1
|
Tadayasu Y, Sarubbi D, Furuichi T, Eleftheraki A, Nakamura S, Sauter W, Hanada R. A randomized phase I study of the safety and pharmacokinetics of BI 1291583 in healthy Japanese male subjects. Br J Clin Pharmacol 2024. [PMID: 39299301 DOI: 10.1111/bcp.16249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 08/02/2024] [Accepted: 08/22/2024] [Indexed: 09/22/2024] Open
Abstract
AIMS Bronchiectasis patients face an unmet need for treatment options that reduce inflammation. Cathepsin C inhibition is expected to achieve this by reducing the activation of neutrophil-derived serine proteases. Here, we present safety and pharmacokinetic (PK) data from a Phase I trial evaluating the novel cathepsin C inhibitor BI 1291583 in healthy Japanese male subjects. METHODS This randomized, double-blind, placebo-controlled, parallel-group study investigated BI 1291583 in healthy Japanese male subjects (jRCT2071210111) and consisted of a single-rising-dose (SRD) part and a multiple-dose (MD) part. The primary endpoint was the percentage of subjects with drug-related treatment-emergent adverse events (AEs). Secondary PK endpoints (SRD: AUC0-∞ and Cmax; MD: AUCτ,1 and Cmax,1 after first dose and AUCτ,ss and Cmax,ss after last dose), as well as further safety and PK endpoints, were also assessed. RESULTS Overall, 36 subjects (n = 24 for SRD part; n = 12 for MD part) entered this Phase I trial. BI 1291583 was safe and well tolerated across the doses tested. All AEs were of mild intensity, with no drug-related treatment-emergent AEs, deaths, serious AEs or AEs of special interest reported in either part of the trial. Following both SRD and MD administration, BI 1291583 was readily absorbed, and PK was supraproportional over the doses assessed. CONCLUSION The results show that BI 1291583 has an appropriate benefit-risk ratio for Japanese patients, with no safety or exposure concerns at the doses studied. Japanese patients with bronchiectasis can be safely integrated into future global clinical trials of BI 1291583, with no dose adjustment required.
Collapse
Affiliation(s)
| | - Donald Sarubbi
- Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT, USA
| | | | | | | | - Wiebke Sauter
- Boehringer Ingelheim International GmbH, Biberach, Germany
| | | |
Collapse
|
2
|
Badorrek P, Diefenbach C, Kögler H, Eleftheraki A, Seitz F, Hohlfeld JM. Novel cathepsin C inhibitor, BI 1291583, intended for treatment of bronchiectasis: Phase I characterization in healthy volunteers. Clin Transl Sci 2024; 17:e13891. [PMID: 39175217 PMCID: PMC11341832 DOI: 10.1111/cts.13891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 07/24/2024] [Accepted: 06/27/2024] [Indexed: 08/24/2024] Open
Abstract
Novel treatments are needed to reduce inflammation, improve symptoms, address exacerbations, and slow disease progression in bronchiectasis. Cathepsin C (CatC) inhibition promises to achieve this through reduction of neutrophil-derived serine protease (including neutrophil elastase [NE] and proteinase 3 [PR3]) activation. Here, we present the phase I characterization of the novel CatC inhibitor, BI 1291583. Five phase I trials of BI 1291583 in healthy subjects are presented: a single-rising-dose study (NCT03414008) and two multiple-rising-dose studies (NCT03868540 and NCT04866160) assessing the safety, tolerability, pharmacodynamics, and pharmacokinetics of BI 1291583; a food effect study (NCT03837964); and a drug-drug interaction study (NCT03890887) of BI 1291583 and itraconazole. BI 1291583 was safe and well tolerated across the doses tested in these trials. Most adverse events (AEs) were mild or moderate in intensity, with no serious AEs, AEs of special interest or deaths reported in any trial. Drug-related skin exfoliation was not reported more frequently in subjects treated with BI 1291583 compared with placebo. BI 1291583 was readily absorbed, and pharmacokinetics were supra-proportional over the dose ranges assessed. Additionally, BI 1291583 inhibited CatC in a dose-dependent manner, inhibited downstream NE activity, and decreased PR3 levels. No food effect was observed. Co-administration of multiple doses of itraconazole increased BI 1291583 exposure approximately twofold. Due to these promising phase I results, a multinational phase II program of BI 1291583 in adults with bronchiectasis is ongoing (Airleaf™ [NCT05238675], Clairafly™ [NCT05865886], and Clairleaf™ [NCT05846230]).
Collapse
Affiliation(s)
- Philipp Badorrek
- Department of Clinical Airway ResearchFraunhofer Institute for Toxicology and Experimental Medicine (ITEM)HannoverGermany
| | | | - Harald Kögler
- Boehringer Ingelheim International GmbHIngelheimGermany
| | | | | | - Jens M. Hohlfeld
- Department of Clinical Airway ResearchFraunhofer Institute for Toxicology and Experimental Medicine (ITEM)HannoverGermany
- Department of Respiratory MedicineHannover Medical SchoolHannoverGermany
- German Centre for Lung Research (DZL)HannoverGermany
| |
Collapse
|
3
|
Perea L, Faner R, Chalmers JD, Sibila O. Pathophysiology and genomics of bronchiectasis. Eur Respir Rev 2024; 33:240055. [PMID: 38960613 PMCID: PMC11220622 DOI: 10.1183/16000617.0055-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 05/02/2024] [Indexed: 07/05/2024] Open
Abstract
Bronchiectasis is a complex and heterogeneous inflammatory chronic respiratory disease with an unknown cause in around 30-40% of patients. The presence of airway infection together with chronic inflammation, airway mucociliary dysfunction and lung damage are key components of the vicious vortex model that better describes its pathophysiology. Although bronchiectasis research has significantly increased over the past years and different endotypes have been identified, there are still major gaps in the understanding of the pathophysiology. Genomic approaches may help to identify new endotypes, as has been shown in other chronic airway diseases, such as COPD.Different studies have started to work in this direction, and significant contributions to the understanding of the microbiome and proteome diversity have been made in bronchiectasis in recent years. However, the systematic application of omics approaches to identify new molecular insights into the pathophysiology of bronchiectasis (endotypes) is still limited compared with other respiratory diseases.Given the complexity and diversity of these technologies, this review describes the key components of the pathophysiology of bronchiectasis and how genomics can be applied to increase our knowledge, including the study of new techniques such as proteomics, metabolomics and epigenomics. Furthermore, we propose that the novel concept of trained innate immunity, which is driven by microbiome exposures leading to epigenetic modifications, can complement our current understanding of the vicious vortex. Finally, we discuss the challenges, opportunities and implications of genomics application in clinical practice for better patient stratification into new therapies.
Collapse
Affiliation(s)
- Lidia Perea
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Rosa Faner
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias M.P. (CIBERES), Barcelona, Spain
| | - James D Chalmers
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Oriol Sibila
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias M.P. (CIBERES), Barcelona, Spain
- Respiratory Department, Hospital Clínic, University of Barcelona, Barcelona, Spain
| |
Collapse
|
4
|
Mall MA, Davies JC, Donaldson SH, Jain R, Chalmers JD, Shteinberg M. Neutrophil serine proteases in cystic fibrosis: role in disease pathogenesis and rationale as a therapeutic target. Eur Respir Rev 2024; 33:240001. [PMID: 39293854 PMCID: PMC11409056 DOI: 10.1183/16000617.0001-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 07/09/2024] [Indexed: 09/20/2024] Open
Abstract
Chronic airway inflammation is a central feature in the pathogenesis of bronchiectasis (BE), which can be caused by cystic fibrosis (CFBE; hereafter referred to as CF lung disease) and non-CF-related conditions (NCFBE). Inflammation in both CF lung disease and NCFBE is predominantly driven by neutrophils, which release proinflammatory cytokines and granule proteins, including neutrophil serine proteases (NSPs). NSPs include neutrophil elastase, proteinase 3 and cathepsin G. An imbalance between NSPs and their antiproteases has been observed in people with CF lung disease and people with NCFBE. While the role of the protease/antiprotease imbalance is well established in both CF lung disease and NCFBE, effective therapies targeting NSPs are lacking. In recent years, the introduction of CF transmembrane conductance regulator (CFTR) modulator therapy has immensely improved outcomes in many people with CF (pwCF). Despite this, evidence suggests that airway inflammation persists, even in pwCF treated with CFTR modulator therapy. In this review, we summarise current data on neutrophilic inflammation in CF lung disease to assess whether neutrophilic inflammation and high, uncontrolled NSP levels play similar roles in CF lung disease and in NCFBE. We discuss similarities between the neutrophilic inflammatory profiles of people with CF lung disease and NCFBE, potentially supporting a similar therapeutic approach. Additionally, we present evidence suggesting that neutrophilic inflammation persists in pwCF treated with CFTR modulator therapy, at levels similar to those in people with NCFBE. Collectively, these findings highlight the ongoing need for new treatment strategies targeting neutrophilic inflammation in CF lung disease.
Collapse
Affiliation(s)
- Marcus A Mall
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine and Cystic Fibrosis Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Center for Lung Research (DZL), associated partner site, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Jane C Davies
- National Heart and Lung Institute, Imperial College London, London, UK
- Royal Brompton Hospital, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Scott H Donaldson
- Department of Medicine, Division of Pulmonary Diseases and Critical Care Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Raksha Jain
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | - Michal Shteinberg
- Lady Davis Carmel Medical Center, Haifa, Israel
- The B. Rappaport Faculty of Medicine, Technion Institute of Technology, Haifa, Israel
| |
Collapse
|
5
|
Dougherty GW, Ostrowski LE, Nöthe-Menchen T, Raidt J, Schramm A, Olbrich H, Yin W, Sears PR, Dang H, Smith AJ, Beule AG, Hjeij R, Rutjes N, Haarman EG, Maas SM, Ferkol TW, Noone PG, Olivier KN, Bracht DC, Barbry P, Zaragosi LE, Fierville M, Kliesch S, Wohlgemuth K, König J, George S, Loges NT, Ceppe A, Markovetz MR, Luo H, Guo T, Rizk H, Eldesoky T, Dahlke K, Boldt K, Ueffing M, Hill DB, Pang YP, Knowles MR, Zariwala MA, Omran H. Recessively Inherited Deficiency of Secreted WFDC2 (HE4) Causes Nasal Polyposis and Bronchiectasis. Am J Respir Crit Care Med 2024; 210:63-76. [PMID: 38626355 PMCID: PMC11197063 DOI: 10.1164/rccm.202308-1370oc] [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: 08/07/2023] [Accepted: 04/16/2024] [Indexed: 04/18/2024] Open
Abstract
Rationale: Bronchiectasis is a pathological dilatation of the bronchi in the respiratory airways associated with environmental or genetic causes (e.g., cystic fibrosis, primary ciliary dyskinesia, and primary immunodeficiency disorders), but most cases remain idiopathic. Objectives: To identify novel genetic defects in unsolved cases of bronchiectasis presenting with severe rhinosinusitis, nasal polyposis, and pulmonary Pseudomonas aeruginosa infection. Methods: DNA was analyzed by next-generation or targeted Sanger sequencing. RNA was analyzed by quantitative PCR and single-cell RNA sequencing. Patient-derived cells, cell cultures, and secretions (mucus, saliva, seminal fluid) were analyzed by Western blotting and immunofluorescence microscopy, and mucociliary activity was measured. Blood serum was analyzed by electrochemiluminescence immunoassay. Protein structure and proteomic analyses were used to assess the impact of a disease-causing founder variant. Measurements and Main Results: We identified biallelic pathogenic variants in WAP four-disulfide core domain 2 (WFDC2) in 11 individuals from 10 unrelated families originating from the United States, Europe, Asia, and Africa. Expression of WFDC2 was detected predominantly in secretory cells of control airway epithelium and also in submucosal glands. We demonstrate that WFDC2 is below the limit of detection in blood serum and hardly detectable in samples of saliva, seminal fluid, and airway surface liquid from WFDC2-deficient individuals. Computer simulations and deglycosylation assays indicate that the disease-causing founder variant p.Cys49Arg structurally hampers glycosylation and, thus, secretion of mature WFDC2. Conclusions: WFDC2 dysfunction defines a novel molecular etiology of bronchiectasis characterized by the deficiency of a secreted component of the airways. A commercially available blood test combined with genetic testing allows its diagnosis.
Collapse
Grants
- UM1 HG006504 NHGRI NIH HHS
- U2C TR002818 NCATS NIH HHS
- U54 HL096458 NHLBI NIH HHS
- R01 HL117836 NHLBI NIH HHS
- R01 HL071798 NHLBI NIH HHS
- Wellcome Trust
- P30 DK065988 NIDDK NIH HHS
- OM6/7-2 Deutsche Forschungsgemeinschaft
- OM6/8-2 Deutsche Forschungsgemeinschaft
- OM6/10-1 Deutsche Forschungsgemeinschaft
- CRU 326 Deutsche Forschungsgemeinschaft
- OL450/3-1 Deutsche Forschungsgemeinschaft
- RA3522/1-1 Deutsche Forschungsgemeinschaft
- HJ 7/1-1 Deutsche Forschungsgemeinschaft
- HJ 7/1-3 Deutsche Forschungsgemeinschaft
- Om2/009/12 Interdisziplinäres Zentrum für klinische Forschung Münster (IZKF)
- Om2/015/16 Interdisziplinäres Zentrum für klinische Forschung Münster (IZKF)
- Om2/010/20 Interdisziplinäres Zentrum für klinische Forschung Münster (IZKF)
- Horizon2020 GA 777295 European Commission Registry Warehouse
- EU FP7 GA 305404 BESTCILIA
- Tistou and Charlotte Kerstan Stiftung
- 210585/Z/18/Z Wellcome Trust
- U54HL096458 US NIH/ Office of Rare Diseases Research/National Center for Advancing Translational Sciences (NCATS)/National Heart, Lung, and Blood Institute
- R01HL071798 US NIH
- R01HL117836 US NIH
- X01HL115246-01 US NIH
- 82070003 National Natural Science Foundation of China
- 82270048 National Natural Science Foundation of China
- 91967 Mayo Foundation for Medical Education and Research
- 21EQUI09Z6RCHX CNRS, Inserm, the infrastructure France Génomique and the French Government (Agence Nationale de Recherche, ANR)
- ANR-19-P3IA-0002 CNRS, Inserm, the infrastructure France Génomique and the French Government (Agence Nationale de Recherche, ANR)
- ANR-19-CE14-0027 CNRS, Inserm, the infrastructure France Génomique and the French Government (Agence Nationale de Recherche, ANR)
- ANR-19-P3IA-0002-3IA CNRS, Inserm, the infrastructure France Génomique and the French Government (Agence Nationale de Recherche, ANR)
- ANR-21-ESRE-0052 CNRS, Inserm, the infrastructure France Génomique and the French Government (Agence Nationale de Recherche, ANR)
- ANR-10-INBS-09-03 CNRS, Inserm, the infrastructure France Génomique and the French Government (Agence Nationale de Recherche, ANR)
- ANR-10-INBS-09-02 CNRS, Inserm, the infrastructure France Génomique and the French Government (Agence Nationale de Recherche, ANR)
- 2017-175159-5022 Canceropôle PACA, the H2020 Health (Discovair) and the Chan Zuckerberg Initiative
Collapse
Affiliation(s)
| | - Lawrence E. Ostrowski
- Department of Pediatrics
- Marsico Lung Institute/Cystic Fibrosis Research and Treatment Center
| | | | | | | | | | - Weining Yin
- Marsico Lung Institute/Cystic Fibrosis Research and Treatment Center
| | - Patrick R. Sears
- Marsico Lung Institute/Cystic Fibrosis Research and Treatment Center
| | - Hong Dang
- Marsico Lung Institute/Cystic Fibrosis Research and Treatment Center
| | - Amanda J. Smith
- Marsico Lung Institute/Cystic Fibrosis Research and Treatment Center
| | | | | | - Niels Rutjes
- Department of Pediatric Pulmonology and Allergy, Emma Children’s Hospital, Amsterdam, the Netherlands
| | - Eric G. Haarman
- Department of Pediatric Pulmonology and Allergy, Emma Children’s Hospital, Amsterdam, the Netherlands
| | - Saskia M. Maas
- Department of Human Genetics, Amsterdam Reproduction and Development Research Institute, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | | | - Peadar G. Noone
- Marsico Lung Institute/Cystic Fibrosis Research and Treatment Center
- Department of Medicine
| | - Kenneth N. Olivier
- Marsico Lung Institute/Cystic Fibrosis Research and Treatment Center
- Department of Medicine
| | | | - Pascal Barbry
- Université Côte d’Azur, CNRS, Institut Pharmacologie Moléculaire et Cellulaire, Sophia-Antipolis, France
| | - Laure-Emmanuelle Zaragosi
- Université Côte d’Azur, CNRS, Institut Pharmacologie Moléculaire et Cellulaire, Sophia-Antipolis, France
| | - Morgane Fierville
- Université Côte d’Azur, CNRS, Institut Pharmacologie Moléculaire et Cellulaire, Sophia-Antipolis, France
| | - Sabine Kliesch
- Department of Clinical and Surgical Andrology, Centre of Reproductive Medicine and Andrology, University Hospital Muenster, Muenster, Germany
| | | | | | | | | | - Agathe Ceppe
- Marsico Lung Institute/Cystic Fibrosis Research and Treatment Center
| | | | - Hong Luo
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, China
| | - Ting Guo
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, China
| | - Hoda Rizk
- Department of Pediatrics, Faculty of Medicine, University of Mansoura, Mansoura, Egypt
| | - Tarek Eldesoky
- Department of Pediatrics, Faculty of Medicine, University of Mansoura, Mansoura, Egypt
| | - Katrin Dahlke
- Institute for Ophthalmic Research and Core Facility for Medical Proteomics, Tübingen, Germany
- Eberhard Karls University Tübingen, Tübingen, Germany; and
| | - Karsten Boldt
- Institute for Ophthalmic Research and Core Facility for Medical Proteomics, Tübingen, Germany
- Eberhard Karls University Tübingen, Tübingen, Germany; and
| | - Marius Ueffing
- Institute for Ophthalmic Research and Core Facility for Medical Proteomics, Tübingen, Germany
- Eberhard Karls University Tübingen, Tübingen, Germany; and
| | - David B. Hill
- Marsico Lung Institute/Cystic Fibrosis Research and Treatment Center
- Department of Physics and Astronomy, and
| | - Yuan-Ping Pang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| | - Michael R. Knowles
- Marsico Lung Institute/Cystic Fibrosis Research and Treatment Center
- Department of Medicine
| | - Maimoona A. Zariwala
- Marsico Lung Institute/Cystic Fibrosis Research and Treatment Center
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | | |
Collapse
|
6
|
Chalmers JD, Burgel PR, Daley CL, De Soyza A, Haworth CS, Mauger D, Mange K, Teper A, Fernandez C, Conroy D, Metersky M. Brensocatib in non-cystic fibrosis bronchiectasis: ASPEN protocol and baseline characteristics. ERJ Open Res 2024; 10:00151-2024. [PMID: 39040578 PMCID: PMC11261371 DOI: 10.1183/23120541.00151-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 03/20/2024] [Indexed: 07/24/2024] Open
Abstract
Introduction Brensocatib is an investigational, oral, reversible inhibitor of dipeptidyl peptidase-1 shown to prolong time to first exacerbation in adults with bronchiectasis. Outlined here are the clinical trial design, and baseline characteristics and treatment patterns of adult patients enrolled in the phase 3 ASPEN trial (NCT04594369). Methods The ASPEN trial is a global study enrolling patients with a clinical history consistent with bronchiectasis (cough, chronic sputum production and/or recurrent respiratory infections), diagnosis confirmed radiologically and ≥2 exacerbations in the prior 12 months. It was designed to evaluate the impact of two brensocatib doses (10 mg and 25 mg) on exacerbation rate over a 52-week treatment period versus placebo. Comprehensive clinical data, including demographics, disease severity, lung function, Pseudomonas aeruginosa status and quality of life, were collected at baseline. Results 1682 adults from 35 countries were randomised from December 2020 to March 2023. Mean age was 61.3 years and 64.7% were female. ∼70% had moderate-to-severe Bronchiectasis Severity Index (BSI) scores, 29.3% had ≥3 exacerbations in the prior 12 months and 35.7% were positive for P. aeruginosa. Mean BSI scores were highest in Australia/New Zealand (8.3) and lowest in Latin America (5.9). Overall, the most common aetiology was idiopathic (58.4%). In P. aeruginosa-positive versus P. aeruginosa-negative patients, lung function was lower, with greater long-term macrolide (21.5% versus 14.0%) and inhaled corticosteroid use (63.5% versus 53.9%). There was wide regional variation in long-term antibiotic use in patients with bronchiectasis and P. aeruginosa. Discussion ASPEN baseline characteristics and treatment profiles were representative of a global bronchiectasis population.
Collapse
Affiliation(s)
- James D. Chalmers
- Division of Molecular and Clinical Medicine, University of Dundee, Dundee, UK
| | - Pierre-Régis Burgel
- Hôpital Cochin and Cystic Fibrosis National Reference Center, Service de Pneumologie, AP-HP and Université Paris-Cité, Inserm U1016-Institut Cochin, Paris, France
| | - Charles L. Daley
- National Jewish Health and the University of Colorado, Denver, CO, USA
| | - Anthony De Soyza
- Population and Health Sciences Institute, NIHR Biomedical Research Centre for Aging Newcastle University and Department of Respiratory Medicine, Newcastle upon Tyne NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Charles S. Haworth
- Royal Papworth Hospital NHS Foundation Trust and University of Cambridge, Cambridge, UK
| | | | | | | | | | - Dan Conroy
- Insmed Incorporated, Bridgewater, NJ, USA
| | - Mark Metersky
- University of Connecticut School of Medicine, Farmington, CT, USA
| |
Collapse
|
7
|
Johnson E, Long MB, Chalmers JD. Biomarkers in bronchiectasis. Eur Respir Rev 2024; 33:230234. [PMID: 38960612 PMCID: PMC11220624 DOI: 10.1183/16000617.0234-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 03/09/2024] [Indexed: 07/05/2024] Open
Abstract
Bronchiectasis is a heterogeneous disease with multiple aetiologies and diverse clinical features. There is a general consensus that optimal treatment requires precision medicine approaches focused on specific treatable disease characteristics, known as treatable traits. Identifying subtypes of conditions with distinct underlying biology (endotypes) depends on the identification of biomarkers that are associated with disease features, prognosis or treatment response and which can be applied in clinical practice. Bronchiectasis is a disease characterised by inflammation, infection, structural lung damage and impaired mucociliary clearance. Increasingly there are available methods to measure each of these components of the disease, revealing heterogeneous inflammatory profiles, microbiota, radiology and mucus and epithelial biology in patients with bronchiectasis. Using emerging biomarkers and omics technologies to guide treatment in bronchiectasis is a promising field of research. Here we review the most recent data on biomarkers in bronchiectasis.
Collapse
Affiliation(s)
- Emma Johnson
- Division of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| | - Merete B Long
- Division of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| | - James D Chalmers
- Division of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| |
Collapse
|
8
|
Chotirmall SH, Chalmers JD. The Precision Medicine Era of Bronchiectasis. Am J Respir Crit Care Med 2024; 210:24-34. [PMID: 38949497 PMCID: PMC11197062 DOI: 10.1164/rccm.202403-0473pp] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 04/10/2024] [Indexed: 07/02/2024] Open
Affiliation(s)
- Sanjay H. Chotirmall
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Department of Respiratory and Critical Care Medicine, Tan Tock Seng Hospital, Singapore, Singapore; and
| | - James D. Chalmers
- Division of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, United Kingdom
| |
Collapse
|
9
|
Hou JJ, Ding L, Yang T, Yang YF, Jin YP, Zhang XP, Ma AH, Qin YH. The proteolytic activity in inflammatory bowel disease: insight from gut microbiota. Microb Pathog 2024; 188:106560. [PMID: 38272327 DOI: 10.1016/j.micpath.2024.106560] [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: 08/02/2023] [Revised: 01/20/2024] [Accepted: 01/22/2024] [Indexed: 01/27/2024]
Abstract
Inflammatory bowel disease (IBD) is a chronic, recurrent inflammatory disease caused by the destruction of the intestinal mucosal epithelium that affects a growing number of people worldwide. Although the etiology of IBD is complex and still elucidated, the role of dysbiosis and dysregulated proteolysis is well recognized. Various studies observed altered composition and diversity of gut microbiota, as well as increased proteolytic activity (PA) in serum, plasma, colonic mucosa, and fecal supernatant of IBD compared to healthy individuals. The imbalance of intestinal microecology and intestinal protein hydrolysis were gradually considered to be closely related to IBD. Notably, the pivotal role of intestinal microbiota in maintaining proteolytic balance received increasing attention. In summary, we have speculated a mesmerizing story, regarding the hidden role of PA and microbiota-derived PA hidden in IBD. Most importantly, we provided the diagnosis and therapeutic targets for IBD as well as the formulation of new treatment strategies for other digestive diseases and protease-related diseases.
Collapse
Affiliation(s)
- Jun-Jie Hou
- Department of Gastroenterology, Shaoxing People's Hospital, Shaoxing, PR China
| | - Liang Ding
- Department of Gastroenterology, Shaoxing People's Hospital, Shaoxing, PR China
| | - Tao Yang
- Department of Gastroenterology, Shaoxing People's Hospital, Shaoxing, PR China
| | - Yan-Fei Yang
- Department of Gastroenterology, Shaoxing People's Hospital, Shaoxing, PR China
| | - Yue-Ping Jin
- Department of Gastroenterology, Shaoxing People's Hospital, Shaoxing, PR China
| | - Xiao-Ping Zhang
- Department of Gastroenterology, Shaoxing People's Hospital, Shaoxing, PR China
| | - A-Huo Ma
- Department of Gastroenterology, Shaoxing People's Hospital, Shaoxing, PR China
| | - Yue-Hua Qin
- Department of Gastroenterology, Shaoxing People's Hospital, Shaoxing, PR China.
| |
Collapse
|
10
|
Chalmers JD, Badorrek P, Diefenbach C, Kögler H, Sauter W, Kreideweiss S, Hohlfeld JM. The preclinical and phase 1 development of the novel oral cathepsin C inhibitor BI 1291583. ERJ Open Res 2024; 10:00725-2023. [PMID: 38529344 PMCID: PMC10962448 DOI: 10.1183/23120541.00725-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 01/15/2024] [Indexed: 03/27/2024] Open
Abstract
Preclinical and phase 1 study results indicate that BI 1291583 is a reversible, highly potent and highly selective CatC inhibitor that markedly inhibits active NSP production in a dose-dependent manner, supporting phase 2 trials in bronchiectasis patients https://bit.ly/47PZ8E5.
Collapse
Affiliation(s)
- James D. Chalmers
- Division of Molecular and Clinical Medicine, University of Dundee, Dundee, UK
| | - Philipp Badorrek
- Department of Clinical Airway Research, Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hannover, Germany
| | | | - Harald Kögler
- Boehringer Ingelheim International GmbH, Ingelheim, Germany
| | - Wiebke Sauter
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | | | - Jens M. Hohlfeld
- Department of Clinical Airway Research, Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hannover, Germany
- Department of Respiratory Medicine, Hannover Medical School, Hannover, Germany
- German Centre for Lung Research (DZL), Hannover, Germany
| |
Collapse
|
11
|
Yun C, Wang Y, Wang D, Zang J, Lv Z, Liu R, Cong H. CircABCB10 Promotes the Apoptosis and Inflammatory Response of 16HBE Cells by Cigarette Smoke Extract by Targeting miR-130a/PTEN Axis. IRANIAN JOURNAL OF PUBLIC HEALTH 2024; 53:592-604. [PMID: 38919307 PMCID: PMC11194655 DOI: 10.18502/ijph.v53i3.15141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 09/19/2023] [Indexed: 06/27/2024]
Abstract
Background Chronic obstructive pulmonary disease (COPD) has become a global public health problem due to its high mortality. So there is an urgent need to find an effective treatment. Methods The targeting relationship among circABCB10, miR-130a and PTEN was predicted by the targetscan database (TargetScanHuman 8.0, https://www.targetscan.org/vert_80/). A total of 60 patients which were from the second affiliated hospital of Qiqihar Medical University from 2022 to 2023 were enrolled. The lung condition was detected by CT (Computed Tomography). The expression levels of circABCB10, miR-130a and PTEN in lung tissues were determined by qRT-PCR. The COPD model was established by stimulating normal and silenced 16HBE cells in circABCB10 genes with cigarette smoke extract (CSE) at different concentrations. qRT-PCR was conducted for the expression levels of circABCB10, miR-130a and PTEN, WB for the expression levels of apoptotic proteins, ELISA for the content of inflammatory factors, and CCK8 for the effect of CSE on the proliferation of cells. Results CircABCB10 expression increased in lung tissues from patients with COPD and in 16HBE cells treated with CSE. The stimulation on cells with CSE increased the expression of inflammatory factors, while knocking down circABCB10 could reverse this response. The inflammatory response to the knockdown of circABCB10 was reversed by miR-130a inhibitor, which increased the expression of c-caspase 3. The targetscan database predicted the target factor downstream miR-130a was PTEN. Transfecting OE-PTEN reversed the inflammation of knocking down circABCB10, and increased the apoptosis and inflammation. Conclusion CircABCB10 can cause the inflammatory response by targeting miR-130a/PTEN axis, which is a mechanism that may lead to the occurrence and development of COPD.
Collapse
Affiliation(s)
- Changping Yun
- Department of Respiration, The Second Affiliated Hospital of Qiqihar Medical University, Qiqihar 161000, China
| | - Yuguang Wang
- CT Room, the Second Affiliated Hospital of Qiqihar Medical University, Qiqihar 161000, China
| | - Dongxu Wang
- CT Room, the Second Affiliated Hospital of Qiqihar Medical University, Qiqihar 161000, China
| | - Jialin Zang
- CT Room, the Second Affiliated Hospital of Qiqihar Medical University, Qiqihar 161000, China
| | - Zhen Lv
- School of Pharmacy, Qiqihar Medical University, Qiqihar 161000, China
| | - Ruinan Liu
- CT Room, the Second Affiliated Hospital of Qiqihar Medical University, Qiqihar 161000, China
| | - Houyi Cong
- CT Room, the Second Affiliated Hospital of Qiqihar Medical University, Qiqihar 161000, China
| |
Collapse
|
12
|
Blin T, Parent C, Pichon G, Guillon A, Jouan Y, Allouchi H, Aubrey N, Boursin F, Domain R, Korkmaz B, Sécher T, Heuzé-Vourc'h N. The proteolytic airway environment associated with pneumonia acts as a barrier for treatment with anti-infective antibodies. Eur J Pharm Biopharm 2024; 195:114163. [PMID: 38086491 DOI: 10.1016/j.ejpb.2023.12.003] [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: 10/18/2023] [Revised: 12/04/2023] [Accepted: 12/08/2023] [Indexed: 01/29/2024]
Abstract
Like pneumonia, coronavirus disease 2019 (COVID-19) is characterized by a massive infiltration of innate immune cells (such as polymorphonuclear leukocytes) into the airways and alveolar spaces. These cells release proteases that may degrade therapeutic antibodies and thus limit their effectiveness. Here, we investigated the in vitro and ex vivo impact on anti-severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) IgG1s and other IgG subclasses (IgG2 and IgG4) of the neutrophil elastase, proteinase 3 and cathepsin G (the three main neutrophil serine proteases) found in endotracheal aspirates from patients with severe COVID-19. Although the IgGs were sensitive to neutrophil serine proteases, IgG2 was most resistant to proteolytic degradation. The two anti-SARS CoV2 antibodies (casirivimab and imdevimab) were sensitive to the lung's proteolytic environment, although neutrophil serine protease inhibitors only partly limited the degradation. Overall, our results show that the pneumonia-associated imbalance between proteases and their inhibitors in the airways contributes to degradation of antiviral antibodies.
Collapse
Affiliation(s)
- Timothée Blin
- INSERM, Respiratory Disease Research Centre, U1100, F-37032 Tours, France; University of Tours, F-37032 Tours, France; Tours University Hospital (CHRU), Department of Pulmonary Medicine, Cystic Fibrosis Resource Center, F-37032 Tours, France
| | - Christelle Parent
- INSERM, Respiratory Disease Research Centre, U1100, F-37032 Tours, France; University of Tours, F-37032 Tours, France
| | - Gabrielle Pichon
- INSERM, Respiratory Disease Research Centre, U1100, F-37032 Tours, France; University of Tours, F-37032 Tours, France
| | - Antoine Guillon
- INSERM, Respiratory Disease Research Centre, U1100, F-37032 Tours, France; University of Tours, F-37032 Tours, France; Tours University Hospital (CHRU), Critical Care Department, F-37032 Tours, France
| | - Youenn Jouan
- INSERM, Respiratory Disease Research Centre, U1100, F-37032 Tours, France; University of Tours, F-37032 Tours, France; Tours University Hospital (CHRU), Cardiac Surgery Department, F-37032 Tours, France
| | - Hassan Allouchi
- INSERM, Respiratory Disease Research Centre, U1100, F-37032 Tours, France; University of Tours, F-37032 Tours, France; Tours University Hospital (CHRU), Pharmacy Department, F-37032 Tours, France
| | - Nicolas Aubrey
- University of Tours, F-37032 Tours, France; UMR INRA ISP 1282, BioMap Team, F-37032 Tours, France
| | - Fanny Boursin
- University of Tours, F-37032 Tours, France; UMR INRA ISP 1282, BioMap Team, F-37032 Tours, France
| | - Roxane Domain
- INSERM, Respiratory Disease Research Centre, U1100, F-37032 Tours, France; University of Tours, F-37032 Tours, France
| | - Baris Korkmaz
- INSERM, Respiratory Disease Research Centre, U1100, F-37032 Tours, France; University of Tours, F-37032 Tours, France
| | - Thomas Sécher
- INSERM, Respiratory Disease Research Centre, U1100, F-37032 Tours, France; University of Tours, F-37032 Tours, France
| | - Nathalie Heuzé-Vourc'h
- INSERM, Respiratory Disease Research Centre, U1100, F-37032 Tours, France; University of Tours, F-37032 Tours, France.
| |
Collapse
|
13
|
McShane PJ. Investigation and Management of Bronchiectasis in Nontuberculous Mycobacterial Pulmonary Disease. Clin Chest Med 2023; 44:731-742. [PMID: 37890912 DOI: 10.1016/j.ccm.2023.07.005] [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] [Indexed: 10/29/2023]
Abstract
Patients with nontuberculous mycobacterial (NTM) lung infection require life-long attention to their bronchiectasis, whether or not their NTM infection has been cured. The identification of the cause of bronchiectasis and/or coexisting diseases is important because it may affect therapeutic strategies. Airway clearance is the mainstay of bronchiectasis management. It can include multiple breathing techniques, devices, and mucoactive agents. The exact airway clearance regimen should be customized to each individual patient. Chronic pathogenic airway bacteria, such as Pseudomonas aeruginosa, may warrant consideration of eradication therapy and/or chronic use of maintenance inhaled antibiotics.
Collapse
Affiliation(s)
- Pamela J McShane
- Department of Medicine, University of Texas Health Science Center at Tyler, 11937 Hwy 271, Tyler, TX 75708, USA.
| |
Collapse
|
14
|
Martins M, Keir HR, Chalmers JD. Endotypes in bronchiectasis: moving towards precision medicine. A narrative review. Pulmonology 2023; 29:505-517. [PMID: 37030997 DOI: 10.1016/j.pulmoe.2023.03.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 03/10/2023] [Accepted: 03/13/2023] [Indexed: 04/09/2023] Open
Abstract
Bronchiectasis is a highly complex entity that can be very challenging to investigate and manage. Patients are diverse in their aetiology, symptoms, risk of complications and outcomes. "Endotypes"- subtypes of disease with distinct biological mechanisms, has been proposed as a means of better managing bronchiectasis. This review discusses the emerging field of endotyping in bronchiectasis. We searched PubMed and Google Scholar for randomized controlled trials (RCT), observational studies, systematic reviews and meta-analysis published from inception until October 2022, using the terms: "bronchiectasis", "endotypes", "biomarkers", "microbiome" and "inflammation". Exclusion criteria included commentaries and non-English language articles as well as case reports. Duplicate articles between databases were initially identified and appropriately excluded. Studies identified suggest that it is possible to classify bronchiectasis patients into multiple endotypes deriving from their co-morbidities or underlying causes to complex infective or inflammatory endotypes. Specific biomarkers closely related to a particular endotype might be used to determine response to treatment and prognosis. The most clearly defined examples of endotypes in bronchiectasis are the underlying causes such as immunodeficiency or allergic bronchopulmonary aspergillosis where the underlying causes are clearly related to a specific treatment. The heterogeneity of bronchiectasis extends, however, far beyond aetiology and it is now possible to identify subtypes of disease based on inflammatory mechanisms such airway neutrophil extracellular traps and eosinophilia. In future biomarkers of host response and infection, including the microbiome may be useful to guide treatments and to increase the success of randomized trials. Advances in the understanding the inflammatory pathways, microbiome, and genetics in bronchiectasis are key to move towards a personalized medicine in bronchiectasis.
Collapse
Affiliation(s)
- M Martins
- Pulmonology Department, Centro Hospitalar Universitário de São João, Porto, Portugal.
| | - H R Keir
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, Dundee, DD1 9SY, Scotland, United Kinkdom
| | - J D Chalmers
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, Dundee, DD1 9SY, Scotland, United Kinkdom
| |
Collapse
|
15
|
Lushington GH, Linde A, Melgarejo T. Bacterial Proteases as Potentially Exploitable Modulators of SARS-CoV-2 Infection: Logic from the Literature, Informatics, and Inspiration from the Dog. BIOTECH 2023; 12:61. [PMID: 37987478 PMCID: PMC10660736 DOI: 10.3390/biotech12040061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/19/2023] [Accepted: 10/18/2023] [Indexed: 11/22/2023] Open
Abstract
(1) Background: The COVID-19 pandemic left many intriguing mysteries. Retrospective vulnerability trends tie as strongly to odd demographics as to exposure profiles, genetics, health, or prior medical history. This article documents the importance of nasal microbiome profiles in distinguishing infection rate trends among differentially affected subgroups. (2) Hypothesis: From a detailed literature survey, microbiome profiling experiments, bioinformatics, and molecular simulations, we propose that specific commensal bacterial species in the Pseudomonadales genus confer protection against SARS-CoV-2 infections by expressing proteases that may interfere with the proteolytic priming of the Spike protein. (3) Evidence: Various reports have found elevated Moraxella fractions in the nasal microbiomes of subpopulations with higher resistance to COVID-19 (e.g., adolescents, COVID-19-resistant children, people with strong dietary diversity, and omnivorous canines) and less abundant ones in vulnerable subsets (the elderly, people with narrower diets, carnivorous cats and foxes), along with bioinformatic evidence that Moraxella bacteria express proteases with notable homology to human TMPRSS2. Simulations suggest that these proteases may proteolyze the SARS-CoV-2 spike protein in a manner that interferes with TMPRSS2 priming.
Collapse
Affiliation(s)
| | - Annika Linde
- College of Veterinary Medicine, Western University of Health Sciences, Pomona, CA 91766, USA;
| | - Tonatiuh Melgarejo
- College of Veterinary Medicine, Western University of Health Sciences, Pomona, CA 91766, USA;
| |
Collapse
|
16
|
Barbosa M, Chalmers JD. Bronchiectasis. Presse Med 2023; 52:104174. [PMID: 37778637 DOI: 10.1016/j.lpm.2023.104174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 09/26/2023] [Indexed: 10/03/2023] Open
Abstract
Bronchiectasis is a final common pathway of a wide variety of underlying conditions including infectious, autoimmune, allergic, genetic and inflammatory conditions. Patients experience a chronic disease with variable clinical symptoms and course, but most experience cough, sputum production and recurrent exacerbations. Symptoms of bronchiectasis lead to poor quality of life and exacerbations are the major driver of morbidity and mortality. Patients are often chronically infected with bacteria with the most common being Pseudomonas aeruginosa and Haemophilus influenzae. Treatment of bronchiectasis includes standardised testing to identify the underlying cause with targeted treatment if immune deficiency, allergic bronchopulmonary aspergillosis or non-tuberculous mycobacterial infection, for example, are identified. Airway clearance is the mainstay of therapy for patients with symptoms of cough and sputum production. Frequently exacerbating patients may benefit from long term antibiotic or mucoactive therapies. Bronchiectasis is a heterogeneous disease and increasingly precision medicine approaches are advocated to target treatments most appropriately and to limit the emergence of antimicrobial resistance.
Collapse
Affiliation(s)
- Miguel Barbosa
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, Dundee, DD1 9SY, UK
| | - James D Chalmers
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, Dundee, DD1 9SY, UK.
| |
Collapse
|
17
|
Perea L, Faner R, Solarat B, Shoemark A, Aliberti S, Chalmers JD, Sibila O. Low Salivary Secretory Leukocyte Protease Inhibitor Levels Are Related to Airway Pseudomonas aeruginosa Infection in Bronchiectasis. Chest 2023; 164:323-326. [PMID: 36898430 DOI: 10.1016/j.chest.2023.02.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/17/2023] [Accepted: 02/28/2023] [Indexed: 03/12/2023] Open
Affiliation(s)
- Lidia Perea
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Barcelona, Spain
| | - Rosa Faner
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Barcelona, Spain
| | - Belen Solarat
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Respiratory Department, Hospital Clínic, University of Barcelona, Barcelona, Spain
| | - Amelia Shoemark
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Milan, Italy
| | - Stefano Aliberti
- Department of Biomedical Sciences, Humanitas University, Milan, Italy; IRCCS Humanitas Research Hospital, Respiratory Unit, Milan, Italy
| | - James D Chalmers
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Milan, Italy
| | - Oriol Sibila
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Barcelona, Spain; Respiratory Department, Hospital Clínic, University of Barcelona, Barcelona, Spain.
| |
Collapse
|
18
|
Kreideweiss S, Schänzle G, Schnapp G, Vintonyak V, Grundl MA. BI 1291583: a novel selective inhibitor of cathepsin C with superior in vivo profile for the treatment of bronchiectasis. Inflamm Res 2023; 72:1709-1717. [PMID: 37542002 PMCID: PMC10499737 DOI: 10.1007/s00011-023-01774-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 06/26/2023] [Accepted: 07/24/2023] [Indexed: 08/06/2023] Open
Abstract
BACKGROUND Airway inflammation in chronic inflammatory lung diseases (e.g. bronchiectasis) is partly mediated by neutrophil-derived serine protease (NSP)/antiprotease imbalance. NSPs are activated during neutrophil myelopoiesis in bone marrow by cathepsin C (CatC; DPP1). CatC is therefore an attractive target to reduce NSP activity in the lungs of patients with bronchiectasis, restoring the protease/antiprotease balance. We report results from the preclinical pharmacological assessment of the novel CatC inhibitor BI 1291583. METHODS Binding kinetics of BI 1291583 to human CatC were determined by surface plasmon resonance. In vitro inhibition of human CatC activity was determined by CatC-specific fluorescent assay, and selectivity was assessed against related cathepsins and unrelated proteases. Inhibition of NSP neutrophil elastase (NE) production was assessed in a human neutrophil progenitor cell line. In vivo inhibition of NE and NSP proteinase 3 (PR3) in bronchoalveolar lavage fluid (BALF) neutrophils after lipopolysaccharide (LPS) challenge and distribution of BI 1291583 was determined in a mouse model. RESULTS BI 1291583 bound human CatC in a covalent, reversible manner, selectively and fully inhibiting CatC enzymatic activity. This inhibition translated to concentration-dependent inhibition of NE activation in U937 cells and dose-dependent, almost-complete inhibition of NE and PR3 activity in BALF neutrophils in an in vivo LPS-challenge model in mice. BI 1291583 exhibited up to 100 times the exposure in the target tissue bone marrow compared with plasma. CONCLUSION BI 1291583-mediated inhibition of CatC is expected to restore the protease-antiprotease balance in the lungs of patients with chronic airway inflammatory diseases such as bronchiectasis.
Collapse
Affiliation(s)
| | | | - Gisela Schnapp
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | | | - Marc A Grundl
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany.
| |
Collapse
|
19
|
Chalmers JD, Gupta A, Chotirmall SH, Armstrong A, Eickholz P, Hasegawa N, McShane PJ, O'Donnell AE, Shteinberg M, Watz H, Eleftheraki A, Diefenbach C, Sauter W. A Phase 2 randomised study to establish efficacy, safety and dosing of a novel oral cathepsin C inhibitor, BI 1291583, in adults with bronchiectasis: Airleaf. ERJ Open Res 2023; 9:00633-2022. [PMID: 37465817 PMCID: PMC10351677 DOI: 10.1183/23120541.00633-2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 03/28/2023] [Indexed: 07/20/2023] Open
Abstract
New therapies are needed to prevent exacerbations, improve quality of life and slow disease progression in bronchiectasis. Inhibition of cathepsin C (CatC) activity has the potential to decrease activation of neutrophil-derived serine proteases in patients with bronchiectasis, thereby reducing airway inflammation, improving symptoms, reducing exacerbations and preventing further airway damage. Here we present the design of a phase 2 trial (Airleaf™; NCT05238675) assessing the efficacy and safety of a novel CatC inhibitor, BI 1291583, in adult patients with bronchiectasis. This multinational, randomised, double-blind, placebo-controlled, parallel-group, dose-finding study has a screening period of at least 6 weeks, a treatment period of 24-48 weeks and a follow-up period of 4 weeks. ∼240 adults with bronchiectasis of multiple aetiologies will be randomised to placebo once daily, or BI 1291583 1 mg once daily, 2.5 mg once daily or 5 mg once daily in a 2:1:1:2 ratio, stratified by Pseudomonas aeruginosa infection and maintenance use of macrolides. The primary efficacy objective is to evaluate the dose-response relationship for the three oral doses of BI 1291583 versus placebo on time to first pulmonary exacerbation up to Week 48 (the primary end-point). Efficacy will be assessed using exacerbations, patient-reported outcomes, measures of symptoms, sputum neutrophil elastase activity and pulmonary function testing. Safety assessment will include adverse event reporting, physical examination, monitoring of vital signs, safety laboratory parameters, 12-lead electrocardiogram, and periodontal and dermatological assessments. If efficacy and safety are demonstrated, results will support further investigation of BI 1291583 in phase 3 trials.
Collapse
Affiliation(s)
- James D. Chalmers
- Division of Molecular and Clinical Medicine, University of Dundee, Dundee, UK
| | - Abhya Gupta
- Boehringer Ingelheim International GmbH, Biberach, Germany
| | - Sanjay H. Chotirmall
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Department of Respiratory and Critical Care Medicine, Tan Tock Seng Hospital, Singapore, Singapore
| | | | - Peter Eickholz
- Department of Periodontology, Goethe University Frankfurt, Frankfurt, Germany
| | - Naoki Hasegawa
- Department of Infectious Diseases, Keio University, Tokyo, Japan
| | | | | | | | - Henrik Watz
- Pulmonary Research Institute at LungenClinic Grosshansdorf, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Grosshansdorf, Germany
| | | | | | - Wiebke Sauter
- Boehringer Ingelheim International GmbH, Biberach, Germany
| |
Collapse
|
20
|
The Role of MMPs in the Era of CFTR Modulators: An Additional Target for Cystic Fibrosis Patients? Biomolecules 2023; 13:biom13020350. [PMID: 36830719 PMCID: PMC9952876 DOI: 10.3390/biom13020350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/27/2023] [Accepted: 02/08/2023] [Indexed: 02/15/2023] Open
Abstract
Cystic fibrosis (CF) is a high-prevalence disease characterized by significant lung remodeling, responsible for high morbidity and mortality worldwide. The lung structural changes are partly due to proteolytic activity associated with inflammatory cells such as neutrophils and macrophages. Matrix metalloproteases (MMPs) are the major proteases involved in CF, and recent literature data focused on their potential role in the pathogenesis of the disease. In fact, an imbalance of proteases and antiproteases was observed in CF patients, resulting in dysfunction of protease activity and loss of lung homeostasis. Currently, many steps forward have been moved in the field of pharmacological treatment with the recent introduction of triple-combination therapy targeting the CFTR channel. Despite CFTR modulator therapy potentially being effective in up to 90% of patients with CF, there are still patients who are not eligible for the available therapies. Here, we introduce experimental drugs to provide updates on therapy evolution regarding a proportion of CF non-responder patients to current treatment, and we summarize the role of MMPs in pathogenesis and as future therapeutic targets of CF.
Collapse
|
21
|
Aliberti S, Gramegna A, Seia M, Malvestiti F, Mantero M, Sotgiu G, Simonetta E, Prati D, Paganini S, Ferrarotti I, Benzoni E, Stainer A, Santambrogio M, Saderi L, Balderacchi AM, Valenti L, Corsico AG, Amati F, Blasi F. Alpha 1-Antitrypsin Inherited Variants in Patients With Bronchiectasis. Arch Bronconeumol 2023:S0300-2896(23)00009-1. [PMID: 36710175 DOI: 10.1016/j.arbres.2023.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/30/2022] [Accepted: 01/05/2023] [Indexed: 01/19/2023]
Affiliation(s)
- Stefano Aliberti
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Milan, Italy; IRCCS Humanitas Research Hospital, Respiratory Unit, Via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Andrea Gramegna
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Respiratory Unit and Cystic Fibrosis Adult Center, Milan, Italy; Università degli Studi di Milano, Department of Pathophysiology and Transplantation, Milan, Italy
| | - Manuela Seia
- Laboratory of Medical Genetics, IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Francesco Malvestiti
- Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico Milan, Italy
| | - Marco Mantero
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Respiratory Unit and Cystic Fibrosis Adult Center, Milan, Italy; Università degli Studi di Milano, Department of Pathophysiology and Transplantation, Milan, Italy
| | - Giovanni Sotgiu
- Clinical Epidemiology and Medical Statistics Unit, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Edoardo Simonetta
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Milan, Italy; IRCCS Humanitas Research Hospital, Respiratory Unit, Via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Daniele Prati
- Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico Milan, Italy
| | - Stefania Paganini
- Laboratory of Medical Genetics, IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Ilaria Ferrarotti
- Center for Diagnosis of Inherited Alpha1-antitrypsin Deficiency, Department of Internal Medicine and Therapeutics, University of Pavia, and Pneumology Unit IRCCS San Matteo Hospital Foundation, University of Pavia, Pavia, Italy
| | - Elena Benzoni
- Laboratory of Medical Genetics, IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Anna Stainer
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Milan, Italy; IRCCS Humanitas Research Hospital, Respiratory Unit, Via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Martina Santambrogio
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Respiratory Unit and Cystic Fibrosis Adult Center, Milan, Italy
| | - Laura Saderi
- Clinical Epidemiology and Medical Statistics Unit, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Alice M Balderacchi
- Center for Diagnosis of Inherited Alpha1-antitrypsin Deficiency, Department of Internal Medicine and Therapeutics, University of Pavia, and Pneumology Unit IRCCS San Matteo Hospital Foundation, University of Pavia, Pavia, Italy
| | - Luca Valenti
- Università degli Studi di Milano, Department of Pathophysiology and Transplantation, Milan, Italy; Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico Milan, Italy
| | - Angelo G Corsico
- Center for Diagnosis of Inherited Alpha1-antitrypsin Deficiency, Department of Internal Medicine and Therapeutics, University of Pavia, and Pneumology Unit IRCCS San Matteo Hospital Foundation, University of Pavia, Pavia, Italy
| | - Francesco Amati
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Milan, Italy; IRCCS Humanitas Research Hospital, Respiratory Unit, Via Manzoni 56, 20089 Rozzano, Milan, Italy.
| | - Francesco Blasi
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Respiratory Unit and Cystic Fibrosis Adult Center, Milan, Italy; Università degli Studi di Milano, Department of Pathophysiology and Transplantation, Milan, Italy
| |
Collapse
|
22
|
Bigalke A, Sponholz C, Schnabel C, Bauer M, Kiehntopf M. Multiplex quantification of C-terminal alpha-1-antitrypsin peptides provides a novel approach for characterizing systemic inflammation. Sci Rep 2022; 12:3844. [PMID: 35264629 PMCID: PMC8907207 DOI: 10.1038/s41598-022-07752-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 02/22/2022] [Indexed: 01/09/2023] Open
Abstract
C-terminal peptides (CAAPs) of the highly abundant serine protease alpha-1-antitrypsin (A1AT) have been identified at various lengths in several human materials and have been proposed to serve as putative biomarkers for a variety of diseases. CAAPs are enzymatically formed and these enzymatic activities are often associated with excessive immune responses (e.g. sepsis, allergies). However, most of those CAAPs have been either detected using in vitro incubation experiments or in human materials which are not easily accessible. To gain a comprehensive understanding about the occurrence and function of CAAPs in health and disease, a LC-MS/MS method for the simultaneous detection of nine CAAPs was developed and validated for human plasma (EDTA and lithium-heparin) and serum. Using this newly developed method, we were able to detect and quantify five CAAPs in healthy individuals thereby providing an initial proof for the presence of C36, C37, C40 and C44 in human blood. Concentrations of four CAAPs in a clinical test cohort of patients suffering from sepsis were significantly higher compared to healthy controls. These results reveal that in addition to C42 other fragments of A1AT seem to play a crucial role during systemic infections. The proposed workflow is simple, rapid and robust; thus this method could be used as diagnostic tool in routine clinical chemistry as well as for research applications for elucidating the diagnostic potential of CAAPs in numerous diseases. To this end, we also provide an overview about the current state of knowledge for CAAPs identified in vitro and in vivo.
Collapse
Affiliation(s)
- Arite Bigalke
- grid.275559.90000 0000 8517 6224Department of Clinical Chemistry and Laboratory Medicine, Jena University Hospital, Jena, Germany
| | - Christoph Sponholz
- grid.275559.90000 0000 8517 6224Department of Anesthesiology and Intensive Care Therapy, Jena University Hospital, Jena, Germany
| | - Claudia Schnabel
- grid.275559.90000 0000 8517 6224Department of Clinical Chemistry and Laboratory Medicine, Jena University Hospital, Jena, Germany
| | - Michael Bauer
- grid.275559.90000 0000 8517 6224Department of Anesthesiology and Intensive Care Therapy, Jena University Hospital, Jena, Germany
| | - Michael Kiehntopf
- grid.275559.90000 0000 8517 6224Department of Clinical Chemistry and Laboratory Medicine, Jena University Hospital, Jena, Germany
| |
Collapse
|
23
|
Blanter M, Cockx M, Wittebols L, Abouelasrar Salama S, De Bondt M, Berghmans N, Pörtner N, Vanbrabant L, Lorent N, Gouwy M, Boon M, Struyf S. Sputum from patients with primary ciliary dyskinesia contains high numbers of dysfunctional neutrophils and inhibits efferocytosis. Respir Res 2022; 23:359. [PMID: 36528664 PMCID: PMC9758951 DOI: 10.1186/s12931-022-02280-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Primary ciliary dyskinesia (PCD) is a genetic disorder characterized by recurrent airway infection and inflammation. There is no cure for PCD and to date there are no specific treatments available. Neutrophils are a crucial part of the immune system and are known to be dysfunctional in many inflammatory diseases. So far, the role of the neutrophils in PCD airways is largely unknown. The purpose of this study was to investigate the phenotype and function of airway neutrophils in PCD, and compare them to blood neutrophils. METHODS Paired peripheral blood and spontaneously expectorated sputum samples from patients with PCD (n = 32) and a control group of patients with non-PCD, non-cystic fibrosis bronchiectasis (n = 5) were collected. The expression of neutrophil-specific surface receptors was determined by flow cytometry. Neutrophil function was assessed by measuring the extent of actin polymerization, production of reactive oxygen species (ROS) and release of neutrophil extracellular traps (NETs) in response to activating stimuli. RESULTS Sputum neutrophils displayed a highly activated phenotype and were unresponsive to stimuli that would normally induce ROS production, actin polymerization and the expulsion of NETs. In addition, PCD sputum displayed high activity of neutrophil elastase, and impaired the efferocytosis by healthy donor macrophages. CONCLUSIONS Sputum neutrophils in PCD are dysfunctional and likely contribute to ongoing inflammation in PCD airways. Further research should focus on anti-inflammatory therapies and stimulation of efferocytosis as a strategy to treat PCD.
Collapse
Affiliation(s)
- Marfa Blanter
- grid.5596.f0000 0001 0668 7884Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, University of Leuven, 3000 Leuven, Belgium
| | - Maaike Cockx
- grid.5596.f0000 0001 0668 7884Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, University of Leuven, 3000 Leuven, Belgium
| | - Liesel Wittebols
- grid.5596.f0000 0001 0668 7884Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, University of Leuven, 3000 Leuven, Belgium
| | - Sara Abouelasrar Salama
- grid.5596.f0000 0001 0668 7884Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, University of Leuven, 3000 Leuven, Belgium
| | - Mirre De Bondt
- grid.5596.f0000 0001 0668 7884Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, University of Leuven, 3000 Leuven, Belgium
| | - Nele Berghmans
- grid.5596.f0000 0001 0668 7884Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, University of Leuven, 3000 Leuven, Belgium
| | - Noëmie Pörtner
- grid.5596.f0000 0001 0668 7884Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, University of Leuven, 3000 Leuven, Belgium
| | - Lotte Vanbrabant
- grid.5596.f0000 0001 0668 7884Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, University of Leuven, 3000 Leuven, Belgium
| | - Natalie Lorent
- grid.410569.f0000 0004 0626 3338Pneumology and Cystic Fibrosis Unit, Department of Pneumology, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Mieke Gouwy
- grid.5596.f0000 0001 0668 7884Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, University of Leuven, 3000 Leuven, Belgium
| | - Mieke Boon
- grid.410569.f0000 0004 0626 3338Pediatric Pneumology and Cystic Fibrosis Unit, Department of Pediatrics, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Sofie Struyf
- grid.5596.f0000 0001 0668 7884Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, University of Leuven, 3000 Leuven, Belgium
| |
Collapse
|