
- Allergy Department, 2nd Pediatric Clinic, National and Kapodistrian University of Athens, Athens, Greece
| | - Styliani Taka
- Allergy Department, 2nd Pediatric Clinic, National and Kapodistrian University of Athens, Athens, Greece.
| | - Chrysanthi Skevaki
- Allergy Department, 2nd Pediatric Clinic, National and Kapodistrian University of Athens, Athens, Greece.,Institute of Laboratory Medicine, Philipps-Universität Marburg, Marburg, Germany.,Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Philipps-Universität Marburg, Hannover, Germany.,German Center for Lung Research (DZL), Hannover, Germany
| | - Aikaterini Trochoutsou
- Allergy Department, 2nd Pediatric Clinic, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikolaos G Papadopoulos
- Allergy Department, 2nd Pediatric Clinic, National and Kapodistrian University of Athens, Athens, Greece.,Division of Infection, Inflammation and Respiratory Medicine, University of Manchester, Manchester, UK
| |
Collapse
|
3
|
Boulet LP, Côté A, Abd-Elaziz K, Gauvreau G, Diamant Z. Allergen bronchoprovocation test: an important research tool supporting precision medicine. Curr Opin Pulm Med 2021; 27:15-22. [PMID: 33065599 DOI: 10.1097/mcp.0000000000000742] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE OF REVIEW Allergen bronchoprovocation test (ABT) has been used to study asthma pathophysiology and as a disease-modelling tool to assess the properties and efficacy of new asthma drugs. In view of the complexity and heterogeneity of asthma, which has driven the definition of several phenotypes and endotypes, we aim to discuss the role of ABT in the era of precision medicine and provide guidance for clinicians how to interpret and use available data to understand the implications for the benefits of asthma treatment. RECENT FINDINGS In this review, we summarize background knowledge and applications of ABT and provide an update with recent publications on this topic. In the past years, several studies have been published on ABT in combination with non-invasive and invasive airway samplings and innovative detection techniques allowing to study several inflammatory mechanisms linked to Th2-pathway and allergen-induced pathophysiology throughout the airways. SUMMARY ABT is a valuable research tool, which has strongly contributed to precision medicine by helping to define allergen-triggered key inflammatory pathways and airway pathophysiology, and thus helped to shape our understanding of allergen-driven asthma phenotypes and endotypes. In addition, ABT has been instrumental to assess the interactions and effects of new-targeted asthma treatments along these pathways.
Collapse
Affiliation(s)
- Louis-Philippe Boulet
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec Heart and Lung Institute, Université Laval, Québec, Canada
| | - Andréanne Côté
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec Heart and Lung Institute, Université Laval, Québec, Canada
| | | | - Gail Gauvreau
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Zuzana Diamant
- Department of Respiratory Medicine and Allergology, Institute for Clinical Science, Skane University Hospital, Lund University, Lund, Sweden
- Department of Respiratory Medicine, First Faculty of Medicine, Charles University and Thomayer Hospital, Prague, Czech Republic
- Department Clinical Pharmacy and Pharmacology, University Groningen, University Medicine Ctr Groningen, Groningen, The Netherlands
| |
Collapse
|
4
|
Sun CP, Zhang XY, Morisseau C, Hwang SH, Zhang ZJ, Hammock BD, Ma XC. Discovery of Soluble Epoxide Hydrolase Inhibitors from Chemical Synthesis and Natural Products. J Med Chem 2020; 64:184-215. [PMID: 33369424 DOI: 10.1021/acs.jmedchem.0c01507] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Soluble epoxide hydrolase (sEH) is an α/β hydrolase fold protein and widely distributed in numerous organs including the liver, kidney, and brain. The inhibition of sEH can effectively maintain endogenous epoxyeicosatrienoic acids (EETs) levels and reduce dihydroxyeicosatrienoic acids (DHETs) levels, resulting in therapeutic potentials for cardiovascular, central nervous system, and metabolic diseases. Therefore, since the beginning of this century, the development of sEH inhibitors is a hot research topic. A variety of potent sEH inhibitors have been developed by chemical synthesis or isolated from natural sources. In this review, we mainly summarized the interconnected aspects of sEH with cardiovascular, central nervous system, and metabolic diseases and then focus on representative inhibitors, which would provide some useful guidance for the future development of potential sEH inhibitors.
Collapse
Affiliation(s)
- Cheng-Peng Sun
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College (Institute) of Integrative Medicine, College of Pharmacy, Dalian Medical University, Dalian 116044, People's Republic of China
| | - Xin-Yue Zhang
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College (Institute) of Integrative Medicine, College of Pharmacy, Dalian Medical University, Dalian 116044, People's Republic of China
| | - Christophe Morisseau
- Department of Entomology and Nematology, UC Davis Comprehensive Cancer Center, University of California, Davis, California 95616, United States
| | - Sung Hee Hwang
- Department of Entomology and Nematology, UC Davis Comprehensive Cancer Center, University of California, Davis, California 95616, United States
| | - Zhan-Jun Zhang
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, People's Republic of China
| | - Bruce D Hammock
- Department of Entomology and Nematology, UC Davis Comprehensive Cancer Center, University of California, Davis, California 95616, United States
| | - Xiao-Chi Ma
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College (Institute) of Integrative Medicine, College of Pharmacy, Dalian Medical University, Dalian 116044, People's Republic of China.,College of Pharmacy, School of Medicine, Hangzhou Normal University, Hangzhou 311121, People's Republic of China
| |
Collapse
|
5
|
De Volder J, Vereecke L, Joos G, Maes T. Targeting neutrophils in asthma: A therapeutic opportunity? Biochem Pharmacol 2020; 182:114292. [PMID: 33080186 DOI: 10.1016/j.bcp.2020.114292] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 10/14/2020] [Accepted: 10/15/2020] [Indexed: 02/06/2023]
Abstract
Suppression of airway inflammation with inhaled corticosteroids has been the key therapeutic approach for asthma for many years. Identification of inflammatory phenotypes in asthma has moreover led to important breakthroughs, e.g. with specific targeting of the IL-5 pathway as add-on treatment in difficult-to-treat eosinophilic asthma. However, the impact of interfering with the neutrophilic component in asthma is less documented and understood. This review provides an overview of established and recent insights with regard to the role of neutrophils in asthma, focusing on research in humans. We will describe the main drivers of neutrophilic responses in asthma, the heterogeneity in neutrophils and how they could contribute to asthma pathogenesis. Moreover we will describe findings from clinical trials, in which neutrophilic inflammation was targeted. It is clear that neutrophils are important actors in asthma development and play a role in exacerbations. However, more research is required to fully understand how modulation of neutrophil activity could lead to a significant benefit in asthma patients with airway neutrophilia.
Collapse
Affiliation(s)
- Joyceline De Volder
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Lars Vereecke
- VIB Inflammation Research Center, Ghent, Belgium; Ghent Gut Inflammation Group (GGIG), Ghent University, Belgium; Department of Rheumatology, Ghent University Hospital, Belgium
| | - Guy Joos
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Tania Maes
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium.
| |
Collapse
|
6
|
Current state and future prospect of the therapeutic strategy targeting cysteinyl leukotriene metabolism in asthma. Respir Investig 2019; 57:534-543. [PMID: 31591069 DOI: 10.1016/j.resinv.2019.08.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 08/12/2019] [Accepted: 08/29/2019] [Indexed: 12/18/2022]
Abstract
Asthma is an allergic disorder with dominant type 2 airway inflammation, and its prevalence is increasing worldwide. Inhalation of corticosteroids is the primary treatment for asthma along with add-on drugs, including long-acting β2 agonists and/or cysteinyl leukotriene (cys-LT) receptor antagonists, in patients with poorly controlled asthma. Cys-LTs are composed of leukotriene C4 (LTC4), LTD4, and LTE4, which are enzymatically metabolized from arachidonic acid. These molecules act as inflammatory mediators through different types of high-affinity receptors, namely, CysLT1, CysLT2, and CysLT3 (also named as GPR99). CysLT1 antagonists possessing anti-inflammatory and bronchodilatory effects can be orally administered to patients with asthma. Recently, molecular biology-based studies have revealed the mechanism of inflammatory responses via other receptors, such as CysLT2 and CysLT3, as well as the importance of upstream inflammatory regulators, including type 2 cytokines (e.g., interleukins 4 and 5), in controlling cys-LT metabolism. These findings indicate the therapeutic potential of pharmacological agents targeting cys-LT metabolism-related receptors and enzymes, and antibody drugs neutralizing or antagonizing type 2 cytokines. This review focuses on the current state and future prospect of the therapeutic strategy targeting cys-LT metabolism.
Collapse
|
7
|
Nunns GR, Stringham JR, Gamboni F, Moore EE, Fragoso M, Stettler GR, Silliman CC, Banerjee A. Trauma and hemorrhagic shock activate molecular association of 5-lipoxygenase and 5-lipoxygenase-Activating protein in lung tissue. J Surg Res 2018; 229:262-270. [PMID: 29936999 DOI: 10.1016/j.jss.2018.03.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 02/02/2018] [Accepted: 03/14/2018] [Indexed: 12/20/2022]
Abstract
BACKGROUND Post-traumatic lung injury following trauma and hemorrhagic shock (T/HS) is associated with significant morbidity. Leukotriene-induced inflammation has been implicated in the development of post-traumatic lung injury through a mechanism that is only partially understood. Postshock mesenteric lymph returning to the systemic circulation is rich in arachidonic acid, the substrate of 5-lipoxygenase (ALOX5). ALOX5 is the rate-limiting enzyme in leukotriene synthesis and, following T/HS, contributes to the development of lung dysfunction. ALOX5 colocalizes with its cofactor, 5-lipoxygenase-activating protein (ALOX5AP), which is thought to potentiate ALOX5 synthetic activity. We hypothesized that T/HS results in the molecular association and nuclear colocalization of ALOX5 and ALOX5AP, which ultimately increases leukotriene production and potentiates lung injury. MATERIALS AND METHODS To examine these molecular interactions, a rat T/HS model was used. Post-T/HS tissue was evaluated for lung injury through both histologic analysis of lung sections and biochemical analysis of bronchoalveolar lavage fluid. Lung tissue was immunostained for ALOX5 and ALOX5AP with association and colocalization evaluated by fluorescence resonance energy transfer. In addition, rats undergoing T/HS were treated with MK-886, a known ALOX5AP inhibitor. RESULTS ALOX5 levels increase and ALOX5/ALOX5AP association occurred after T/HS, as evidenced by increases in total tissue fluorescence and fluorescence resonance energy transfer signal intensity, respectively. These findings coincided with increased leukotriene production and with the histological changes characteristic of lung injury. ALOX5/ALOX5AP complex formation, leukotriene production, and lung injury were decreased after inhibition of ALOX5AP with MK-886. CONCLUSIONS These results suggest that the association of ALOX5/ALOX5AP contributes to leukotriene-induced inflammation and predisposes the T/HS animal to lung injury.
Collapse
Affiliation(s)
- Geoffrey R Nunns
- School of Medicine, Department of Surgery, Trauma Research Center, University of Colorado Denver, Aurora, Colorado.
| | - John R Stringham
- School of Medicine, Department of Surgery, Trauma Research Center, University of Colorado Denver, Aurora, Colorado
| | - Fabia Gamboni
- School of Medicine, Department of Surgery, Trauma Research Center, University of Colorado Denver, Aurora, Colorado
| | - Ernest E Moore
- School of Medicine, Department of Surgery, Trauma Research Center, University of Colorado Denver, Aurora, Colorado; Denver Health Medical Center, Department of Surgery, Denver, Colorado
| | - Miguel Fragoso
- School of Medicine, Department of Surgery, Trauma Research Center, University of Colorado Denver, Aurora, Colorado; Denver Health Medical Center, Department of Surgery, Denver, Colorado
| | - Gregory R Stettler
- School of Medicine, Department of Surgery, Trauma Research Center, University of Colorado Denver, Aurora, Colorado
| | - Christopher C Silliman
- School of Medicine, Department of Surgery, Trauma Research Center, University of Colorado Denver, Aurora, Colorado; School of Medicine, Department of Pediatrics-Hematology/Oncology, Children's Hospital Colorado, University of Colorado Denver, Aurora, Colorado; Research Laboratory, Bonfils Blood Center, Denver, Colorado
| | - Anirban Banerjee
- School of Medicine, Department of Surgery, Trauma Research Center, University of Colorado Denver, Aurora, Colorado
| |
Collapse
|
8
|
Singh D. Evaluation of New Drugs for Asthma and COPD: Endpoints, Biomarkers and Clinical Trial Design. Handb Exp Pharmacol 2017; 237:243-264. [PMID: 27838852 DOI: 10.1007/164_2016_70] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
There remains a considerable need to develop novel therapies for patients with asthma and chronic obstructive pulmonary disease (COPD). The greatest challenge at the moment is measuring the effects of novel anti-inflammatory drugs, as these drugs often cause only small effects on lung function. Measurements that demonstrate the pharmacological and clinical effects of these drugs are needed. Furthermore, we now recognise that only subgroups of patients are likely to respond to these novel drugs, so using biomarkers to determine the clinical phenotype most suitable for such therapies is important. An endotype is a subtype of a (clinical) condition defined by a distinct pathophysiological mechanism. An endotype-driven approach may be more helpful in drug development, enabling drugs to be targeted specifically towards specific biological mechanisms rather than clinical characteristics. This requires the development of biomarkers to define endotypes and/or to measure drug effects. This newer approach should continue alongside efforts to optimise the measurement of clinical endpoints, including patient-reported outcome measurements, required by drug regulatory authorities.
Collapse
Affiliation(s)
- Dave Singh
- Medicines Evaluation Unit, University of Manchester, University Hospital of South Manchester Foundations Trust, Langley Building, Southmoor Road, Wythenshawe, Manchester, M23 9Q2, UK.
| |
Collapse
|
9
|
Kowal-Bielecka O, Chwiesko-Minarowska S, Bernatowicz PL, Allanore Y, Radstake T, Matucci-Cerinic M, Broen J, Hesselstrand R, Krasowska D, Riemekasten G, Vonk M, Kowalczuk O, Bielecki M, Milewski R, Chyczewski L, Niklinski J, Kowal K. The arachidonate 5-lipoxygenase activating protein gene polymorphism is associated with the risk of scleroderma-related interstitial lung disease: a multicentre European Scleroderma Trials and Research group (EUSTAR) study. Rheumatology (Oxford) 2017; 56:844-852. [DOI: 10.1093/rheumatology/kew499] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Indexed: 01/02/2023] Open
|
10
|
Discovery of the Novel Oxadiazole-Containing 5-Lipoxygenase Activating Protein (FLAP) Inhibitor BI 665915. ACTA ACUST UNITED AC 2016. [DOI: 10.1021/bk-2016-1239.ch004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
|
11
|
Singh D, Leaker B, Boyce M, Nandeuil MA, Collarini S, Mariotti F, Santoro D, Barnes PJ. A novel inhaled phosphodiesterase 4 inhibitor (CHF6001) reduces the allergen challenge response in asthmatic patients. Pulm Pharmacol Ther 2016; 40:1-6. [PMID: 27373438 DOI: 10.1016/j.pupt.2016.06.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Revised: 06/08/2016] [Accepted: 06/28/2016] [Indexed: 12/18/2022]
Abstract
CHF6001 is an inhaled phosphodiesterase 4 (PDE4) inhibitor in development for the treatment of obstructive lung diseases. The efficacy and safety of CHF6001 were investigated in a double blind, placebo controlled, 3-way cross-over study using the allergen challenge model. Thirty-six atopic asthmatics who were not taking inhaled corticosteroids and who demonstrated a late asthmatic response (LAR) to inhaled allergen at screening were randomised to receive CHF6001 400 μg or 1200 μg or placebo administered once a day using a dry powder inhaler. The three treatment periods were 9 days; allergen challenges were performed on day 9 and induced sputum was obtained after 10 h from challenge. Washout periods between treatments were up to 5 weeks. Both CHF6001 doses significantly attenuated the LAR; the primary endpoint analysis showed that CHF6001 400 μg and 1200 μg caused reductions of 19.7% (p = 0.015) and 28.2% (p < 0.001) respectively of the weighted FEV1 AUC4-10h compared with placebo. The difference between the CHF6001 doses was not statistically significant (p = 0.223). Compared with placebo, CHF6001 caused greater reduction in sputum eosinophil counts, although these changes were not statistically significant. CHF6001 was well tolerated, with similar numbers of adverse events in each treatment period. This inhaled PDE4 inhibitor has the potential to provide clinical benefits in patients with atopic asthma.
Collapse
Affiliation(s)
- D Singh
- University of Manchester, Medicines Evaluation Unit, University Hospital of South Manchester Foundation Trust, Southmoor Road, Manchester, M23 9QZ, United Kingdom.
| | - B Leaker
- Respiratory Clinical Trials Ltd, 18-22 Queen Anne St, London, W1G 8HU, United Kingdom
| | - M Boyce
- Hammersmith Medicines Research, Cumberland Avenue, London, NW10 7EW, United Kingdom
| | - M A Nandeuil
- Chiesi S.A., 11 Avenue Dubonnet, 92400, Courbevoie, France
| | - S Collarini
- Chiesi Farmaceutici S.p.A., Via Palermo 26/A, 43122, Parma, Italy
| | - F Mariotti
- Chiesi Farmaceutici S.p.A., Via Palermo 26/A, 43122, Parma, Italy
| | - D Santoro
- Chiesi Farmaceutici S.p.A., Via Palermo 26/A, 43122, Parma, Italy
| | - P J Barnes
- National Heart & Lung Institute, Imperial College, London, SW3 6LY, United Kingdom
| |
Collapse
|
12
|
Arthur G, Bradding P. New Developments in Mast Cell Biology: Clinical Implications. Chest 2016; 150:680-93. [PMID: 27316557 DOI: 10.1016/j.chest.2016.06.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 04/26/2016] [Accepted: 06/03/2016] [Indexed: 12/12/2022] Open
Abstract
Mast cells (MCs) are present in connective tissue and at mucosal surfaces in all classes of vertebrates. In health, they contribute to tissue homeostasis, host defense, and tissue repair via multiple receptors regulating the release of a vast stockpile of proinflammatory mediators, proteases, and cytokines. However, these potentially protective cells are a double-edged sword. When there is a repeated or long-term stimulus, MC activation leads to tissue damage and dysfunction. Accordingly, MCs are implicated in the pathophysiologic aspects of numerous diseases covering all organs. Understanding the biology of MCs, their heterogeneity, mechanisms of activation, and signaling cascades may lead to the development of novel therapies for many diseases for which current treatments are lacking or are of poor efficacy. This review will focus on updates and developments in MC biology and their clinical implications, with a particular focus on their role in respiratory diseases.
Collapse
Affiliation(s)
- Greer Arthur
- Department of Infection, Immunity and Inflammation, Institute for Lung Health, University of Leicester, England
| | - Peter Bradding
- Department of Infection, Immunity and Inflammation, Institute for Lung Health, University of Leicester, England.
| |
Collapse
|
13
|
Abstract
Noneosinophilic airway inflammation occurs in approximately 50% of patients with asthma. It is subdivided into neutrophilic or paucigranulocytic inflammation, although the proportion of each subtype is uncertain because of variable cut-off points used to define neutrophilia. This article reviews the evidence for noneosinophilic inflammation being a target for therapy in asthma and assesses clinical trials of licensed drugs, novel small molecules and biologics agents in noneosinophilic inflammation. Current symptoms, rate of exacerbations and decline in lung function are generally less in noneosinophilic asthma than eosinophilic asthma. Noneosinophilic inflammation is associated with corticosteroid insensitivity. Neutrophil activation in the airways and systemic inflammation is reported in neutrophilic asthma. Neutrophilia in asthma may be due to corticosteroids, associated chronic pulmonary infection, altered airway microbiome or delayed neutrophil apoptosis. The cause of poorly controlled noneosinophilic asthma may differ between patients and involve several mechanism including neutrophilic inflammation, T helper 2 (Th2)-low or other subtypes of airway inflammation or corticosteroid insensitivity as well as noninflammatory pathways such as airway hyperreactivity and remodelling. Smoking cessation in asthmatic smokers and removal from exposure to some occupational agents reduces neutrophilic inflammation. Preliminary studies of 'off-label' use of licensed drugs suggest that macrolides show efficacy in nonsmokers with noneosinophilic severe asthma and statins, low-dose theophylline and peroxisome proliferator-activated receptor gamma (PPARγ) agonists may benefit asthmatic smokers with noneosinophilic inflammation. Novel small molecules targeting neutrophilic inflammation, such as chemokine (CXC) receptor 2 (CXCR2) antagonists reduce neutrophils, but do not improve clinical outcomes in studies to date. Inhaled phosphodiesterase (PDE)4 inhibitors, dual PDE3 and PDE4 inhibitors, p38MAPK (mitogen-activated protein kinase) inhibitors, tyrosine kinase inhibitors and PI (phosphoinositide) 3kinase inhibitors are under development and these compounds may be of benefit in noneosinophilic inflammation. The results of clinical trials of biological agents targeting mediators associated with noneosinophilic inflammation, such as interleukin (IL)-17 and tumor necrosis factor (TNF)-α are disappointing. Greater understanding of the mechanisms of noneosinophilic inflammation in asthma should lead to improved therapies.
Collapse
Affiliation(s)
- Neil C Thomson
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow G12 0YN, UK
| |
Collapse
|
14
|
Exploring the roles of UGT1A1 and UGT1A3 in oral clearance of GSK2190915, a 5-lipoxygenase-activating protein inhibitor. Pharmacogenet Genomics 2015; 24:618-21. [PMID: 25192553 DOI: 10.1097/fpc.0000000000000090] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Pharmacokinetic variability in drug exposure is a concern for all compounds in development including those for the treatment of asthma and other respiratory disorders. Substantial variability in the oral clearance of GSK2190915, a 5-lipoxygenase-activating protein inhibitor that attenuates the production of leukotriene B4 and cysteinyl leukotrienes, is largely unaccounted for by clinical variables. A study of 41 patients, 78% (32/41) of whom were non-Hispanic whites, with mild to moderate asthma identified an association of UGT1A1*28 and UGT1A3*2 with the oral clearance of GSK2190915 (P=3.8×10⁻⁴ and 1.2×10⁻⁵, respectively). However, in a subsequent replication study of 403 non-Hispanic white patients with asthma, we failed to observe a statistically significant association between oral clearance of GSK2190915 and either UGT1A1*28 or UGT1A3*2 (P>0.05). Therefore, genetic effects that could explain the systemic exposure level variability of GSK2190915 were not identified.
Collapse
|
15
|
Abstract
Environmental allergens are an important cause of asthma and can contribute to loss of asthma control and exacerbations. Allergen inhalation challenge has been a useful clinical model to examine the mechanisms of allergen-induced airway responses and inflammation. Allergen bronchoconstrictor responses are the early response, which reaches a maximum within 30 min and resolves by 1-3 h, and late responses, when bronchoconstriction recurs after 3-4 h and reaches a maximum over 6-12 h. Late responses are followed by an increase in airway hyperresponsiveness. These responses occur when IgE on mast cells is cross-linked by an allergen, causing degranulation and the release of histamine, neutral proteases and chemotactic factors, and the production of newly formed mediators, such as cysteinyl leukotrienes and prostaglandin D2. Allergen-induced airway inflammation consists of an increase in airway eosinophils, basophils and, less consistently, neutrophils. These responses are mediated by the trafficking and activation of myeloid dendritic cells into the airways, probably as a result of the release of epithelial cell-derived thymic stromal lymphopoietin, and the release of pro-inflammatory cytokines from type 2 helper T-cells. Allergen inhalation challenge has also been a widely used model to study potential new therapies for asthma and has an excellent negative predictive value for this purpose.
Collapse
Affiliation(s)
- Gail M Gauvreau
- Firestone Institute for Respiratory Health and the Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Amani I El-Gammal
- Firestone Institute for Respiratory Health and the Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Paul M O'Byrne
- Firestone Institute for Respiratory Health and the Department of Medicine, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
16
|
Blankestijn MA, Boyle RJ, Gore R, Hawrylowicz C, Jarvis D, Knulst AC, Wardlaw AJ. Developments in the field of allergy in 2013 through the eyes of Clinical and Experimental Allergy. Clin Exp Allergy 2015; 44:1436-57. [PMID: 25346287 DOI: 10.1111/cea.12442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
2013 was another exciting year for allergy in general and Clinical and Experimental Allergy in particular. In the field of asthma and rhinitis, there continued to be a focus on heterogeneity and phenotypes with increasing use of biostatistical techniques to determine clusters of similar populations. Obesity- and aspirin-associated disease are intriguing associations with asthma which were explored in a number of papers. We published a number of excellent papers on mechanisms of airway inflammation and how this relates to physiology, pathology, genetics and biomarkers in both human and experimental model systems. In terms of mechanisms, there is less on individual cell types in allergic disease at the moment, but the immunology of allergic disease continued to fascinate our authors. Another area that was popular both in the mechanisms and in the epidemiology sections was early life events and how these lead to allergic disease, with an increasing focus on the role of the microbiome and how this influences immune tolerance. In the clinical allergy section, oral immunotherapy for food allergy is clearly a major topic of interest at the moment as was in vitro testing to distinguish between sensitization and allergic disease. There was less on inhalant allergy this year, but a good representation from the drug allergy community including some interesting work on non-IgE-mediated mechanisms. In the allergen section, important new allergens continue to be discovered, but the major focus as in the last couple of years was on working out how component-resolved approaches can improve diagnosis and management of food and venom allergy.
Collapse
Affiliation(s)
- M A Blankestijn
- Department of Dermatology and Allergology, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
17
|
Krug N, Hohlfeld JM, Kirsten AM, Kornmann O, Beeh KM, Kappeler D, Korn S, Ignatenko S, Timmer W, Rogon C, Zeitvogel J, Zhang N, Bille J, Homburg U, Turowska A, Bachert C, Werfel T, Buhl R, Renz J, Garn H, Renz H. Allergen-induced asthmatic responses modified by a GATA3-specific DNAzyme. N Engl J Med 2015; 372:1987-95. [PMID: 25981191 DOI: 10.1056/nejmoa1411776] [Citation(s) in RCA: 236] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND The most prevalent phenotype of asthma is characterized by eosinophil-dominated inflammation that is driven by a type 2 helper T cell (Th2). Therapeutic targeting of GATA3, an important transcription factor of the Th2 pathway, may be beneficial. We evaluated the safety and efficacy of SB010, a novel DNA enzyme (DNAzyme) that is able to cleave and inactivate GATA3 messenger RNA (mRNA). METHODS We conducted a randomized, double-blind, placebo-controlled, multicenter clinical trial of SB010 involving patients who had allergic asthma with sputum eosinophilia and who also had biphasic early and late asthmatic responses after laboratory-based allergen provocation. A total of 40 patients could be evaluated; 21 were assigned to receive 10 mg of SB010, and 19 were assigned to receive placebo, with each study drug administered by means of inhalation once daily for 28 days. An allergen challenge was performed before and after the 28-day period. The primary end point was the late asthmatic response as quantified by the change in the area under the curve (AUC) for forced expiratory volume in 1 second (FEV1). RESULTS After 28 days, SB010 attenuated the mean late asthmatic response by 34%, as compared with the baseline response, according to the AUC for FEV1, whereas placebo was associated with a 1% increase in the AUC for FEV1 (P=0.02). The early asthmatic response with SB010 was attenuated by 11% as measured by the AUC for FEV1, whereas the early response with placebo was increased by 10% (P=0.03). Inhibition of the late asthmatic response by SB010 was associated with attenuation of allergen-induced sputum eosinophilia and with lower levels of tryptase in sputum and lower plasma levels of interleukin-5. Allergen-induced levels of fractional exhaled nitric oxide and airway hyperresponsiveness to methacholine were not affected by either SB010 or placebo. CONCLUSIONS Treatment with SB010 significantly attenuated both late and early asthmatic responses after allergen provocation in patients with allergic asthma. Biomarker analysis showed an attenuation of Th2-regulated inflammatory responses. (Funded by Sterna Biologicals and the German Federal Ministry of Education and Research; ClinicalTrials.gov number, NCT01743768.).
Collapse
Affiliation(s)
- Norbert Krug
- From the Fraunhofer Institute for Toxicology and Experimental Medicine (N.K., J.M.H.) and the Department of Dermatology and Allergy, Hannover Medical School (J.Z., T.W.), Hannover, Pulmonary Research Institute at Lung Clinic Grosshansdorf, Grosshansdorf (A.-M.K.), Institut für klinische Forschung Pneumologie, Clinical Research Center Respiratory Medicine, Frankfurt (O.K.), Insaf Respiratory Research Institute, Wiesbaden (K.M.B.), Inamed, Gauting (D.K., W.T.), Pulmonary Department, Medical Clinic, University Hospital Mainz, Mainz (S.K., R.B.), Charité Research Organization, Berlin (S.I.), FGK Clinical Research, Munich (C.R.), Sterna Biologicals (J.B., U.H., A.T., J.R.) and Institute of Laboratory Medicine, Philipps University Marburg, a member of Universities Giessen and Marburg Lung Center (H.G., H.R.), Marburg - all in Germany; Upper Airways Research Laboratory, University Hospital Ghent, Ghent, Belgium (N.Z., C.B.); and the Division of Ear, Nose, and Throat Diseases, Clintec, Karolinska Institute, Stockholm (N.Z., C.B.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Kulinski JM, Muñoz-Cano R, Olivera A. Sphingosine-1-phosphate and other lipid mediators generated by mast cells as critical players in allergy and mast cell function. Eur J Pharmacol 2015; 778:56-67. [PMID: 25941085 DOI: 10.1016/j.ejphar.2015.02.058] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 02/09/2015] [Accepted: 02/17/2015] [Indexed: 12/20/2022]
Abstract
Sphingosine-1-phosphate (S1P), platelet activating factor (PAF) and eicosanoids are bioactive lipid mediators abundantly produced by antigen-stimulated mast cells that exert their function mostly through specific cell surface receptors. Although it has long been recognized that some of these bioactive lipids are potent regulators of allergic diseases, their exact contributions to disease pathology have been obscured by the complexity of their mode of action and the regulation of their metabolism. Indeed, the effects of such lipids are usually mediated by multiple receptor subtypes that may differ in their signaling mechanisms and functions. In addition, their actions may be elicited by cell surface receptor-independent mechanisms. Furthermore, these lipids may be converted into metabolites that exhibit different functionalities, adding another layer of complexity to their overall biological responses. In some instances, a second wave of lipid mediator synthesis by both mast cell and non-mast cell sources may occur late during inflammation, bringing about additional roles in the altered environment. New evidence also suggests that bioactive lipids in the local environment can fine-tune mast cell maturation and phenotype, and thus their responsiveness. A better understanding of the subtleties of the spatiotemporal regulation of these lipid mediators, their receptors and functions may aid in the pursuit of pharmacological applications for allergy treatments.
Collapse
Affiliation(s)
- Joseph M Kulinski
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA.
| | - Rosa Muñoz-Cano
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA.
| | - Ana Olivera
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA.
| |
Collapse
|
19
|
Pergola C, Gerstmeier J, Mönch B, Çalışkan B, Luderer S, Weinigel C, Barz D, Maczewsky J, Pace S, Rossi A, Sautebin L, Banoglu E, Werz O. The novel benzimidazole derivative BRP-7 inhibits leukotriene biosynthesis in vitro and in vivo by targeting 5-lipoxygenase-activating protein (FLAP). Br J Pharmacol 2015; 171:3051-64. [PMID: 24641614 DOI: 10.1111/bph.12625] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Revised: 01/25/2014] [Accepted: 02/03/2014] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Leukotrienes (LTs) are inflammatory mediators produced via the 5-lipoxygenase (5-LOX) pathway and are linked to diverse disorders, including asthma, allergic rhinitis and cardiovascular diseases. We recently identified the benzimidazole derivative BRP-7 as chemotype for anti-LT agents by virtual screening targeting 5-LOX-activating protein (FLAP). Here, we aimed to reveal the in vitro and in vivo pharmacology of BRP-7 as an inhibitor of LT biosynthesis. EXPERIMENTAL APPROACH We analysed LT formation and performed mechanistic studies in human neutrophils and monocytes, in human whole blood (HWB) and in cell-free assays. The effectiveness of BRP-7 in vivo was evaluated in rat carrageenan-induced pleurisy and mouse zymosan-induced peritonitis. KEY RESULTS BRP-7 potently suppressed LT formation in neutrophils and monocytes and this was accompanied by impaired 5-LOX co-localization with FLAP. Neither the cellular viability nor the activity of 5-LOX in cell-free assays was affected by BRP-7, indicating that a functional FLAP is needed for BRP-7 to inhibit LTs, and FLAP bound to BRP-7 linked to a solid matrix. Compared with the FLAP inhibitor MK-886, BRP-7 did not significantly inhibit COX-1 or microsomal prostaglandin E2 synthase-1, implying the selectivity of BRP-7 for FLAP. Finally, BRP-7 was effective in HWB and impaired inflammation in vivo, in rat pleurisy and mouse peritonitis, along with reducing LT levels. CONCLUSIONS AND IMPLICATIONS BRP-7 potently suppresses LT biosynthesis by interacting with FLAP and exhibits anti-inflammatory effectiveness in vivo, with promising potential for further development.
Collapse
Affiliation(s)
- C Pergola
- Institute of Pharmacy, Friedrich-Schiller-University Jena, Jena, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Antoniu SA. Targeting 5-lipoxygenase-activating protein in asthma and chronic obstructive pulmonary disease. Expert Opin Ther Targets 2014; 18:1285-92. [PMID: 25213852 DOI: 10.1517/14728222.2014.945425] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION In asthma and chronic obstructive pulmonary disease (COPD), there is an unmet therapeutic need for the anti-inflammatory therapies, and the identification of therapeutic targets and potent corresponding therapies is necessary. Although inhaled corticosteroids and leukotriene modifiers are most effective in asthma they are still not always capable of appropriately controlling the disease. In COPD, the therapeutic gap is even larger because inhaled corticosteroids and other anti-inflammatory therapies are not beneficial in all disease subsets. AREAS COVERED The role of the 5-lipoxygenase-activating protein (FLAP) in generating proinflammatory molecules such as leukotrienes is discussed, highlighting, in particular, its potential as a therapeutic target in asthma and COPD. The preclinical data on FLAP inhibitors are discussed. The clinical data on the FLAP inhibitors investigated so far for these diseases are analyzed. EXPERT OPINION FLAP inhibitors have emerged during the past decade as a promising therapeutic class in asthma and COPD, but there exists only a limited amount of data supporting their efficacy in these diseases. This might be due to the fact that the development of some of the molecules discussed was abandoned. Such therapies might be of particular interest in COPD and in asthma-COPD overlap syndrome.
Collapse
Affiliation(s)
- Sabina Antonela Antoniu
- University of Medicine and Pharmacy Grigore T Popa Iasi, Interdisciplinary Medicine , 16 Universitatii Str, Iasi, 700115 , Romania
| |
Collapse
|
21
|
Molecular targets on mast cells and basophils for novel therapies. J Allergy Clin Immunol 2014; 134:530-44. [DOI: 10.1016/j.jaci.2014.03.007] [Citation(s) in RCA: 110] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 02/24/2014] [Accepted: 03/07/2014] [Indexed: 01/14/2023]
|
22
|
Chaudhuri R, Norris V, Kelly K, Zhu CQ, Ambery C, Lafferty J, Cameron E, Thomson NC. Effects of a FLAP inhibitor, GSK2190915, in asthmatics with high sputum neutrophils. Pulm Pharmacol Ther 2013; 27:62-9. [PMID: 24333186 DOI: 10.1016/j.pupt.2013.11.007] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Revised: 11/19/2013] [Accepted: 11/30/2013] [Indexed: 11/18/2022]
Abstract
Patients with refractory asthma frequently have neutrophilic airway inflammation and respond poorly to inhaled corticosteroids. This study evaluated the effects of an oral 5-lipoxygenase-activating protein (FLAP) inhibitor, GSK2190915, in patients with asthma and elevated sputum neutrophils. Patients received 14 (range 13-16) days treatment with GSK2190915 100 mg and placebo with a minimum 14 day washout in a double-blind, cross-over, randomised design (N = 14). Sputum induction was performed twice pre-dose in each treatment period to confirm sputum neutrophilia, and twice at the end of each treatment period. The primary endpoint was the percentage and absolute sputum neutrophil count, averaged for end-of-treatment visits. GSK2190915 did not significantly reduce mean percentage sputum neutrophils (GSK2190915-placebo difference [95% CI]: -0.9 [-12.0, 10.3]), or mean sputum neutrophil counts (GSK2190915/placebo ratio [95% CI]: 1.06 [0.43, 2.61]). GSK2190915 resulted in a marked suppression (>90%) of sputum LTB4 and urine LTE4, but did not alter clinical endpoints. There were no safety issues. Despite suppressing the target mediator LTB4, FLAP inhibitor GSK2190915 had no short-term effect on sputum cell counts or clinical endpoints in patients with asthma and sputum neutrophilia.
Collapse
Affiliation(s)
- R Chaudhuri
- Institute of Infection, Immunity & Inflammation, University of Glasgow and Respiratory Medicine, Gartnavel General Hospital, Glasgow, UK.
| | - V Norris
- Immuno-Inflammation Therapy Area, GlaxoSmithKline, Stevenage, UK
| | - K Kelly
- Clinical Pharmacology Science and Study Operations, GlaxoSmithKline, Uxbridge, UK
| | - C-Q Zhu
- Clinical Statistics, GlaxoSmithKline, Uxbridge, UK
| | - C Ambery
- Clinical Pharmacology Modelling and Simulation, GlaxoSmithKline, Uxbridge, UK
| | - J Lafferty
- Institute of Infection, Immunity & Inflammation, University of Glasgow and Respiratory Medicine, Gartnavel General Hospital, Glasgow, UK
| | - E Cameron
- Institute of Infection, Immunity & Inflammation, University of Glasgow and Respiratory Medicine, Gartnavel General Hospital, Glasgow, UK
| | - N C Thomson
- Institute of Infection, Immunity & Inflammation, University of Glasgow and Respiratory Medicine, Gartnavel General Hospital, Glasgow, UK
| |
Collapse
|
23
|
Singh D, Boyce M, Norris V, Kent SE, Bentley JH. Inhibition of the early asthmatic response to inhaled allergen by the 5-lipoxygenase activating protein inhibitor GSK2190915: a dose-response study. Int J Gen Med 2013; 6:897-903. [PMID: 24357936 PMCID: PMC3862733 DOI: 10.2147/ijgm.s51364] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Background GSK2190915, a 5-lipoxygenase activating protein inhibitor, inhibits the production of cysteinyl leukotrienes and leukotriene B4 and 5-oxo-6,8,11,14-eicosatetraenoic acid. We have previously reported that GSK2190915 100 mg daily inhibits early and late asthmatic responses to inhaled allergen; the effects of lower doses have not been reported. This study assessed the dose–response effects of GSK2190915 10 mg and 50 mg on the early asthmatic response (EAR) to inhaled allergen. Methods Nineteen subjects with mild asthma and an EAR were enrolled in a randomized, double-blind, three-way crossover study of GSK2190915 10 mg, 50 mg, and placebo orally once-daily for 3 days. Allergen challenge was performed 2 hours after the third dose. Results Compared with placebo, GSK2190915 10 mg and 50 mg caused significant, dose-dependent attenuation of the minimum forced expiratory volume at 1 second (FEV1) absolute change from baseline; mean treatment differences were 0.21 L (95% confidence interval [CI] 0.04 L, 0.38 L) and 0.41 L (95% CI 0.24 L, 0.58 L), respectively. GSK2190915 50 mg was more effective than 10 mg; mean difference between treatments was 0.20 L, (95% CI 0.03 L, 0.36 L). Compared with placebo, GSK2190915 50 mg, but not 10 mg, significantly inhibited the weighted mean FEV1 absolute change from baseline. Conclusion GSK2190915 50 mg attenuated the EAR similarly to GSK2190915 100 mg in our previous study, suggesting 50 mg is at the top of the dose–response curve. GSK2190915 10 mg is a suboptimal dose. The EAR can be used to assess the therapeutic dose of a new treatment for asthma.
Collapse
Affiliation(s)
- Dave Singh
- University of Manchester, Medicines Evaluation Unit, University Hospital of South Manchester, Manchester, UK
| | | | | | | | | |
Collapse
|
24
|
Inhaled allergen bronchoprovocation tests. J Allergy Clin Immunol 2013; 132:1045-1055.e6. [PMID: 24119772 DOI: 10.1016/j.jaci.2013.08.023] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Revised: 07/16/2013] [Accepted: 08/20/2013] [Indexed: 11/23/2022]
Abstract
The allergen bronchoprovocation test is a long-standing exacerbation model of allergic asthma that can induce several clinical and pathophysiologic features of asthma in sensitized subjects. Standardized allergen challenge is primarily a research tool, and when properly conducted by qualified and experienced investigators, it is safe and highly reproducible. In combination with validated airway sampling and sensitive detection techniques, allergen challenge allows the study of several features of the physiology of mainly TH2 cell-driven asthma in relation to the kinetics of the underlying airway pathology occurring during the allergen-induced late response. Furthermore, given the small within-subject variability in allergen-induced airway responses, allergen challenge offers an adequate disease model for the evaluation of new (targeted) controller therapies for asthma in a limited number of subjects. In proof-of-efficacy studies thus far, allergen challenge showed a fair positive predicted value and an excellent negative predictive value for the actual clinical efficacy of new antiasthma therapies, underscoring its important role in early drug development. In this review we provide recommendations on challenge methods, response measurements, sample size, safety, and harmonization for future applications.
Collapse
|
25
|
Di Gennaro A, Haeggström JZ. Targeting leukotriene B4 in inflammation. Expert Opin Ther Targets 2013; 18:79-93. [PMID: 24090264 DOI: 10.1517/14728222.2013.843671] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Leukotriene (LT) B(4) is a powerful proinflammatory lipid mediator and triggers adherence to the endothelium, activates and recruits leukocytes to the site of injury. When formed in excess, LTB(4) plays a pathogenic role and may sustain chronic inflammation in diseases such as asthma, rheumatoid arthritis, and inflammatory bowel disease. Recent investigations have also indicated that LTB(4) is involved in cardiovascular diseases. AREAS COVERED As the 5-lipoxygenase pathway involves several discrete, tightly coupled, enzymes, which convert the substrate, 'step by step', into bioactive products, several different strategies have been used to target LTB(4) as a means to treat inflammation. Here, we discuss recent findings regarding the development of selective enzyme inhibitors and antagonists for LTB(4) receptors, as well as their application in preclinical and clinical studies. EXPERT OPINION Components of the 5-lipoxygenase pathway have received considerable attention as candidate drug targets resulting in one new class of medications against asthma, that is, the antileukotrienes. However, efforts to specifically target LTB(4) have not yet been fruitful in the clinical setting, in spite of very promising preclinical data. Recently, crystal structures along with hitherto unknown functions of key enzymes in the leukotriene cascade have emerged, offering new opportunities for drug development and, with time, pharmacological intervention in LTB(4)-mediated pathologies.
Collapse
Affiliation(s)
- Antonio Di Gennaro
- Karolinska Institutet, Department of Medical Biochemistry and Biophysics, Division of Chemistry 2 , Scheeles väg 2, Stockholm, S-171 77 , Sweden
| | | |
Collapse
|
26
|
Bain G, King CD, Schaab K, Rewolinski M, Norris V, Ambery C, Bentley J, Yamada M, Santini AM, van de Wetering de Rooij J, Stock N, Zunic J, Hutchinson JH, Evans JF. Pharmacodynamics, pharmacokinetics and safety of GSK2190915, a novel oral anti-inflammatory 5-lipoxygenase-activating protein inhibitor. Br J Clin Pharmacol 2013; 75:779-90. [PMID: 22803688 DOI: 10.1111/j.1365-2125.2012.04386.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2012] [Accepted: 06/16/2012] [Indexed: 11/27/2022] Open
Abstract
AIM To assess the pharmacokinetics, pharmacodynamics, safety and tolerability of the 5-lipoxygenase-activating protein inhibitor, GSK2190915, after oral dosing in two independent phase I studies, one in Western European and one in Japanese subjects, utilizing different formulations. METHOD Western European subjects received single (50-1000 mg) or multiple (10-450 mg) oral doses of GSK2190915 or placebo in a dose-escalating manner. Japanese subjects received three of four GSK2190915 doses (10-200 mg) plus placebo once in a four period crossover design. Blood samples were collected for GSK2190915 concentrations and blood and urine were collected to measure leukotriene B₄ and leukotriene E₄, respectively, as pharmacodynamic markers of drug activity. RESULTS There was no clear difference in adverse events between placebo and active drug-treated subjects in either study. Maximum plasma concentrations of GSK2190915 and area under the curve increased in a dose-related manner and mean half-life values ranged from 16-34 h. Dose-dependent inhibition of blood leukotriene B₄ production was observed and near complete inhibition of urinary leukotriene E₄ excretion was shown at all doses except the lowest dose. The EC₅₀ values for inhibition of LTB₄ were 85 nM and 89 nM in the Western European and Japanese studies, respectively. CONCLUSION GSK2190915 is well-tolerated with pharmacokinetics and pharmacodynamics in Western European and Japanese subjects that support once daily dosing for 24 h inhibition of leukotrienes. Doses of ≥50 mg show near complete inhibition of urinary leukotriene E₄ at 24 h post-dose, whereas doses of ≥150 mg are required for 24 h inhibition of blood LTB₄.
Collapse
|
27
|
Hofmann B, Steinhilber D. 5-Lipoxygenase inhibitors: a review of recent patents (2010-2012). Expert Opin Ther Pat 2013; 23:895-909. [PMID: 23600432 DOI: 10.1517/13543776.2013.791678] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION 5-Lipoxygenase (5-LO) is a crucial enzyme of the arachidonic acid (AA) cascade and catalyzes the formation of bioactive leukotrienes (LTs) with the help of FLAP, the 5-LO-activating protein. LTs are inflammatory mediators playing a pathophysiological role in different diseases like asthma, allergic rhinitis as well as cardiovascular diseases and certain types of cancer. With the rising number of indications for anti-LT therapy, 5-LO inhibitor drug development becomes increasingly important. AREAS COVERED Here, both recent findings regarding the pathophysiological role of 5-LO and the patents claimed for 5-LO inhibitors are discussed. Focusing on direct inhibitors, several patents disclosing FLAP antagonists are also subject of this review. Novel compounds include 1,5-diarylpyrazoles, indolizines and indoles and several natural product extracts. EXPERT OPINION Evaluation of the patent activities revealed only quite moderate action. Nevertheless, several auspicious drug-like molecules were disclosed. It seems that in the near future, FLAP inhibitors can be expected to enter the market for the treatment of asthma. With the resolved structure of 5-LO, structure-based drug design is now applicable. Together with the identification of downstream enzyme inhibitors and dual-targeting drugs within the AA cascade, several tools are at hand to cope with 5-LOs increasing pathophysiological roles.
Collapse
Affiliation(s)
- Bettina Hofmann
- Goethe University Frankfurt, Institute of Pharmaceutical Chemistry, Max-von-Laue-Str. 9, 60438, Frankfurt, Germany
| | | |
Collapse
|