1
|
Barzaghi F, Moratti M, Panza G, Rivalta B, Giardino G, De Rosa A, Baselli LA, Chinello M, Marzollo A, Montin D, Marinoni M, Costagliola G, Ricci S, Lodi L, Martire B, Milito C, Trizzino A, Tommasini A, Zecca M, Badolato R, Cancrini C, Lougaris V, Pignata C, Conti F. Report of the Italian Cohort with Activated Phosphoinositide 3-Kinase δ Syndrome in the Target Therapy Era. J Clin Immunol 2024; 45:58. [PMID: 39714594 DOI: 10.1007/s10875-024-01835-1] [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: 08/12/2024] [Accepted: 11/05/2024] [Indexed: 12/24/2024]
Abstract
BACKGROUND Activated Phosphoinositide 3-Kinase (PI3K) δ Syndrome (APDS), an inborn error of immunity due to upregulation of the PI3K pathway, leads to recurrent infections and immune dysregulation (lymphoproliferation and autoimmunity). METHODS Clinical and genetic data of 28 APDS patients from 25 unrelated families were collected from fifteen Italian centers. RESULTS Patients were genetically confirmed with APDS-1 (n = 20) or APDS-2 (n = 8), with pathogenic mutations in the PIK3CD or PIK3R1 genes. The median age at diagnosis was 15.5 years, with a median follow-up of 74 months (range 6-384). The main presenting symptoms were respiratory tract infections alone (57%) or associated with lymphoproliferation (17%). Later, non-clonal lymphoproliferation was the leading clinical sign (86%), followed by respiratory infections (79%) and gastrointestinal complications (43%). Malignant lymphoproliferative disorders, all EBV-encoding RNA (EBER)-positive at the histological analysis, occurred in 14% of patients aged 17-19 years, highlighting the role of EBV in lymphomagenesis in this disorder. Diffuse large B-cell lymphoma was the most frequent. Immunological work-up revealed combined T/B cell abnormalities in most patients. Treatment strategies included immunosuppression and PI3K/Akt/mTOR inhibitor therapy. Rapamycin, employed in 36% of patients, showed efficacy in controlling lymphoproliferation, while selective PI3Kδ inhibitor leniolisib, administered in 32% of patients, was beneficial on both infections and immune dysregulation. Additionally, three patients underwent successful HSCT due to recurrent infections despite ongoing prophylaxis or lymphoproliferation poorly responsive to Rapamycin. CONCLUSIONS This study underscores the clinical heterogeneity and challenging diagnosis of APDS, highlighting the importance of multidisciplinary management tailored to individual needs and further supporting leniolisib efficacy.
Collapse
Affiliation(s)
- Federica Barzaghi
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
- Pediatric Immunohematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Mattia Moratti
- Pediatric Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, University of Bologna, Bologna, Italy
- Department of Systems Medicine, University of Tor Vergata, Rome, Italy
| | - Giuseppina Panza
- Pediatric Immunohematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Beatrice Rivalta
- Research and Clinical Unit of Primary Immunodeficiencies, IRCCS Bambin Gesù Children Hospital, Rome, Italy
- PhD Program in Immunology, Molecular Medicine and Applied Biotechnology, University of Rome Tor Vergata, Rome, Italy
| | - Giuliana Giardino
- Pediatric Section, Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Antonio De Rosa
- Pediatric Section, Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Lucia Augusta Baselli
- Pediatric Immunorheumatology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Matteo Chinello
- Pediatric Hematology-Oncology, Department of Mother and Child, Azienda Ospedaliera Universitaria Integrata Verona, Verona, Italy
| | - Antonio Marzollo
- Pediatric Hematology, Oncology and Stem Cell Transplant Division, Padua University Hospital, Padua, Italy
| | - Davide Montin
- Department of Pediatric and Public Health Sciences, University of Torino and Regina Margherita Children's Hospital, AOU Città della Salute e della Scienza di Torino, Turin, Italy
| | - Maddalena Marinoni
- SSD Oncoematologia Pediatrica, Dipartimento materno infantile, Ospedale Filippo del Ponte, ASST Sette Laghi, Varese, Italy
| | - Giorgio Costagliola
- Section of Pediatric Hematology and Oncology, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Silvia Ricci
- Department of Health Sciences, University of Florence, Florence, Italy
- Immunology Unit, Department of Pediatrics, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Lorenzo Lodi
- Department of Health Sciences, University of Florence, Florence, Italy
- Immunology Unit, Department of Pediatrics, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Baldassarre Martire
- Maternal and Child Department, Unit of Pediatrics and Neonatology, "Monsignor A.R. Dimiccoli" Hospital, Barletta, Italy
| | - Cinzia Milito
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Antonino Trizzino
- Department of Pediatric Hematology and Oncology, "ARNAS Civico Di Cristina Benfratelli" Hospital, Palermo, Italy
| | - Alberto Tommasini
- Department of Medical Sciences, University of Trieste, Trieste, Italy
- Institute for Maternal and Child Health, IRCCS Burlo Garofalo, Trieste, Italy
| | - Marco Zecca
- Paediatric Haematology and Oncology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Raffaele Badolato
- Molecular Medicine Institute "Angelo Nocivelli", Department of Clinical and Experimental Sciences, University of Brescia and ASST Spedali civili, Brescia, Italy
| | - Caterina Cancrini
- Research and Clinical Unit of Primary Immunodeficiencies, IRCCS Bambin Gesù Children Hospital, Rome, Italy
- Department of System Medicine, Pediatric Chair, University of Tor Vergata, Rome, Italy
| | - Vassilios Lougaris
- Pediatrics Clinic, Department of Clinical and Experimental Sciences, University of Brescia, Azienda Socio Sanitaria Territoriale Spedali Civili di Brescia, Brescia, Italy
| | - Claudio Pignata
- Department of Translational Medical Science, Pediatric Section, Federico II University, Via S. Pansini, 5, 80131 , Naples, Italy.
| | - Francesca Conti
- Pediatric Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, University of Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| |
Collapse
|
2
|
Govoni M, Bassi M, Girardello L, Lucci G, Rony F, Charretier R, Galkin D, Faietti ML, Pioselli B, Modafferi G, Benfeitas R, Bonatti M, Miglietta D, Clark J, Pedersen F, Kirsten AM, Beeh KM, Kornmann O, Korn S, Ludwig-Sengpiel A, Watz H. CHF6523 data suggest that the phosphoinositide 3-kinase delta isoform is not a suitable target for the management of COPD. Respir Res 2024; 25:380. [PMID: 39427187 PMCID: PMC11491004 DOI: 10.1186/s12931-024-02999-5] [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: 06/11/2024] [Accepted: 10/02/2024] [Indexed: 10/21/2024] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is a chronic inflammatory condition. Given patients with COPD continue to experience exacerbations despite the availability of effective therapies, anti-inflammatory treatments targeting novel pathways are needed. Kinases, notably the phosphoinositide 3-kinases (PI3K), are thought to be involved in chronic airway inflammation, with this pathway proposed as a critical regulator of inflammation and oxidative stress response in COPD. CHF6523 is an inhaled PI3Kδ inhibitor that has shown positive preclinical results. This manuscript reports the results of a study of CHF6523 in patients with stable COPD (chronic bronchitis phenotype), and who had evidence of type-2 inflammation. METHODS This randomised, double-blind, placebo-controlled, two-way crossover study comprised two 28-day treatment periods separated by a 28-day washout. Patients (N = 44) inhaled CHF6523 in one period, and placebo in the other, both twice daily. The primary objective was to assess the safety and tolerability of CHF6523; the secondary objective was to assess CHF6523 pharmacokinetics. Exploratory endpoints included target engagement (the relative reduction in phosphatidylinositol (3,4,5)-trisphosphate [PIP3]), pharmacodynamic evaluations such as airflow obstruction, and hyperinflation, and to identify biomarker(s) of drug response using proteomics and transcriptomics. RESULTS CHF6523 plasma pharmacokinetics were characterised by an early maximum concentration (Cmax), reached 15 and 10 min after dosing on Days 1 and 28, respectively, followed by a rapid decline. Systemic exposure on Day 28 showed limited accumulation, with ratios < 1.6 for Cmax and area under the curve from 0 to 12 h post-dose, and with steady state achieved on Day 20. Target engagement was confirmed by a significant 29.7% reduction from baseline in induced sputum PIP3 (29.5% reduction vs. placebo; adjusted ratio 0.705 [0.580, 0.856]; p = 0.001), but this did not translate into an anti-inflammatory pharmacodynamic effect, as assessed through measures including biomarkers and multi-omics. Additionally, although CHF6523 was generally well-tolerated, 95.2% of patients reported cough as an adverse event, most mild to moderate and resolving within one-hour post-dose. CONCLUSIONS These data, together with those from other PI3K inhibitors, suggest that PI3Kδ is not a suitable pathway for the management of COPD, as the achieved target engagement did not translate into any pharmacodynamic anti-inflammatory effect. TRIAL REGISTRATION ClinicalTrials.gov (NCT04032535); posted 23rd July 2019.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Martina Bonatti
- Chiesi Farmaceutici SpA, Parma, Italy
- Respiratory Medicine Unit, Department of Medicine Solna and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | | | - Frauke Pedersen
- Velocity Clinical Research Grosshansdorf, Formerly Known as Pulmonary Research Institute at LungenClinic Grosshansdorf, Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Grosshansdorf, Germany
| | - Anne-Marie Kirsten
- Velocity Clinical Research Grosshansdorf, Formerly Known as Pulmonary Research Institute at LungenClinic Grosshansdorf, Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Grosshansdorf, Germany
| | | | - Oliver Kornmann
- IKF Pneumologie Frankfurt, Clinical Research Centre Respiratory Diseases, Frankfurt, Germany
| | - Stephanie Korn
- IKF Pneumologie Mainz, Mainz, Germany
- Thoraxklinik Heidelberg, Heidelberg, Germany
| | - Andrea Ludwig-Sengpiel
- Velocity Clinical Research Lübeck GmbH, Formerly Known as KLB Health Research, Lübeck, Germany
| | - Henrik Watz
- Velocity Clinical Research Grosshansdorf, Formerly Known as Pulmonary Research Institute at LungenClinic Grosshansdorf, Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Grosshansdorf, Germany
| |
Collapse
|
3
|
Pinkerton JW, Preite S, Piras A, Zervas D, Markou T, Freeman MS, Hofving T, Ivarsson E, Bonvini SJ, Brailsford W, Yrlid L, Belvisi MG, Birrell MA. PI3Kγδ inhibition suppresses key disease features in a rat model of asthma. Respir Res 2024; 25:175. [PMID: 38654248 PMCID: PMC11040934 DOI: 10.1186/s12931-024-02814-1] [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: 01/05/2024] [Accepted: 04/15/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND Two isoforms of Phosphoinositide 3-kinase (PI3K), p110γ and p110δ, are predominantly expressed in leukocytes and represent attractive therapeutic targets for the treatment of allergic asthma. The study aim was to assess the impact of administration of an inhaled PI3Kγδ inhibitor (AZD8154) in a rat model of asthma. METHODS Firstly, we checked that the tool compound, AZD8154, inhibited rat PI3K γ & δ kinases using rat cell-based assays. Subsequently, a time-course study was conducted in a rat model of asthma to assess PI3K activity in the lung and how it is temporally associated with other key transcription pathways and asthma like features of the model. Finally, the impact on lung dosed AZD8154 on target engagement, pathway specificity, airway inflammation and lung function changes was assessed. RESULTS Data showed that AZD8154 could inhibit rat PI3K γ & δ isoforms and, in a rat model of allergic asthma the PI3K pathway was activated in the lung. Intratracheal administration of AZD8154 caused a dose related suppression PI3K pathway activation (reduction in pAkt) and unlike after budesonide treatment, STAT and NF-κB pathways were not affected by AZD8154. The suppression of the PI3K pathway led to a marked inhibition of airway inflammation and reduction in changes in lung function. CONCLUSION These data show that a dual PI3Kγδ inhibitor suppress key features of disease in a rat model of asthma to a similar degree as budesonide and indicate that dual PI3Kγδ inhibition may be an effective treatment for people suffering from allergic asthma.
Collapse
Affiliation(s)
- James W Pinkerton
- Early Respiratory & Immunology, Biopharmaceuticals R&D AstraZeneca, Gothenburg, Sweden
- Respiratory Pharmacology group, Airway Disease section, NHLI, Imperial College, London, UK
| | - Silvia Preite
- Early Respiratory & Immunology, Biopharmaceuticals R&D AstraZeneca, Gothenburg, Sweden
| | - Antonio Piras
- Early Respiratory & Immunology, Biopharmaceuticals R&D AstraZeneca, Gothenburg, Sweden
| | - Dimitrios Zervas
- Early Respiratory & Immunology, Biopharmaceuticals R&D AstraZeneca, Gothenburg, Sweden
- Respiratory Pharmacology group, Airway Disease section, NHLI, Imperial College, London, UK
| | - Thomais Markou
- Early Respiratory & Immunology, Biopharmaceuticals R&D AstraZeneca, Gothenburg, Sweden
- Respiratory Pharmacology group, Airway Disease section, NHLI, Imperial College, London, UK
| | - Mark S Freeman
- Early Respiratory & Immunology, Biopharmaceuticals R&D AstraZeneca, Gothenburg, Sweden
- Respiratory Pharmacology group, Airway Disease section, NHLI, Imperial College, London, UK
| | - Tobias Hofving
- Early Respiratory & Immunology, Biopharmaceuticals R&D AstraZeneca, Gothenburg, Sweden
| | - Emil Ivarsson
- Early Respiratory & Immunology, Biopharmaceuticals R&D AstraZeneca, Gothenburg, Sweden
| | - Sara J Bonvini
- Early Respiratory & Immunology, Biopharmaceuticals R&D AstraZeneca, Gothenburg, Sweden
- Respiratory Pharmacology group, Airway Disease section, NHLI, Imperial College, London, UK
| | - Wayne Brailsford
- Early Respiratory & Immunology, Biopharmaceuticals R&D AstraZeneca, Gothenburg, Sweden
| | - Linda Yrlid
- Early Respiratory & Immunology, Biopharmaceuticals R&D AstraZeneca, Gothenburg, Sweden
| | - Maria G Belvisi
- Early Respiratory & Immunology, Biopharmaceuticals R&D AstraZeneca, Gothenburg, Sweden
- Respiratory Pharmacology group, Airway Disease section, NHLI, Imperial College, London, UK
| | - Mark A Birrell
- Early Respiratory & Immunology, Biopharmaceuticals R&D AstraZeneca, Gothenburg, Sweden.
- Respiratory Pharmacology group, Airway Disease section, NHLI, Imperial College, London, UK.
| |
Collapse
|
4
|
Zhu J, Meng H, Li X, Jia L, Xu L, Cai Y, Chen Y, Jin J, Yu L. Optimization of virtual screening against phosphoinositide 3-kinase delta: Integration of common feature pharmacophore and multicomplex-based molecular docking. Comput Biol Chem 2024; 109:108011. [PMID: 38198965 DOI: 10.1016/j.compbiolchem.2023.108011] [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: 11/23/2023] [Revised: 12/29/2023] [Accepted: 12/29/2023] [Indexed: 01/12/2024]
Abstract
Extensive research has accumulated which suggests that phosphatidylinositol 3-kinase delta (PI3Kδ) is closely related to the occurrence and development of various human diseases, making PI3Kδ a highly promising drug target. However, PI3Kδ exhibits high homology with other members of the PI3K family, which poses significant challenges to the development of PI3Kδ inhibitors. Therefore, in the present study, a hybrid virtual screening (VS) approach based on a ligand-based pharmacophore model and multicomplex-based molecular docking was developed to find novel PI3Kδ inhibitors. 13 crystal structures of the human PI3Kδ-inhibitor complex were collected to establish models. The inhibitors were extracted from the crystal structures to generate the common feature pharmacophore. The crystallographic protein structures were used to construct a naïve Bayesian classification model that integrates molecular docking based on multiple PI3Kδ conformations. Subsequently, three VS protocols involving sequential or parallel molecular docking and pharmacophore approaches were employed. External predictions demonstrated that the protocol combining molecular docking and pharmacophore resulted in a significant improvement in the enrichment of active PI3Kδ inhibitors. Finally, the optimal VS method was utilized for virtual screening against a large chemical database, and some potential hit compounds were identified. We hope that the developed VS strategy will provide valuable guidance for the discovery of novel PI3Kδ inhibitors.
Collapse
Affiliation(s)
- Jingyu Zhu
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, Jiangsu 214122, China.
| | - Huiqin Meng
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Xintong Li
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Lei Jia
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Lei Xu
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, Jiangsu University of Technology, Changzhou 213001, China
| | - Yanfei Cai
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Yun Chen
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Jian Jin
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Li Yu
- School of Inspection and Testing Certification, Changzhou Vocational Institute of Engineering, Changzhou, Jiangsu 213164, China.
| |
Collapse
|
5
|
McClean N, Hasday JD, Shapiro P. Progress in the development of kinase inhibitors for treating asthma and COPD. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2023; 98:145-178. [PMID: 37524486 DOI: 10.1016/bs.apha.2023.04.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/02/2023]
Abstract
Current therapies to mitigate inflammatory responses involved in airway remodeling and associated pathological features of asthma and chronic obstructive pulmonary disease (COPD) are limited and largely ineffective. Inflammation and the release of cytokines and growth factors activate kinase signaling pathways that mediate changes in airway mesenchymal cells such as airway smooth muscle cells and lung fibroblasts. Proliferative and secretory changes in mesenchymal cells exacerbate the inflammatory response and promote airway remodeling, which is often characterized by increased airway smooth muscle mass, airway hyperreactivity, increased mucus secretion, and lung fibrosis. Thus, inhibition of relevant kinases has been viewed as a potential therapeutic approach to mitigate the debilitating and, thus far, irreversible airway remodeling that occurs in asthma and COPD. Despite FDA approval of several kinase inhibitors for the treatment of proliferative disorders, such as cancer and inflammation associated with rheumatoid arthritis and ulcerative colitis, none of these drugs have been approved to treat asthma or COPD. This review will provide a brief overview of the role kinases play in the pathology of asthma and COPD and an update on the status of kinase inhibitors currently in clinical trials for the treatment of obstructive pulmonary disease. In addition, potential issues associated with the current kinase inhibitors, which have limited their success as therapeutic agents in treating asthma or COPD, and alternative approaches to target kinase functions will be discussed.
Collapse
Affiliation(s)
- Nathaniel McClean
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, United States
| | - Jeffery D Hasday
- Department of Medicine, Division of Pulmonary Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Paul Shapiro
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, United States.
| |
Collapse
|
6
|
Begg M, Amour A, Jarvis E, Tang T, Franco SS, Want A, Beerahee M, Fernando D, Karkera Y, Sander C, Southworth T, Singh D, Clark J, Nejentsev S, Okkenhaug K, Condliffe A, Chandra A, Cahn A, Hall EB. An open label trial of nemiralisib, an inhaled PI3 kinase delta inhibitor for the treatment of Activated PI3 kinase Delta Syndrome. Pulm Pharmacol Ther 2023; 79:102201. [PMID: 36841351 DOI: 10.1016/j.pupt.2023.102201] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 02/08/2023] [Accepted: 02/22/2023] [Indexed: 02/26/2023]
Abstract
Activated PI3Kδ Syndrome (APDS) is a rare inherited inborn error of immunity caused by mutations that constitutively activate the p110 delta isoform of phosphoinositide 3-kinase (PI3Kδ), resulting in recurring pulmonary infections. Currently no licensed therapies are available. Here we report the results of an open-label trial in which five subjects were treated for 12 weeks with nemiralisib, an inhaled inhibitor of PI3Kδ, to determine safety, systemic exposure, together with lung and systemic biomarker profiles (Clinicaltrial.gov: NCT02593539). Induced sputum was captured to measure changes in phospholipids and inflammatory mediators, and blood samples were collected to assess pharmacokinetics of nemiralisib, and systemic biomarkers. Nemiralisib was shown to have an acceptable safety and tolerability profile, with cough being the most common adverse event, and no severe adverse events reported during the study. No meaningful changes in phosphatidylinositol (3,4,5)-trisphosphate (PIP3; the enzyme product of PI3Kδ) or downstream inflammatory markers in induced sputum, were observed following nemiralisib treatment. Similarly, there were no meaningful changes in blood inflammatory markers, or lymphocytes subsets. Systemic levels of nemiralisib were higher in subjects in this study compared to previous observations. While nemiralisib had an acceptable safety profile, there was no convincing evidence of target engagement in the lung following inhaled dosing and no downstream effects observed in either the lung or blood compartments. We speculate that this could be explained by nemiralisib not being retained in the lung for sufficient duration, suggested by the increased systemic exposure, perhaps due to pre-existing structural lung damage. In this study investigating a small number of subjects with APDS, nemiralisib appeared to be safe and well-tolerated. However, data from this study do not support the hypothesis that inhaled treatment with nemiralisib would benefit patients with APDS.
Collapse
Affiliation(s)
- Malcolm Begg
- Research, GlaxoSmithKline, Medicines Research Centre, Gunnels Wood Road, Stevenage, UK.
| | - Augustin Amour
- Research, GlaxoSmithKline, Medicines Research Centre, Gunnels Wood Road, Stevenage, UK
| | - Emily Jarvis
- Clinical Statistics, Development, GlaxoSmithKline, Medicines Research Centre, Gunnels Wood Road, Stevenage, UK
| | - Teresa Tang
- Global Medical Safety, Development, GlaxoSmithKline, GSK House, London, UK
| | - Sara Santos Franco
- Clinical Unit Cambridge, GlaxoSmithKline, Addenbrooke's Hospital, Cambridge, UK
| | - Andrew Want
- Clinical Unit Cambridge, GlaxoSmithKline, Addenbrooke's Hospital, Cambridge, UK
| | - Misba Beerahee
- Research, GlaxoSmithKline, Medicines Research Centre, Gunnels Wood Road, Stevenage, UK
| | - Disala Fernando
- Clinical Unit Cambridge, GlaxoSmithKline, Addenbrooke's Hospital, Cambridge, UK
| | - Yakshitha Karkera
- Clinical Statistics, Development, GlaxoSmithKline, Prestige Trade Tower, Palace Road, Bangalore, India
| | | | - Thomas Southworth
- Centre for Respiratory Medicine and Allergy, Institute of Inflammation and Repair, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK; Medicines Evaluation Unit, Manchester University NHS Hospital Trust, Manchester, UK
| | - Dave Singh
- Centre for Respiratory Medicine and Allergy, Institute of Inflammation and Repair, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK; Medicines Evaluation Unit, Manchester University NHS Hospital Trust, Manchester, UK
| | | | - Sergey Nejentsev
- Department of Medicine, University of Cambridge, Cambridge, UK; Amsterdam UMC location Vrije Universiteit Amsterdam, Molecular Cell Biology and Immunology, Amsterdam, the Netherlands; Amsterdam Infection and Immunity, Infectious diseases, Amsterdam, the Netherlands
| | - Klaus Okkenhaug
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Alison Condliffe
- Department of Infection, Immunity and Cardiovascular Disease, The Medical School, University of Sheffield, Sheffield, UK
| | - Anita Chandra
- Department of Medicine, University of Cambridge, Cambridge, UK; Department of Clinical Immunology, Addenbrooke's Hospital, Cambridge, UK
| | - Anthony Cahn
- Research, GlaxoSmithKline, Medicines Research Centre, Gunnels Wood Road, Stevenage, UK
| | - Edward Banham Hall
- Clinical Unit Cambridge, GlaxoSmithKline, Addenbrooke's Hospital, Cambridge, UK
| |
Collapse
|
7
|
Yin X, Rang X, Hong X, Zhou Y, Xu C, Fu J. Immune cells transcriptome-based drug repositioning for multiple sclerosis. Front Immunol 2022; 13:1020721. [PMID: 36341423 PMCID: PMC9630342 DOI: 10.3389/fimmu.2022.1020721] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 10/03/2022] [Indexed: 11/13/2022] Open
Abstract
Objective Finding target genes and target pathways of existing drugs for drug repositioning in multiple sclerosis (MS) based on transcriptomic changes in MS immune cells. Materials and Methods Based on transcriptome data from Gene Expression Omnibus (GEO) database, differentially expressed genes (DEGs) in MS patients without treatment were identified by bioinformatics analysis according to the type of immune cells, as well as DEGs in MS patients before and after drug administration. Hub target genes of the drug for MS were analyzed by constructing the protein-protein interaction network, and candidate drugs targeting 2 or more hub target genes were obtained through the connectivity map (CMap) database and Drugbank database. Then, the enriched pathways of MS patients without treatment and the enriched pathways of MS patients before and after drug administration were intersected to obtain the target pathways of the drug for MS, and the candidate drugs targeting 2 or more target pathways were obtained through Kyoto Encyclopedia of Genes and Genomes (KEGG) database. Results We obtained 50 hub target genes for CD4+ T cells in Fingolimod for MS, 15 hub target genes for Plasmacytoid dendritic cells (pDCs) and 7 hub target genes for Peripheral blood mononuclear cells (PBMC) in interferon-β (IFN-β) for MS. 6 candidate drugs targeting two or more hub targets (Fostamatinib, Copper, Artenimol, Phenethyl isothiocyanate, Aspirin and Zinc) were obtained. In addition, we obtained 4 target pathways for CD19+ B cells and 15 target pathways for CD4+ T cells in Fingolimod for MS, 7 target pathways for pDCs and 6 target pathways for PBMC in IFN-β for MS, most of which belong to the immune system and viral infectious disease pathways. We obtained 69 candidate drugs targeting two target pathways. Conclusion We found that applying candidate drugs that target both the “PI3K-Akt signaling pathway” and “Chemokine signaling pathway” (e.g., Nemiralisib and Umbralisib) or applying tyrosine kinase inhibitors (e.g., Fostamatinib) may be potential therapies for the treatment of MS.
Collapse
Affiliation(s)
- Xinyue Yin
- Department of Neurology, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xinming Rang
- Department of Neurology, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiangxiang Hong
- Department of Neurology, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yinglian Zhou
- Department of Neurology, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chaohan Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
- *Correspondence: Jin Fu, ; Chaohan Xu,
| | - Jin Fu
- Department of Neurology, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
- *Correspondence: Jin Fu, ; Chaohan Xu,
| |
Collapse
|
8
|
Diver S, Brightling CE, Greening NJ. Novel Therapeutic Strategies in Asthma-Chronic Obstructive Pulmonary Disease Overlap. Immunol Allergy Clin North Am 2022; 42:671-690. [DOI: 10.1016/j.iac.2022.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
9
|
Cazzola M, Rogliani P, Naviglio S, Calzetta L, Matera MG. An update on the currently available and emerging synthetic pharmacotherapy for uncontrolled asthma. Expert Opin Pharmacother 2022; 23:1205-1216. [PMID: 35621331 DOI: 10.1080/14656566.2022.2083955] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
INTRODUCTION : The treatment of uncontrolled asthma has improved because of triple therapy that includes a long-acting muscarinic antagonist (LAMA) and biological drugs, but several patients are resistant to corticosteroids and/or cannot achieve adequate asthma control using such therapies. AREAS COVERED : Herein, the authors review the current and emerging synthetic pharmacotherapy for uncontrolled asthma to overcome obstacles and limitations of biological therapies. The authors also provide their expert perspectives and opinion on the treatment of uncontrolled asthma. EXPERT OPINION : LAMAs should be added to inhaled corticosteroid/long-acting β2-agonist combinations much earlier than currently recommended by the Global Initiative for Asthma strategy because they can influence the course of small airways disease, reducing lung hyperinflation and improving asthma control. Biological therapies are a major advance in the treatment of severe asthma, but their use is still very limited for several reasons. An alternative to overcome the use of biological therapies is to synthesise compounds that target inflammation-signalling pathways. Several pathways have been identified as potential targets to design either therapeutic or prophylactic drugs against asthma. Some new compounds have already been tested in humans, but results have often been disappointing probably because existing phenotypic and endotypic variants may unpredictably limit the therapeutic value of blocking a specific pathway in most asthmatics, although there may be a substantial benefit for a subgroup of patients.
Collapse
Affiliation(s)
- Mario Cazzola
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Paola Rogliani
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Silvio Naviglio
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Luigino Calzetta
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Maria Gabriella Matera
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| |
Collapse
|
10
|
Zhang J, Jiang H, Lin S, Wu D, Tian H, Jiang L, Cui Y, Jin J, Chen X, Xu H. Design and Optimization of Thienopyrimidine Derivatives as Potent and Selective PI3Kδ Inhibitors for the Treatment of B-Cell Malignancies. J Med Chem 2022; 65:8011-8028. [PMID: 35609190 DOI: 10.1021/acs.jmedchem.2c00530] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Phosphoinositide 3-kinase δ (PI3Kδ) plays a critical role in B lymphocyte (B-cell) development and activation and has been a validated target for the treatment of B-cell malignancies. Herein, we report a series of thienopyrimidine derivatives as novel potent and selective PI3Kδ inhibitors based on a scaffold hopping design strategy. Among them, compound 6 exhibited nanomolar PI3Kδ potency and a favorable selectivity profile compared to other class I PI3K isoforms. In cellular assays, compound 6 showed antiproliferative activity against a panel of B-cell lymphoma cell lines in a low micromolar range, caused cell cycle arrest, and induced apoptosis in Pfeiffer and SU-DHL-6 cells. Further, compound 6 inhibited the activation of mouse B-cells. With support from in vivo pharmacokinetic studies, compound 6 demonstrated significant anticancer efficacy in a Pfeiffer xenograft mouse model. Overall, compound 6 is a promising PI3Kδ inhibitor worthy of further preclinical investigation for the treatment of B-cell malignancies.
Collapse
Affiliation(s)
- Jingbo Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.,Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Huimin Jiang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Songwen Lin
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.,Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Deyu Wu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.,Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Hua Tian
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.,Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Lin Jiang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.,Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yiman Cui
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.,Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Jing Jin
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Xiaoguang Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Heng Xu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.,Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
11
|
Bai Y, Guedes AGP, Krishnan R, Ai X. CD38 plays an age-related role in cholinergic deregulation of airway smooth muscle contractility. J Allergy Clin Immunol 2022; 149:1643-1654.e8. [PMID: 34800431 PMCID: PMC9081122 DOI: 10.1016/j.jaci.2021.10.033] [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: 05/17/2021] [Revised: 10/18/2021] [Accepted: 10/26/2021] [Indexed: 10/19/2022]
Abstract
BACKGROUND Allergen-induced airway hyperresponsiveness in neonatal mice, but not adult mice, is caused by elevated innervation and consequent cholinergic hyperstimulation of airway smooth muscle (ASM). Whether this inflammation-independent mechanism contributes to ASM hypercontraction in childhood asthma warrants investigation. OBJECTIVE We aimed to establish the functional connection between cholinergic stimulation and ASM contractility in different human age groups. METHODS First, we used a neonatal mouse model of asthma to identify age-related mediators of cholinergic deregulation of ASM contractility. Next, we conducted validation and mechanistic studies in primary human ASM cells and precision-cut lung slices from young (<5 years old) and adult (>20 years old) donor lungs. Finally, we evaluated the therapeutic potential of the identified cholinergic signaling mediators using culture models of human ASM hypercontraction. RESULTS ASM hypercontraction due to cholinergic deregulation in early postnatal life requires CD38. Mechanistically, cholinergic signaling activates the phosphatidylinositol 3-kinase/protein kinase B pathway in immature ASM cells to upregulate CD38 levels, thereby augmenting the Ca2+ response to contractile agonists. Strikingly, this early-life, CD38-mediated ASM hypercontraction is not alleviated by the β-agonist formoterol. CONCLUSIONS The acetylcholine-phosphatidylinositol 3-kinase/protein kinase B-CD38 axis is a critical mechanism of airway hyperresponsiveness in early postnatal life. Targeting this axis may provide a tailored treatment for children at high risk for allergic asthma.
Collapse
Affiliation(s)
- Yan Bai
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass; Department of Pediatrics, Division of Neonatology and Newborn Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Mass.
| | - Alonso G P Guedes
- Department of Veterinary Clinical Science, College of Veterinary Medicine, University of Minnesota, St Paul, Minn
| | - Ramaswamy Krishnan
- Department of Emergency Medicine, Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Boston, Mass
| | - Xingbin Ai
- Department of Pediatrics, Division of Neonatology and Newborn Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Mass.
| |
Collapse
|
12
|
Fagone E, Fruciano M, Gili E, Sambataro G, Vancheri C. Developing PI3K Inhibitors for Respiratory Diseases. Curr Top Microbiol Immunol 2022; 436:437-466. [DOI: 10.1007/978-3-031-06566-8_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
13
|
Uddin S, Amour A, Lewis DJ, Edwards CD, Williamson MG, Hall S, Lione LA, Hessel EM. PI3Kδ inhibition prevents IL33, ILC2s and inflammatory eosinophils in persistent airway inflammation. BMC Immunol 2021; 22:78. [PMID: 34920698 PMCID: PMC8684271 DOI: 10.1186/s12865-021-00461-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 09/30/2021] [Indexed: 01/30/2023] Open
Abstract
Background Phosphoinositide-3-kinase-delta (PI3Kδ) inhibition is a promising therapeutic approach for inflammatory conditions due to its role in leucocyte proliferation, migration and activation. However, the effect of PI3Kδ inhibition on group 2 innate lymphoid cells (ILC2s) and inflammatory eosinophils remains unknown. Using a murine model exhibiting persistent airway inflammation we sought to understand the effect of PI3Kδ inhibition, montelukast and anti-IL5 antibody treatment on IL33 expression, group-2-innate lymphoid cells, inflammatory eosinophils, and goblet cell metaplasia. Results Mice were sensitised to house dust mite and after allowing inflammation to resolve, were re-challenged with house dust mite to re-initiate airway inflammation. ILC2s were found to persist in the airways following house dust mite sensitisation and after re-challenge their numbers increased further along with accumulation of inflammatory eosinophils. In contrast to montelukast or anti-IL5 antibody treatment, PI3Kδ inhibition ablated IL33 expression and prevented group-2-innate lymphoid cell accumulation. Only PI3Kδ inhibition and IL5 neutralization reduced the infiltration of inflammatory eosinophils. Moreover, PI3Kδ inhibition reduced goblet cell metaplasia. Conclusions Hence, we show that PI3Kδ inhibition dampens allergic inflammatory responses by ablating key cell types and cytokines involved in T-helper-2-driven inflammatory responses. Supplementary Information The online version contains supplementary material available at 10.1186/s12865-021-00461-5.
Collapse
Affiliation(s)
- Sorif Uddin
- Immunology Research Unit, Respiratory Therapy Area Unit, GSK Medicines Research Centre, GlaxoSmithKline Research and Development Limited, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, UK.
| | - Augustin Amour
- Immunology Research Unit, Respiratory Therapy Area Unit, GSK Medicines Research Centre, GlaxoSmithKline Research and Development Limited, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, UK
| | - David J Lewis
- In Vivo/In Vitro Translation, GlaxoSmithKline Research and Development Limited, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, UK
| | - Chris D Edwards
- In Vivo/In Vitro Translation, GlaxoSmithKline Research and Development Limited, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, UK
| | - Matthew G Williamson
- Immunology Research Unit, Respiratory Therapy Area Unit, GSK Medicines Research Centre, GlaxoSmithKline Research and Development Limited, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, UK
| | - Simon Hall
- Immunology Research Unit, Respiratory Therapy Area Unit, GSK Medicines Research Centre, GlaxoSmithKline Research and Development Limited, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, UK
| | - Lisa A Lione
- Department of Clinical and Pharmaceutical Sciences, School of Life and Medical Sciences, University of Hertfordshire, College Lane, Hatfield, Hertfordshire, AL10 9AB, UK
| | - Edith M Hessel
- Eligo Bioscience, 29 Rue du Faubourg Saint-Jacques, 75014, Paris, France
| |
Collapse
|
14
|
Vanhaesebroeck B, Perry MWD, Brown JR, André F, Okkenhaug K. PI3K inhibitors are finally coming of age. Nat Rev Drug Discov 2021; 20:741-769. [PMID: 34127844 PMCID: PMC9297732 DOI: 10.1038/s41573-021-00209-1] [Citation(s) in RCA: 236] [Impact Index Per Article: 59.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/08/2021] [Indexed: 01/08/2023]
Abstract
Overactive phosphoinositide 3-kinase (PI3K) in cancer and immune dysregulation has spurred extensive efforts to develop therapeutic PI3K inhibitors. Although progress has been hampered by issues such as poor drug tolerance and drug resistance, several PI3K inhibitors have now received regulatory approval - the PI3Kα isoform-selective inhibitor alpelisib for the treatment of breast cancer and inhibitors mainly aimed at the leukocyte-enriched PI3Kδ in B cell malignancies. In addition to targeting cancer cell-intrinsic PI3K activity, emerging evidence highlights the potential of PI3K inhibitors in cancer immunotherapy. This Review summarizes key discoveries that aid the clinical translation of PI3Kα and PI3Kδ inhibitors, highlighting lessons learnt and future opportunities.
Collapse
Affiliation(s)
| | - Matthew W D Perry
- Medicinal Chemistry, Research and Early Development, Respiratory & Immunology BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Jennifer R Brown
- CLL Center, Dana-Farber Cancer Institute, Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Fabrice André
- Institut Gustave Roussy, INSERM U981, Université Paris Saclay, Paris, France
| | - Klaus Okkenhaug
- Department of Pathology, University of Cambridge, Cambridge, UK
| |
Collapse
|
15
|
Baker JR, Donnelly LE. Leukocyte Function in COPD: Clinical Relevance and Potential for Drug Therapy. Int J Chron Obstruct Pulmon Dis 2021; 16:2227-2242. [PMID: 34354348 PMCID: PMC8331105 DOI: 10.2147/copd.s266394] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 07/19/2021] [Indexed: 11/23/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a progressive lung condition affecting 10% of the global population over 45 years. Currently, there are no disease-modifying treatments, with current therapies treating only the symptoms of the disease. COPD is an inflammatory disease, with a high infiltration of leukocytes being found within the lung of COPD patients. These leukocytes, if not kept in check, damage the lung, leading to the pathophysiology associated with the disease. In this review, we focus on the main leukocytes found within the COPD lung, describing how the release of chemokines from the damaged epithelial lining recruits these cells into the lung. Once present, these cells become active and may be driven towards a more pro-inflammatory phenotype. These cells release their own subtypes of inflammatory mediators, growth factors and proteases which can all lead to airway remodeling, mucus hypersecretion and emphysema. Finally, we describe some of the current therapies and potential new targets that could be utilized to target aberrant leukocyte function in the COPD lung. Here, we focus on old therapies such as statins and corticosteroids, but also look at the emerging field of biologics describing those which have been tested in COPD already and potential new monoclonal antibodies which are under review.
Collapse
Affiliation(s)
- Jonathan R Baker
- Airway Disease, National Heart and Lung Institute, Imperial College London, London, UK
| | - Louise E Donnelly
- Airway Disease, National Heart and Lung Institute, Imperial College London, London, UK
| |
Collapse
|
16
|
Sadiq MW, Asimus S, Belvisi MG, Brailsford W, Fransson R, Fuhr R, Hagberg A, Hashemi M, Jellesmark Jensen T, Jonsson J, Keen C, Körnicke T, Kristensson C, Mäenpää J, Necander S, Nemes S, Betts J. Characterisation of pharmacokinetics, safety and tolerability in a first-in-human study for AZD8154, a novel inhaled selective PI3Kγδ dual inhibitor targeting airway inflammatory disease. Br J Clin Pharmacol 2021; 88:260-270. [PMID: 34182611 DOI: 10.1111/bcp.14956] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 05/18/2021] [Accepted: 05/30/2021] [Indexed: 02/04/2023] Open
Abstract
AIMS This 3-part, randomised, phase 1 first-in-human study (NCT03436316) investigated the safety, tolerability and pharmacokinetics (PK) of AZD8154, a dual phosphoinositide 3-kinase (PI3K) γδ inhibitor developed as a novel inhaled anti-inflammatory treatment for respiratory disease. METHODS Healthy men, and women of nonchildbearing potential, were enrolled to receive single and multiple ascending inhaled doses of AZD8154 in parts 1 and 3 of the study, respectively, while part 2 characterised the systemic PK after a single intravenous (IV) dose. In part 1, participants received 0.1-7.7 mg AZD8154 in 6 cohorts. In part 2, participants were given 0.15 mg AZD8154 as an IV infusion. In part 3, AZD8154 was given in 3 cohorts of 0.6, 1.8 and 3.1 mg, with a single dose on Day 1 followed by repeated once-daily doses on Days 4-12. RESULTS In total, 78 volunteers were randomised. All single inhaled, single IV and multiple inhaled doses were shown to be well tolerated without any safety concerns. A population PK model, using nonlinear mixed-effect modelling, was developed to describe the PK of AZD8154. The terminal mean half-life of AZD8154 was 18.0-32.0 hours. The geometric mean of the absolute pulmonary bioavailability of AZD8154 via the inhaled route was 94.1%. CONCLUSION AZD8154 demonstrated an acceptable safety profile, with no reports of serious adverse events and no clinically significant drug-associated safety concerns reported in healthy volunteers. AZD8154 demonstrated prolonged lung retention and a half-life supporting once-daily dosing.
Collapse
Affiliation(s)
- Muhammad Waqas Sadiq
- Clinical Pharmacology & Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Sara Asimus
- Clinical Pharmacology & Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Gaithersburg, MD, Sweden
| | - Maria G Belvisi
- Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden.,Respiratory Pharmacology Group, National Heart & Lung Institute, Imperial College London, London, UK
| | - Wayne Brailsford
- Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Rebecca Fransson
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Rainard Fuhr
- Parexel Early Phase Clinical Unit, Berlin, Germany
| | - Anette Hagberg
- Patient Safety, Respiratory & Immunology, Chief Medical Office, R&D, AstraZeneca, Gothenburg, Sweden
| | - Mahdi Hashemi
- Early Biostats & Statistical Innovation, Data Science & AI, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Tina Jellesmark Jensen
- Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Julia Jonsson
- Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Christina Keen
- Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | | | - Cecilia Kristensson
- Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Jukka Mäenpää
- Patient Safety, Respiratory & Immunology, Chief Medical Office, R&D, AstraZeneca, Gothenburg, Sweden
| | - Sofia Necander
- Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Szilárd Nemes
- Early Biostats & Statistical Innovation, Data Science & AI, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Joanne Betts
- Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
17
|
Perry MWD, Björhall K, Bold P, Brűlls M, Börjesson U, Carlsson J, Chang HFA, Chen Y, Eriksson A, Fihn BM, Fransson R, Fredlund L, Ge H, Huang H, Karabelas K, Lamm Bergström E, Lever S, Lindmark H, Mogemark M, Nikitidis A, Palmgren AP, Pemberton N, Petersen J, Rodrigo Blomqvist M, Smith RW, Thomas MJ, Ullah V, Tyrchan C, Wennberg T, Westin Eriksson A, Yang W, Zhao S, Öster L. Discovery of AZD8154, a Dual PI3Kγδ Inhibitor for the Treatment of Asthma. J Med Chem 2021; 64:8053-8075. [PMID: 34080862 DOI: 10.1021/acs.jmedchem.1c00434] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Starting from our previously described PI3Kγ inhibitors, we describe the exploration of structure-activity relationships that led to the discovery of highly potent dual PI3Kγδ inhibitors. We explored changes in two positions of the molecules, including macrocyclization, but ultimately identified a simpler series with the desired potency profile that had suitable physicochemical properties for inhalation. We were able to demonstrate efficacy in a rat ovalbumin challenge model of allergic asthma and in cells derived from asthmatic patients. The optimized compound, AZD8154, has a long duration of action in the lung and low systemic exposure coupled with high selectivity against off-targets.
Collapse
Affiliation(s)
- Matthew W D Perry
- Medicinal Chemistry, Research and Early Development, Respiratory and Immunology (R&I), BioPharmaceuticals R&D, AstraZeneca, Gothenburg SE-431 83, Sweden
| | - Karin Björhall
- Bioscience COPD/IPF, Research and Early Development, Respiratory and Immunology (R&I), BioPharmaceuticals R&D, AstraZeneca, Gothenburg SE-431 83, Sweden
| | - Peter Bold
- DMPK, Research and Early Development, Respiratory and Immunology (R&I), BioPharmaceuticals R&D, AstraZeneca, Gothenburg SE-431 83, Sweden
| | - Mikael Brűlls
- Early Product Development & Manufacturing, Pharmaceutical Sciences R&D, AstraZeneca, Gothenburg SE-431 83, Sweden
| | - Ulf Börjesson
- Computational Chemistry, Discovery Sciences, R&D, AstraZeneca, Gothenburg SE-431 83, Sweden
| | - Johan Carlsson
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, Gothenburg SE-431 83, Sweden
| | - Hui-Fang Amy Chang
- Medicinal Chemistry, Research and Early Development, Respiratory and Immunology (R&I), BioPharmaceuticals R&D, AstraZeneca, Gothenburg SE-431 83, Sweden
| | - Yunhua Chen
- Pharmaron Beijing Co., Ltd., No. 6 Taihe Road, BDA, Beijing 100176, P.R. China
| | - Anders Eriksson
- Medicinal Chemistry, Research and Early Development, Respiratory and Immunology (R&I), BioPharmaceuticals R&D, AstraZeneca, Gothenburg SE-431 83, Sweden
| | - Britt-Marie Fihn
- DMPK, Research and Early Development, Respiratory and Immunology (R&I), BioPharmaceuticals R&D, AstraZeneca, Gothenburg SE-431 83, Sweden
| | - Rebecca Fransson
- Advanced Drug Delivery, Pharmaceutical Sciences R&D, AstraZeneca, Gothenburg SE-431 83, Sweden
| | - Linda Fredlund
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, Gothenburg SE-431 83, Sweden
| | - Hongbin Ge
- Pharmaron Beijing Co., Ltd., No. 6 Taihe Road, BDA, Beijing 100176, P.R. China
| | - Haijuan Huang
- Pharmaron Beijing Co., Ltd., No. 6 Taihe Road, BDA, Beijing 100176, P.R. China
| | - Kostas Karabelas
- Projects, Research and Early Development, Respiratory and Immunology (R&I), BioPharmaceuticals R&D, AstraZeneca, Gothenburg SE-431 83, Sweden
| | - Eva Lamm Bergström
- DMPK, Research and Early Development, Respiratory and Immunology (R&I), BioPharmaceuticals R&D, AstraZeneca, Gothenburg SE-431 83, Sweden
| | - Sarah Lever
- Medicinal Chemistry, Research and Early Development, Respiratory and Immunology (R&I), BioPharmaceuticals R&D, AstraZeneca, Gothenburg SE-431 83, Sweden
| | - Helena Lindmark
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, Gothenburg SE-431 83, Sweden
| | - Mickael Mogemark
- Medicinal Chemistry, Research and Early Development, Respiratory and Immunology (R&I), BioPharmaceuticals R&D, AstraZeneca, Gothenburg SE-431 83, Sweden
| | - Antonios Nikitidis
- Medicinal Chemistry, Research and Early Development, Respiratory and Immunology (R&I), BioPharmaceuticals R&D, AstraZeneca, Gothenburg SE-431 83, Sweden
| | - Anna-Pia Palmgren
- Medicinal Chemistry, Research and Early Development, Respiratory and Immunology (R&I), BioPharmaceuticals R&D, AstraZeneca, Gothenburg SE-431 83, Sweden
| | - Nils Pemberton
- Medicinal Chemistry, Research and Early Development, Respiratory and Immunology (R&I), BioPharmaceuticals R&D, AstraZeneca, Gothenburg SE-431 83, Sweden
| | - Jens Petersen
- Structure & Biophysics, Discovery Sciences, R&D, AstraZeneca, Gothenburg SE-431 83, Sweden
| | - Mio Rodrigo Blomqvist
- Bioscience Cough and In Vivo, Research and Early Development, Respiratory and Immunology (R&I), BioPharmaceuticals R&D, AstraZeneca, Gothenburg SE-431 83, Sweden
| | - Reed W Smith
- Medicinal Chemistry, Research and Early Development, Respiratory and Immunology (R&I), BioPharmaceuticals R&D, AstraZeneca, Gothenburg SE-431 83, Sweden
| | - Matthew J Thomas
- Bioscience COPD/IPF, Research and Early Development, Respiratory and Immunology (R&I), BioPharmaceuticals R&D, AstraZeneca, Gothenburg SE-431 83, Sweden
| | - Victoria Ullah
- Medicinal Chemistry, Research and Early Development, Respiratory and Immunology (R&I), BioPharmaceuticals R&D, AstraZeneca, Gothenburg SE-431 83, Sweden
| | - Christian Tyrchan
- Medicinal Chemistry, Research and Early Development, Respiratory and Immunology (R&I), BioPharmaceuticals R&D, AstraZeneca, Gothenburg SE-431 83, Sweden
| | - Tiiu Wennberg
- Bioscience COPD/IPF, Research and Early Development, Respiratory and Immunology (R&I), BioPharmaceuticals R&D, AstraZeneca, Gothenburg SE-431 83, Sweden
| | - Annika Westin Eriksson
- Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gothenburg SE-431 83, Sweden
| | - Wenzhen Yang
- Pharmaron Beijing Co., Ltd., No. 6 Taihe Road, BDA, Beijing 100176, P.R. China
| | - Shuchun Zhao
- Pharmaron Beijing Co., Ltd., No. 6 Taihe Road, BDA, Beijing 100176, P.R. China
| | - Linda Öster
- Structure & Biophysics, Discovery Sciences, R&D, AstraZeneca, Gothenburg SE-431 83, Sweden
| |
Collapse
|
18
|
Ricciardolo FL, Bertolini F, Carriero V, Sprio AE. Asthma phenotypes and endotypes: a systematic review. Minerva Med 2021; 112:547-563. [PMID: 33969960 DOI: 10.23736/s0026-4806.21.07498-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
INTRODUCTION Asthma is a complex disorder characterized by expiratory airflow limitation, wheeze, shortness of breath, chest tightness and cough, which can vary over time and in intensity. Being highly heterogeneous, asthma was characterized and classified in several asthma phenotypes and endotypes from 1947 until today. The present systematic review aims to summarize and describe evidence that was published in the last ten years in the field of asthma phenotyping and endotyping. EVIDENCE ACQUISITION The systematic review resumed high-quality evidence (clinical trials and randomized control trials) retrieved on MEDLINE and EMBASE databanks and involving adult asthmatic populations. Analyses of literature were conducted according to PRISMA and CASP guidelines. EVIDENCE SYNTHESIS Querying MEDLINE and EMBASE databanks, 5019 and 12261 entries were retrieved, respectively. Applying limitations for year of publication, age of participants, and type of publication, the search results were reduced to 98 and 132 articles, respectively. After data abstraction and resolution of duplications, only 50 articles were further evaluated. The research products were then classified first in macro-areas of interest (phenotypes or endotypes) and then in detailed micro-areas. CONCLUSIONS This systematic review overviews the principal findings available from high-quality literature in the last decade concerning asthma phenotypes and endotypes. Asthma has been described from different points of view, characterizing symptoms, microbiota composition, comorbidities, viral infections, and airway and/or systemic inflammatory status. The comprehension of precise mechanisms underlying asthma pathogenesis is thereby the basis for the development of novel therapeutic strategies, likely essential to the development of precision medicine.
Collapse
Affiliation(s)
- Fabio L Ricciardolo
- Department of Clinical and Biological Sciences, Rare Lung Disease Unit and Severe Asthma Centre, San Luigi Gonzaga University Hospital, University of Turin, Turin, Italy -
| | - Francesca Bertolini
- Department of Clinical and Biological Sciences, Rare Lung Disease Unit and Severe Asthma Centre, San Luigi Gonzaga University Hospital, University of Turin, Turin, Italy
| | - Vitina Carriero
- Department of Clinical and Biological Sciences, Rare Lung Disease Unit and Severe Asthma Centre, San Luigi Gonzaga University Hospital, University of Turin, Turin, Italy
| | - Andrea E Sprio
- Department of Clinical and Biological Sciences, Rare Lung Disease Unit and Severe Asthma Centre, San Luigi Gonzaga University Hospital, University of Turin, Turin, Italy.,Department of Research, ASOMI College of Sciences, Marsa, Malta
| |
Collapse
|
19
|
Kyriakopoulos C, Gogali A, Bartziokas K, Kostikas K. Identification and treatment of T2-low asthma in the era of biologics. ERJ Open Res 2021; 7:00309-2020. [PMID: 34109244 PMCID: PMC8181790 DOI: 10.1183/23120541.00309-2020] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 09/08/2020] [Indexed: 12/25/2022] Open
Abstract
Currently, and based on the development of relevant biologic therapies, T2-high is the most well-defined endotype of asthma. Although much progress has been made in elucidating T2-high inflammation pathways, no specific clinically applicable biomarkers for T2-low asthma have been identified. The therapeutic approach of T2-low asthma is a problem urgently needing resolution, firstly because these patients have poor response to steroids, and secondly because they are not candidates for the newer targeted biologic agents. Thus, there is an unmet need for the identification of biomarkers that can help the diagnosis and endotyping of T2-low asthma. Ongoing investigation is focusing on neutrophilic airway inflammation mediators as therapeutic targets, including interleukin (IL)-8, IL-17, IL-1, IL-6, IL-23 and tumour necrosis factor-α; molecules that target restoration of corticosteroid sensitivity, mainly mitogen-activated protein kinase inhibitors, tyrosine kinase inhibitors and phosphatidylinositol 3-kinase inhibitors; phosphodiesterase (PDE)3 inhibitors that act as bronchodilators and PDE4 inhibitors that have an anti-inflammatory effect; and airway smooth muscle mass attenuation therapies, mainly for patients with paucigranulocytic inflammation. This article aims to review the evidence for noneosinophilic inflammation being a target for therapy in asthma; discuss current and potential future therapeutic approaches, such as novel molecules and biologic agents; and assess clinical trials of licensed drugs in the treatment of T2-low asthma.
Collapse
Affiliation(s)
- Chris Kyriakopoulos
- Respiratory Medicine Dept, University of Ioannina School of Medicine, Ioannina, Greece
| | - Athena Gogali
- Respiratory Medicine Dept, University of Ioannina School of Medicine, Ioannina, Greece
| | | | - Konstantinos Kostikas
- Respiratory Medicine Dept, University of Ioannina School of Medicine, Ioannina, Greece
| |
Collapse
|
20
|
Crisford H, Sapey E, Rogers GB, Taylor S, Nagakumar P, Lokwani R, Simpson JL. Neutrophils in asthma: the good, the bad and the bacteria. Thorax 2021; 76:thoraxjnl-2020-215986. [PMID: 33632765 PMCID: PMC8311087 DOI: 10.1136/thoraxjnl-2020-215986] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 01/12/2021] [Accepted: 01/18/2021] [Indexed: 12/30/2022]
Abstract
Airway inflammation plays a key role in asthma pathogenesis but is heterogeneous in nature. There has been significant scientific discovery with regard to type 2-driven, eosinophil-dominated asthma, with effective therapies ranging from inhaled corticosteroids to novel biologics. However, studies suggest that approximately 1 in 5 adults with asthma have an increased proportion of neutrophils in their airways. These patients tend to be older, have potentially pathogenic airway bacteria and do not respond well to classical therapies. Currently, there are no specific therapeutic options for these patients, such as neutrophil-targeting biologics.Neutrophils comprise 70% of the total circulatory white cells and play a critical defence role during inflammatory and infective challenges. This makes them a problematic target for therapeutics. Furthermore, neutrophil functions change with age, with reduced microbial killing, increased reactive oxygen species release and reduced production of extracellular traps with advancing age. Therefore, different therapeutic strategies may be required for different age groups of patients.The pathogenesis of neutrophil-dominated airway inflammation in adults with asthma may reflect a counterproductive response to the defective neutrophil microbial killing seen with age, resulting in bystander damage to host airway cells and subsequent mucus hypersecretion and airway remodelling. However, in children with asthma, neutrophils are less associated with adverse features of disease, and it is possible that in children, neutrophils are less pathogenic.In this review, we explore the mechanisms of neutrophil recruitment, changes in cellular function across the life course and the implications this may have for asthma management now and in the future. We also describe the prevalence of neutrophilic asthma globally, with a focus on First Nations people of Australia, New Zealand and North America.
Collapse
Affiliation(s)
- Helena Crisford
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Elizabeth Sapey
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Geraint B Rogers
- SAHMRI Microbiome Research Laboratory, Flinders University College of Medicine and Public Health, Adelaide, South Australia, Australia
- Microbiome and Host Health, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Steven Taylor
- SAHMRI Microbiome Research Laboratory, Flinders University College of Medicine and Public Health, Adelaide, South Australia, Australia
- Microbiome and Host Health, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Prasad Nagakumar
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- Respiratory Medicine, Birmingham Women's and Children's NHS Foundation Trust, Birmingham, UK
| | - Ravi Lokwani
- Faculty of Health and Medicine, Priority Research Centre for Healthy Lungs, The University of Newcastle, Callaghan, New South Wales, Australia
| | - Jodie L Simpson
- Faculty of Health and Medicine, Priority Research Centre for Healthy Lungs, The University of Newcastle, Callaghan, New South Wales, Australia
| |
Collapse
|
21
|
Agache I, Akdis CA, Akdis M, Canonica GW, Casale T, Chivato T, Corren J, Chu DK, Del Giacco S, Eiwegger T, Flood B, Firinu D, Gern JE, Hamelmann E, Hanania N, Hernández‐Martín I, Knibb R, Mäkelä M, Nair P, O’Mahony L, Papadopoulos NG, Papi A, Park H, Pérez de Llano L, Pfaar O, Quirce S, Sastre J, Shamji M, Schwarze J, Palomares O, Jutel M. EAACI Biologicals Guidelines-Recommendations for severe asthma. Allergy 2021; 76:14-44. [PMID: 32484954 DOI: 10.1111/all.14425] [Citation(s) in RCA: 135] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 05/19/2020] [Indexed: 12/20/2022]
Abstract
Severe asthma imposes a significant burden on patients, families and healthcare systems. Management is difficult, due to disease heterogeneity, co-morbidities, complexity in care pathways and differences between national or regional healthcare systems. Better understanding of the mechanisms has enabled a stratified approach to the management of severe asthma, supporting the use of targeted treatments with biologicals. However, there are still many issues that require further clarification. These include selection of a certain biological (as they all target overlapping disease phenotypes), the definition of response, strategies to enhance the responder rate, the duration of treatment and its regimen (in the clinic or home-based) and its cost-effectiveness. The EAACI Guidelines on the use of biologicals in severe asthma follow the GRADE approach in formulating recommendations for each biological and each outcome. In addition, a management algorithm for the use of biologicals in the clinic is proposed, together with future approaches and research priorities.
Collapse
Affiliation(s)
- Ioana Agache
- Faculty of Medicine Transylvania University Brasov Romania
| | - Cezmi A. 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
| | - Mubeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
| | - Giorgio Walter Canonica
- Personalized Medicine, Asthma and Allergy Humanitas Clinical and Research Center IRCCS Rozzano Italy
| | - Thomas Casale
- Division of Allergy and Immunology University of South Florida Morsani College of Medicine Tampa FL USA
| | - Tomas Chivato
- School of Medicine University CEU San Pablo Madrid Spain
| | | | - Derek K. Chu
- Department of Health Research Methods, Evidence and Impact Division of Immunology and Allergy, and Department of Medicine McMaster University Hamilton ON Canada
| | - Stefano Del Giacco
- Department of Medical Sciences and Public Health University of Cagliari Cagliari Italy
| | - Thomas Eiwegger
- Translational Medicine Program, Research Institute Hospital for Sick Children Toronto ON Canada
- Department of Immunology University of Toronto Toronto ON Canada
- Division of Immunology and Allergy Food Allergy and Anaphylaxis Program The Hospital for Sick Children Departments of Paediatrics and Immunology University of Toronto Toronto ON Canada
| | - Breda Flood
- European Federation of Allergy and Airway Diseases Brussels Belgium
| | - Davide Firinu
- Department of Medical Sciences and Public Health University of Cagliari Cagliari Italy
| | - James E. Gern
- Department of Pediatrics School of Medicine and Public Health University of Wisconsin Madison WI USA
| | - Eckard Hamelmann
- Children’s Center Bethel Evangelical Hospital Bethel University of Bielefeld Bielefeld Germany
| | - Nicola Hanania
- Section of Pulmonary, Critical Care and Sleep Medicine Baylor College of Medicine Houston TX USA
| | | | - Rebeca Knibb
- Department of Psychology School of Life and Health Sciences Aston University Birmingham UK
| | - Mika Mäkelä
- Skin and Allergy Hospital Helsinki University Hospital and University of Helsinki Helsinki Finland
| | - Parameswaran Nair
- Division of Respirology Department of Medicine McMaster University Hamilton ON Canada
- Firestone Institute for Respiratory Health St Joseph's Healthcare Hamilton ON Canada
| | - Liam O’Mahony
- Departments of Medicine and Microbiology APC Microbiome Ireland University College Cork Cork Ireland
| | - Nikolaos G. Papadopoulos
- Division of Infection, Immunity and Respiratory Medicine University of Manchester Manchester UK
- Allergy Department 2nd Pediatric Clinic National Kapodistrian University of Athens Athens Greece
| | - Alberto Papi
- Research Center on Asthma and COPD Department of Medical Sciences University of Ferrara Ferrara Italy
| | - Hae‐Sim Park
- Department of Allergy and Clinical Immunology Ajou University Ajou Korea
| | | | - Oliver Pfaar
- Department of Otorhinolaryngology, Head and Neck Surgery Section of Rhinology and Allergy University Hospital Marburg Philipps‐Universität Marburg Marburg Germany
| | - Santiago Quirce
- Department of Allergy La Paz University Hospital IdiPAZ CIBER of Respiratory Diseases (CIBERES) Universidad Autónoma de Madrid Madrid Spain
| | - Joaquin Sastre
- Facultad de Medicina Universidad Autónoma de Madrid Madrid Spain
| | - Mohamed Shamji
- Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Inflammation, Repair, Development National Heart and Lung Institute London UK
- Imperial College NIHR Biomedical Research Centre Asthma UK Centre in Allergic Mechanisms of Asthma London UK
| | - Jurgen Schwarze
- Centre for Inflammation Research, Child Life and Health The University of Edinburgh Edinburgh UK
| | - Oscar Palomares
- Department of Biochemistry and Molecular Biology Chemistry School Complutense University of Madrid Madrid Spain
| | - Marek Jutel
- Department of Clinical Immunology Wroclaw Medical University Wroclaw Poland
- All‐MED Medical Research Institute Wroclaw Poland
| |
Collapse
|
22
|
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
|
23
|
Palma G, Pasqua T, Silvestri G, Rocca C, Gualtieri P, Barbieri A, De Bartolo A, De Lorenzo A, Angelone T, Avolio E, Botti G. PI3Kδ Inhibition as a Potential Therapeutic Target in COVID-19. Front Immunol 2020; 11:2094. [PMID: 32973818 PMCID: PMC7472874 DOI: 10.3389/fimmu.2020.02094] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 08/03/2020] [Indexed: 01/08/2023] Open
Abstract
The spread of the novel human respiratory coronavirus (SARS-CoV-2) is a global public health emergency. There is no known successful treatment as of this time, and there is a need for medical options to mitigate this current epidemic. SARS-CoV-2 uses the angiotensin-converting enzyme 2 (ACE2) receptor and is primarily trophic for the lower and upper respiratory tract. A number of current studies on COVID-19 have demonstrated the substantial increase in pro-inflammatory factors in the lungs during infection. The virus is also documented in the central nervous system and, particularly in the brainstem, which plays a key role in respiratory and cardiovascular function. Currently, there are few antiviral approaches, and several alternative drugs are under investigation. Two of these are Idelalisib and Ebastine, already proposed as preventive strategies in airways and allergic diseases. The interesting and evolving potential of phosphoinositide 3-kinase δ (PI3Kδ) inhibitors, together with Ebastine, lies in their ability to suppress the release of pro-inflammatory cytokines, such as IL-1β, IL-8, IL-6, and TNF-α, by T cells. This may represent an optional therapeutic choice for COVID-19 to reduce inflammatory reactions and mortality, enabling patients to recover faster. This concise communication aims to provide new potential therapeutic targets capable of mitigating and alleviating SARS-CoV-2 pandemic infection.
Collapse
Affiliation(s)
- Giuseppe Palma
- SSD Sperimentazione Animale, Istituto Nazionale Tumori Fondazione G. Pascale – IRCSS, Naples, Italy
| | - Teresa Pasqua
- Laboratory of Cellular and Molecular Cardiovascular Patho-Physiology, Department of Biology, Ecology and Earth Science, University of Calabria, Rende, Italy
| | - Giovannino Silvestri
- Institute of Human Virology, Division of Infectious Agents and Cancer, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Carmine Rocca
- Laboratory of Cellular and Molecular Cardiovascular Patho-Physiology, Department of Biology, Ecology and Earth Science, University of Calabria, Rende, Italy
| | - Paola Gualtieri
- School of Specialization in Food Science, University of Rome “Tor Vergata”, Rome, Italy
| | - Antonio Barbieri
- SSD Sperimentazione Animale, Istituto Nazionale Tumori Fondazione G. Pascale – IRCSS, Naples, Italy
| | - Anna De Bartolo
- Laboratory of Cellular and Molecular Cardiovascular Patho-Physiology, Department of Biology, Ecology and Earth Science, University of Calabria, Rende, Italy
| | - Antonino De Lorenzo
- Section of Clinical Nutrition and Nutrigenomics, Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, Rome, Italy
| | - Tommaso Angelone
- Laboratory of Cellular and Molecular Cardiovascular Patho-Physiology, Department of Biology, Ecology and Earth Science, University of Calabria, Rende, Italy
- National Institute for Cardiovascular Research (INRC), Bologna, Italy
| | - Ennio Avolio
- School of Specialization in Food Science, University of Rome “Tor Vergata”, Rome, Italy
- Section of Clinical Nutrition and Nutrigenomics, Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, Rome, Italy
| | - Gerardo Botti
- Scientific Director, Istituto Nazionale Tumori Fondazione G. Pascale – IRCSS, Naples, Italy
| |
Collapse
|
24
|
Zhabyeyev P, Chen X, Vanhaesebroeck B, Oudit GY. PI3Kα in cardioprotection: Cytoskeleton, late Na + current, and mechanism of arrhythmias. Channels (Austin) 2020; 13:520-532. [PMID: 31790629 PMCID: PMC6930018 DOI: 10.1080/19336950.2019.1697127] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
PI 3-kinase α (PI3Kα) is a lipid kinase that converts phosphatidylinositol-4,5-bisphosphate (PIP2) to phosphatidylinositol-3,4,5-triphosphate (PIP3). PI3Kα regulates a variety of cellular processes such as nutrient sensing, cell cycle, migration, and others. Heightened activity of PI3Kα in many types of cancer made it a prime oncology drug target, but also raises concerns of possible adverse effects on the heart. Indeed, recent advances in preclinical models demonstrate an important role of PI3Kα in the control of cytoskeletal integrity, Na+ channel activity, cardioprotection, and prevention of arrhythmias.
Collapse
Affiliation(s)
- Pavel Zhabyeyev
- Department of Medicine, University of Alberta, Edmonton, Canada.,Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
| | - Xueyi Chen
- Department of Medicine, University of Alberta, Edmonton, Canada.,Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
| | | | - Gavin Y Oudit
- Department of Medicine, University of Alberta, Edmonton, Canada.,Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
| |
Collapse
|
25
|
Nunes-Santos CJ, Uzel G, Rosenzweig SD. PI3K pathway defects leading to immunodeficiency and immune dysregulation. J Allergy Clin Immunol 2020; 143:1676-1687. [PMID: 31060715 DOI: 10.1016/j.jaci.2019.03.017] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 03/26/2019] [Accepted: 03/26/2019] [Indexed: 12/16/2022]
Abstract
The phosphatidylinositol 3-kinase (PI3K) signaling pathway is involved in a broad range of cellular processes, including growth, metabolism, differentiation, proliferation, motility, and survival. The PI3Kδ enzyme complex is primarily present in the immune system and comprises a catalytic (p110δ) and regulatory (p85α) subunit. Dynamic regulation of PI3Kδ activity is required to ensure normal function and differentiation of immune cells. In the last decade, discovery of germline mutations in genes involved in the PI3Kδ pathway (PIK3CD, PIK3R1, or phosphatase and tensin homolog [PTEN]) proved that both overactivation and underactivation (gain of function and loss of function, respectively) of PI3Kδ lead to impaired and dysregulated immunity. Although a small group of patients reported to underactivate PI3Kδ show predominantly humoral defects and autoimmune features, more than 200 patients have been described with overactivation of PI3Kδ, presenting with a much more complex phenotype of combined immunodeficiency and immune dysregulation. The clinical and immunologic characterization, as well as current pathophysiologic understanding and specific therapies for PI3K pathway defects leading to immunodeficiency and immune dysregulation, are reviewed here.
Collapse
Affiliation(s)
- Cristiane J Nunes-Santos
- Immunology Service, Department of Laboratory Medicine, National Institutes of Health (NIH) Clinical Center, Bethesda, Md; Faculdade de Medicina, Instituto da Crianca, Universidade de São Paulo, São Paulo, Brazil
| | - Gulbu Uzel
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Md
| | - Sergio D Rosenzweig
- Immunology Service, Department of Laboratory Medicine, National Institutes of Health (NIH) Clinical Center, Bethesda, Md.
| |
Collapse
|
26
|
Structurally novel PI3Kδ/γ dual inhibitors characterized by a seven-membered spirocyclic spacer: The SARs investigation and PK evaluation. Eur J Med Chem 2020; 191:112143. [DOI: 10.1016/j.ejmech.2020.112143] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 12/29/2019] [Accepted: 02/11/2020] [Indexed: 12/30/2022]
|
27
|
Abstract
Current management of severe asthma relying either on guidelines (bulk approach) or on disease phenotypes (stratified approach) did not improve the burden of the disease. Several severe phenotypes are described: clinical, functional, morphological, inflammatory, molecular and microbiome-related. However, phenotypes do not necessarily relate to or give insights into the underlying pathogenetic mechanisms which are described by the disease endotypes. Based on the major immune-inflammatory pathway involved type-2 high, type-2 low and mixed endotypes are described for severe asthma, with several shared pathogenetic pathways such as genetic and epigenetic, metabolic, neurogenic and remodelling subtypes. The concept of multidimensional endotyping as un unbiased approach to severe asthma is discussed, together with new tools and targets facilitating the shift from the stratified to the precision medicine approach.
Collapse
|
28
|
Design of selective PI3Kδ inhibitors using an iterative scaffold-hopping workflow. Bioorg Med Chem Lett 2019; 29:2575-2580. [PMID: 31416665 DOI: 10.1016/j.bmcl.2019.08.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 08/01/2019] [Accepted: 08/02/2019] [Indexed: 11/21/2022]
Abstract
PI3Kδ mediates key immune cell signaling pathways and is a target of interest for multiple indications in immunology and oncology. Here we report a structure-based scaffold-hopping strategy for the design of chemically diverse PI3Kδ inhibitors. Using this strategy, we identified several scaffolds that can be combined to generate new PI3Kδ inhibitors with high potency and isoform selectivity. In particular, an oxindole-based scaffold was found to impart exquisite selectivity when combined with several hinge binding motifs.
Collapse
|
29
|
Begg M, Edwards CD, Hamblin JN, Pefani E, Wilson R, Gilbert J, Vitulli G, Mallett D, Morrell J, Hingle MI, Uddin S, Ehtesham F, Marotti M, Harrell A, Newman CF, Fernando D, Clark J, Cahn A, Hessel EM. Translation of Inhaled Drug Optimization Strategies into Clinical Pharmacokinetics and Pharmacodynamics Using GSK2292767A, a Novel Inhaled Phosphoinositide 3-Kinase δ Inhibitor. J Pharmacol Exp Ther 2019; 369:443-453. [PMID: 30940692 DOI: 10.1124/jpet.119.257311] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 03/27/2019] [Indexed: 11/22/2022] Open
Abstract
This study describes the pharmacokinetic (PK) and pharmaco-dynamic (PD) profile of N-(5-(4-(5-(((2R,6S)-2,6-dimethylmorpholino)methyl)oxazol-2-yl)-1H-indazol-6-yl)-2-methoxypyridin-3-yl)methanesulfonamide (GSK2292767A), a novel low-solubility inhaled phosphoinositide 3-kinase delta (PI3Kδ) inhibitor developed as an alternative to 2-(6-(1H-indol-4-yl)-1H-indazol-4-yl)-5-((4-isopropylpiperazin-1-yl)methyl)oxazole (nemiralisib), which is a highly soluble inhaled inhibitor of PI3Kδ with a lung profile consistent with once-daily dosing. GSK2292767A has a similar in vitro cellular profile to nemiralisib and reduces eosinophilia in a murine PD model by 63% (n = 5, P < 0.05). To explore whether a low-soluble compound results in effective PI3Kδ inhibition in humans, a first time in human study was conducted with GSK2292767A in healthy volunteers who smoke. GSK2292767A was generally well tolerated, with headache being the most common reported adverse event. PD changes in induced sputum were measured in combination with drug concentrations in plasma from single (0.05-2 mg, n = 37), and 14-day repeat (2 mg, n = 12) doses of GSK2292767A. Trough bronchoalveolar lavage (BAL) for PK was taken after 14 days of repeat dosing. GSK2292767A displayed a linear increase in plasma exposure with dose, with marginal accumulation after 14 days. Induced sputum showed a 27% (90% confidence interval 15%, 37%) reduction in phosphatidylinositol-trisphosphate (the product of phosphoinositide 3-kinase activation) 3 hours after a single dose. Reduction was not maintained 24 hours after single or repeat dosing. BAL analysis confirmed the presence of GSK2292767A in lung at 24 hours, consistent with the preclinical lung retention profile. Despite good lung retention, target engagement was only present at 3 hours. This exposure-response disconnect is an important observation for future inhaled drug design strategies considering low solubility to drive lung retention.
Collapse
Affiliation(s)
- Malcolm Begg
- Refractory Respiratory Inflammation Discovery Performance Unit, GlaxoSmithKline, Stevenage, United Kingdom (M.B., C.D.E., J.N.H., E.M.H.); Clinical Pharmacology & Model Stimulation, (E.P., R.W.), Drug Metabolism & Pharmacokinetics, (G.V., D.M., J.M.), Refractory Respiratory Inflammation DPU, (S.U.), In Vitro In Vivo Translation, (C.F.N.), Discovery Medicine, (A.C) GlaxoSmithKline, Stevenage, United Kingdom; Global Clinical Science & Delivery, GlaxoSmithKline, Stockley Park, Uxbridge, United Kingdom (M.M., J.G.); In Vitro In Vivo Translation, (A.H.), Drug Product Design & Development, (M.I.H.), GlaxoSmithKline, Ware, United Kingdom (M.I.H.); Department of Biological Chemistry, Babraham Institute, Cambridge, United Kingdom (J.C.); and Clinical Unit Cambridge, Addenbrookes Hospital, Cambridge, United Kingdom (F.E., D.F.)
| | - Chris D Edwards
- Refractory Respiratory Inflammation Discovery Performance Unit, GlaxoSmithKline, Stevenage, United Kingdom (M.B., C.D.E., J.N.H., E.M.H.); Clinical Pharmacology & Model Stimulation, (E.P., R.W.), Drug Metabolism & Pharmacokinetics, (G.V., D.M., J.M.), Refractory Respiratory Inflammation DPU, (S.U.), In Vitro In Vivo Translation, (C.F.N.), Discovery Medicine, (A.C) GlaxoSmithKline, Stevenage, United Kingdom; Global Clinical Science & Delivery, GlaxoSmithKline, Stockley Park, Uxbridge, United Kingdom (M.M., J.G.); In Vitro In Vivo Translation, (A.H.), Drug Product Design & Development, (M.I.H.), GlaxoSmithKline, Ware, United Kingdom (M.I.H.); Department of Biological Chemistry, Babraham Institute, Cambridge, United Kingdom (J.C.); and Clinical Unit Cambridge, Addenbrookes Hospital, Cambridge, United Kingdom (F.E., D.F.)
| | - J Nicole Hamblin
- Refractory Respiratory Inflammation Discovery Performance Unit, GlaxoSmithKline, Stevenage, United Kingdom (M.B., C.D.E., J.N.H., E.M.H.); Clinical Pharmacology & Model Stimulation, (E.P., R.W.), Drug Metabolism & Pharmacokinetics, (G.V., D.M., J.M.), Refractory Respiratory Inflammation DPU, (S.U.), In Vitro In Vivo Translation, (C.F.N.), Discovery Medicine, (A.C) GlaxoSmithKline, Stevenage, United Kingdom; Global Clinical Science & Delivery, GlaxoSmithKline, Stockley Park, Uxbridge, United Kingdom (M.M., J.G.); In Vitro In Vivo Translation, (A.H.), Drug Product Design & Development, (M.I.H.), GlaxoSmithKline, Ware, United Kingdom (M.I.H.); Department of Biological Chemistry, Babraham Institute, Cambridge, United Kingdom (J.C.); and Clinical Unit Cambridge, Addenbrookes Hospital, Cambridge, United Kingdom (F.E., D.F.)
| | - Eleni Pefani
- Refractory Respiratory Inflammation Discovery Performance Unit, GlaxoSmithKline, Stevenage, United Kingdom (M.B., C.D.E., J.N.H., E.M.H.); Clinical Pharmacology & Model Stimulation, (E.P., R.W.), Drug Metabolism & Pharmacokinetics, (G.V., D.M., J.M.), Refractory Respiratory Inflammation DPU, (S.U.), In Vitro In Vivo Translation, (C.F.N.), Discovery Medicine, (A.C) GlaxoSmithKline, Stevenage, United Kingdom; Global Clinical Science & Delivery, GlaxoSmithKline, Stockley Park, Uxbridge, United Kingdom (M.M., J.G.); In Vitro In Vivo Translation, (A.H.), Drug Product Design & Development, (M.I.H.), GlaxoSmithKline, Ware, United Kingdom (M.I.H.); Department of Biological Chemistry, Babraham Institute, Cambridge, United Kingdom (J.C.); and Clinical Unit Cambridge, Addenbrookes Hospital, Cambridge, United Kingdom (F.E., D.F.)
| | - Robert Wilson
- Refractory Respiratory Inflammation Discovery Performance Unit, GlaxoSmithKline, Stevenage, United Kingdom (M.B., C.D.E., J.N.H., E.M.H.); Clinical Pharmacology & Model Stimulation, (E.P., R.W.), Drug Metabolism & Pharmacokinetics, (G.V., D.M., J.M.), Refractory Respiratory Inflammation DPU, (S.U.), In Vitro In Vivo Translation, (C.F.N.), Discovery Medicine, (A.C) GlaxoSmithKline, Stevenage, United Kingdom; Global Clinical Science & Delivery, GlaxoSmithKline, Stockley Park, Uxbridge, United Kingdom (M.M., J.G.); In Vitro In Vivo Translation, (A.H.), Drug Product Design & Development, (M.I.H.), GlaxoSmithKline, Ware, United Kingdom (M.I.H.); Department of Biological Chemistry, Babraham Institute, Cambridge, United Kingdom (J.C.); and Clinical Unit Cambridge, Addenbrookes Hospital, Cambridge, United Kingdom (F.E., D.F.)
| | - Jane Gilbert
- Refractory Respiratory Inflammation Discovery Performance Unit, GlaxoSmithKline, Stevenage, United Kingdom (M.B., C.D.E., J.N.H., E.M.H.); Clinical Pharmacology & Model Stimulation, (E.P., R.W.), Drug Metabolism & Pharmacokinetics, (G.V., D.M., J.M.), Refractory Respiratory Inflammation DPU, (S.U.), In Vitro In Vivo Translation, (C.F.N.), Discovery Medicine, (A.C) GlaxoSmithKline, Stevenage, United Kingdom; Global Clinical Science & Delivery, GlaxoSmithKline, Stockley Park, Uxbridge, United Kingdom (M.M., J.G.); In Vitro In Vivo Translation, (A.H.), Drug Product Design & Development, (M.I.H.), GlaxoSmithKline, Ware, United Kingdom (M.I.H.); Department of Biological Chemistry, Babraham Institute, Cambridge, United Kingdom (J.C.); and Clinical Unit Cambridge, Addenbrookes Hospital, Cambridge, United Kingdom (F.E., D.F.)
| | - Giovanni Vitulli
- Refractory Respiratory Inflammation Discovery Performance Unit, GlaxoSmithKline, Stevenage, United Kingdom (M.B., C.D.E., J.N.H., E.M.H.); Clinical Pharmacology & Model Stimulation, (E.P., R.W.), Drug Metabolism & Pharmacokinetics, (G.V., D.M., J.M.), Refractory Respiratory Inflammation DPU, (S.U.), In Vitro In Vivo Translation, (C.F.N.), Discovery Medicine, (A.C) GlaxoSmithKline, Stevenage, United Kingdom; Global Clinical Science & Delivery, GlaxoSmithKline, Stockley Park, Uxbridge, United Kingdom (M.M., J.G.); In Vitro In Vivo Translation, (A.H.), Drug Product Design & Development, (M.I.H.), GlaxoSmithKline, Ware, United Kingdom (M.I.H.); Department of Biological Chemistry, Babraham Institute, Cambridge, United Kingdom (J.C.); and Clinical Unit Cambridge, Addenbrookes Hospital, Cambridge, United Kingdom (F.E., D.F.)
| | - David Mallett
- Refractory Respiratory Inflammation Discovery Performance Unit, GlaxoSmithKline, Stevenage, United Kingdom (M.B., C.D.E., J.N.H., E.M.H.); Clinical Pharmacology & Model Stimulation, (E.P., R.W.), Drug Metabolism & Pharmacokinetics, (G.V., D.M., J.M.), Refractory Respiratory Inflammation DPU, (S.U.), In Vitro In Vivo Translation, (C.F.N.), Discovery Medicine, (A.C) GlaxoSmithKline, Stevenage, United Kingdom; Global Clinical Science & Delivery, GlaxoSmithKline, Stockley Park, Uxbridge, United Kingdom (M.M., J.G.); In Vitro In Vivo Translation, (A.H.), Drug Product Design & Development, (M.I.H.), GlaxoSmithKline, Ware, United Kingdom (M.I.H.); Department of Biological Chemistry, Babraham Institute, Cambridge, United Kingdom (J.C.); and Clinical Unit Cambridge, Addenbrookes Hospital, Cambridge, United Kingdom (F.E., D.F.)
| | - Josie Morrell
- Refractory Respiratory Inflammation Discovery Performance Unit, GlaxoSmithKline, Stevenage, United Kingdom (M.B., C.D.E., J.N.H., E.M.H.); Clinical Pharmacology & Model Stimulation, (E.P., R.W.), Drug Metabolism & Pharmacokinetics, (G.V., D.M., J.M.), Refractory Respiratory Inflammation DPU, (S.U.), In Vitro In Vivo Translation, (C.F.N.), Discovery Medicine, (A.C) GlaxoSmithKline, Stevenage, United Kingdom; Global Clinical Science & Delivery, GlaxoSmithKline, Stockley Park, Uxbridge, United Kingdom (M.M., J.G.); In Vitro In Vivo Translation, (A.H.), Drug Product Design & Development, (M.I.H.), GlaxoSmithKline, Ware, United Kingdom (M.I.H.); Department of Biological Chemistry, Babraham Institute, Cambridge, United Kingdom (J.C.); and Clinical Unit Cambridge, Addenbrookes Hospital, Cambridge, United Kingdom (F.E., D.F.)
| | - Martin I Hingle
- Refractory Respiratory Inflammation Discovery Performance Unit, GlaxoSmithKline, Stevenage, United Kingdom (M.B., C.D.E., J.N.H., E.M.H.); Clinical Pharmacology & Model Stimulation, (E.P., R.W.), Drug Metabolism & Pharmacokinetics, (G.V., D.M., J.M.), Refractory Respiratory Inflammation DPU, (S.U.), In Vitro In Vivo Translation, (C.F.N.), Discovery Medicine, (A.C) GlaxoSmithKline, Stevenage, United Kingdom; Global Clinical Science & Delivery, GlaxoSmithKline, Stockley Park, Uxbridge, United Kingdom (M.M., J.G.); In Vitro In Vivo Translation, (A.H.), Drug Product Design & Development, (M.I.H.), GlaxoSmithKline, Ware, United Kingdom (M.I.H.); Department of Biological Chemistry, Babraham Institute, Cambridge, United Kingdom (J.C.); and Clinical Unit Cambridge, Addenbrookes Hospital, Cambridge, United Kingdom (F.E., D.F.)
| | - Sorif Uddin
- Refractory Respiratory Inflammation Discovery Performance Unit, GlaxoSmithKline, Stevenage, United Kingdom (M.B., C.D.E., J.N.H., E.M.H.); Clinical Pharmacology & Model Stimulation, (E.P., R.W.), Drug Metabolism & Pharmacokinetics, (G.V., D.M., J.M.), Refractory Respiratory Inflammation DPU, (S.U.), In Vitro In Vivo Translation, (C.F.N.), Discovery Medicine, (A.C) GlaxoSmithKline, Stevenage, United Kingdom; Global Clinical Science & Delivery, GlaxoSmithKline, Stockley Park, Uxbridge, United Kingdom (M.M., J.G.); In Vitro In Vivo Translation, (A.H.), Drug Product Design & Development, (M.I.H.), GlaxoSmithKline, Ware, United Kingdom (M.I.H.); Department of Biological Chemistry, Babraham Institute, Cambridge, United Kingdom (J.C.); and Clinical Unit Cambridge, Addenbrookes Hospital, Cambridge, United Kingdom (F.E., D.F.)
| | - Filzah Ehtesham
- Refractory Respiratory Inflammation Discovery Performance Unit, GlaxoSmithKline, Stevenage, United Kingdom (M.B., C.D.E., J.N.H., E.M.H.); Clinical Pharmacology & Model Stimulation, (E.P., R.W.), Drug Metabolism & Pharmacokinetics, (G.V., D.M., J.M.), Refractory Respiratory Inflammation DPU, (S.U.), In Vitro In Vivo Translation, (C.F.N.), Discovery Medicine, (A.C) GlaxoSmithKline, Stevenage, United Kingdom; Global Clinical Science & Delivery, GlaxoSmithKline, Stockley Park, Uxbridge, United Kingdom (M.M., J.G.); In Vitro In Vivo Translation, (A.H.), Drug Product Design & Development, (M.I.H.), GlaxoSmithKline, Ware, United Kingdom (M.I.H.); Department of Biological Chemistry, Babraham Institute, Cambridge, United Kingdom (J.C.); and Clinical Unit Cambridge, Addenbrookes Hospital, Cambridge, United Kingdom (F.E., D.F.)
| | - Miriam Marotti
- Refractory Respiratory Inflammation Discovery Performance Unit, GlaxoSmithKline, Stevenage, United Kingdom (M.B., C.D.E., J.N.H., E.M.H.); Clinical Pharmacology & Model Stimulation, (E.P., R.W.), Drug Metabolism & Pharmacokinetics, (G.V., D.M., J.M.), Refractory Respiratory Inflammation DPU, (S.U.), In Vitro In Vivo Translation, (C.F.N.), Discovery Medicine, (A.C) GlaxoSmithKline, Stevenage, United Kingdom; Global Clinical Science & Delivery, GlaxoSmithKline, Stockley Park, Uxbridge, United Kingdom (M.M., J.G.); In Vitro In Vivo Translation, (A.H.), Drug Product Design & Development, (M.I.H.), GlaxoSmithKline, Ware, United Kingdom (M.I.H.); Department of Biological Chemistry, Babraham Institute, Cambridge, United Kingdom (J.C.); and Clinical Unit Cambridge, Addenbrookes Hospital, Cambridge, United Kingdom (F.E., D.F.)
| | - Andrew Harrell
- Refractory Respiratory Inflammation Discovery Performance Unit, GlaxoSmithKline, Stevenage, United Kingdom (M.B., C.D.E., J.N.H., E.M.H.); Clinical Pharmacology & Model Stimulation, (E.P., R.W.), Drug Metabolism & Pharmacokinetics, (G.V., D.M., J.M.), Refractory Respiratory Inflammation DPU, (S.U.), In Vitro In Vivo Translation, (C.F.N.), Discovery Medicine, (A.C) GlaxoSmithKline, Stevenage, United Kingdom; Global Clinical Science & Delivery, GlaxoSmithKline, Stockley Park, Uxbridge, United Kingdom (M.M., J.G.); In Vitro In Vivo Translation, (A.H.), Drug Product Design & Development, (M.I.H.), GlaxoSmithKline, Ware, United Kingdom (M.I.H.); Department of Biological Chemistry, Babraham Institute, Cambridge, United Kingdom (J.C.); and Clinical Unit Cambridge, Addenbrookes Hospital, Cambridge, United Kingdom (F.E., D.F.)
| | - Carla F Newman
- Refractory Respiratory Inflammation Discovery Performance Unit, GlaxoSmithKline, Stevenage, United Kingdom (M.B., C.D.E., J.N.H., E.M.H.); Clinical Pharmacology & Model Stimulation, (E.P., R.W.), Drug Metabolism & Pharmacokinetics, (G.V., D.M., J.M.), Refractory Respiratory Inflammation DPU, (S.U.), In Vitro In Vivo Translation, (C.F.N.), Discovery Medicine, (A.C) GlaxoSmithKline, Stevenage, United Kingdom; Global Clinical Science & Delivery, GlaxoSmithKline, Stockley Park, Uxbridge, United Kingdom (M.M., J.G.); In Vitro In Vivo Translation, (A.H.), Drug Product Design & Development, (M.I.H.), GlaxoSmithKline, Ware, United Kingdom (M.I.H.); Department of Biological Chemistry, Babraham Institute, Cambridge, United Kingdom (J.C.); and Clinical Unit Cambridge, Addenbrookes Hospital, Cambridge, United Kingdom (F.E., D.F.)
| | - Disala Fernando
- Refractory Respiratory Inflammation Discovery Performance Unit, GlaxoSmithKline, Stevenage, United Kingdom (M.B., C.D.E., J.N.H., E.M.H.); Clinical Pharmacology & Model Stimulation, (E.P., R.W.), Drug Metabolism & Pharmacokinetics, (G.V., D.M., J.M.), Refractory Respiratory Inflammation DPU, (S.U.), In Vitro In Vivo Translation, (C.F.N.), Discovery Medicine, (A.C) GlaxoSmithKline, Stevenage, United Kingdom; Global Clinical Science & Delivery, GlaxoSmithKline, Stockley Park, Uxbridge, United Kingdom (M.M., J.G.); In Vitro In Vivo Translation, (A.H.), Drug Product Design & Development, (M.I.H.), GlaxoSmithKline, Ware, United Kingdom (M.I.H.); Department of Biological Chemistry, Babraham Institute, Cambridge, United Kingdom (J.C.); and Clinical Unit Cambridge, Addenbrookes Hospital, Cambridge, United Kingdom (F.E., D.F.)
| | - Jonathan Clark
- Refractory Respiratory Inflammation Discovery Performance Unit, GlaxoSmithKline, Stevenage, United Kingdom (M.B., C.D.E., J.N.H., E.M.H.); Clinical Pharmacology & Model Stimulation, (E.P., R.W.), Drug Metabolism & Pharmacokinetics, (G.V., D.M., J.M.), Refractory Respiratory Inflammation DPU, (S.U.), In Vitro In Vivo Translation, (C.F.N.), Discovery Medicine, (A.C) GlaxoSmithKline, Stevenage, United Kingdom; Global Clinical Science & Delivery, GlaxoSmithKline, Stockley Park, Uxbridge, United Kingdom (M.M., J.G.); In Vitro In Vivo Translation, (A.H.), Drug Product Design & Development, (M.I.H.), GlaxoSmithKline, Ware, United Kingdom (M.I.H.); Department of Biological Chemistry, Babraham Institute, Cambridge, United Kingdom (J.C.); and Clinical Unit Cambridge, Addenbrookes Hospital, Cambridge, United Kingdom (F.E., D.F.)
| | - Anthony Cahn
- Refractory Respiratory Inflammation Discovery Performance Unit, GlaxoSmithKline, Stevenage, United Kingdom (M.B., C.D.E., J.N.H., E.M.H.); Clinical Pharmacology & Model Stimulation, (E.P., R.W.), Drug Metabolism & Pharmacokinetics, (G.V., D.M., J.M.), Refractory Respiratory Inflammation DPU, (S.U.), In Vitro In Vivo Translation, (C.F.N.), Discovery Medicine, (A.C) GlaxoSmithKline, Stevenage, United Kingdom; Global Clinical Science & Delivery, GlaxoSmithKline, Stockley Park, Uxbridge, United Kingdom (M.M., J.G.); In Vitro In Vivo Translation, (A.H.), Drug Product Design & Development, (M.I.H.), GlaxoSmithKline, Ware, United Kingdom (M.I.H.); Department of Biological Chemistry, Babraham Institute, Cambridge, United Kingdom (J.C.); and Clinical Unit Cambridge, Addenbrookes Hospital, Cambridge, United Kingdom (F.E., D.F.)
| | - Edith M Hessel
- Refractory Respiratory Inflammation Discovery Performance Unit, GlaxoSmithKline, Stevenage, United Kingdom (M.B., C.D.E., J.N.H., E.M.H.); Clinical Pharmacology & Model Stimulation, (E.P., R.W.), Drug Metabolism & Pharmacokinetics, (G.V., D.M., J.M.), Refractory Respiratory Inflammation DPU, (S.U.), In Vitro In Vivo Translation, (C.F.N.), Discovery Medicine, (A.C) GlaxoSmithKline, Stevenage, United Kingdom; Global Clinical Science & Delivery, GlaxoSmithKline, Stockley Park, Uxbridge, United Kingdom (M.M., J.G.); In Vitro In Vivo Translation, (A.H.), Drug Product Design & Development, (M.I.H.), GlaxoSmithKline, Ware, United Kingdom (M.I.H.); Department of Biological Chemistry, Babraham Institute, Cambridge, United Kingdom (J.C.); and Clinical Unit Cambridge, Addenbrookes Hospital, Cambridge, United Kingdom (F.E., D.F.)
| |
Collapse
|
30
|
Perry MWD, Abdulai R, Mogemark M, Petersen J, Thomas MJ, Valastro B, Westin Eriksson A. Evolution of PI3Kγ and δ Inhibitors for Inflammatory and Autoimmune Diseases. J Med Chem 2018; 62:4783-4814. [DOI: 10.1021/acs.jmedchem.8b01298] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
| | - Raolat Abdulai
- Respiratory, Inflammation & Autoimmunity Translational Medicine Unit, Early Clinical Development, IMED Biotech Unit, AstraZeneca, Boston, Massachusetts 02451, United States
- Brigham and Women’s Hospital, Boston, Massachusetts 02115, United States
| | | | | | | | | | | |
Collapse
|