1
|
Pavord ID, Brightling CE, Korn S, Martin NL, Ponnarambil SS, Molfino NA, Parnes JR, Ambrose CS. Tezepelumab can Restore Normal Lung Function in Patients with Severe, Uncontrolled Asthma: Pooled Results from the PATHWAY and NAVIGATOR Studies. Pulm Ther 2025; 11:315-325. [PMID: 40285963 PMCID: PMC12102424 DOI: 10.1007/s41030-025-00294-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 03/18/2025] [Indexed: 04/29/2025] Open
Abstract
INTRODUCTION This post hoc analysis assessed the ability of tezepelumab treatment to restore normal lung function in patients with severe, uncontrolled asthma with abnormal lung function at baseline pooled from the PATHWAY and NAVIGATOR studies. METHODS PATHWAY and NAVIGATOR were multicentre, randomized, double-blind, placebo-controlled studies. Patients (12-80 years old) included in this analysis received tezepelumab 210 mg subcutaneously every 4 weeks or matched placebo for 52 weeks. Patients had a percent predicted pre-bronchodilator (BD) forced expiratory volume in 1 s (FEV1) of < 80% (< 90% for adolescents in NAVIGATOR) at screening. The change from baseline to week 52 in pre-BD FEV1 was assessed by baseline percent predicted pre-BD FEV1 subgroup [abnormal (< 80%) and normal (≥ 80%)]. The proportion of patients with abnormal lung function at baseline who achieved normal lung function at week 52 was assessed overall and by biomarker level and disease duration subgroups. RESULTS Of the 665 and 669 patients who received tezepelumab or placebo, respectively, 564 and 569 had abnormal lung function at baseline. Tezepelumab improved the pre-BD FEV1 from baseline to week 52 versus placebo by 0.14 L [95% confidence interval (CI) 0.09-0.19] and 0.13 L (95% CI 0.01-0.24) in patients with abnormal and normal lung function at baseline, respectively. A higher proportion of tezepelumab than placebo recipients with abnormal lung function at baseline achieved normal lung function at week 52 (17.2% vs. 9.9%, respectively). Among tezepelumab recipients, those with higher levels of type 2 inflammatory biomarkers and a shorter duration of disease at baseline were more likely to achieve normal lung function at week 52. CONCLUSION In patients with severe, uncontrolled asthma, a greater proportion of tezepelumab than placebo recipients with abnormal lung function at baseline achieved normal lung function at week 52. TRIAL REGISTRATION PATHWAY: NCT02054130; NAVIGATOR: NCT03347279.
Collapse
Affiliation(s)
- Ian D Pavord
- Respiratory Medicine, National Institute for Health and Care Research Oxford Biomedical Research Centre, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| | - Christopher E Brightling
- Institute for Lung Health, National Institute for Health and Care Research Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
| | - Stephanie Korn
- IKF Pneumologie Mainz, Mainz, Germany, and Thoraxklinik Heidelberg, Heidelberg, Germany
| | - Nicole L Martin
- Biometrics, Late-Stage Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cytel Inc., Waltham, MA, USA
| | - Sandhia S Ponnarambil
- Late-Stage Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | | | | | - Christopher S Ambrose
- Respiratory and Immunology, BioPharmaceuticals Medical, AstraZeneca, Gaithersburg, MD, USA
| |
Collapse
|
2
|
Mettler SK, Nardelli P, Campo MI, San José Estépar R, Manapragada PP, Abozeed M, Aziz MU, Zahid M, Grumley S, Nath HP, Yen A, Sonavane S, Clarenbach CF, Wang W, Pistenmaa CL, Washko GR, Estépar RSJ, Cho MH, Diaz AA. Longitudinal Changes in Airway Mucus Plugs and FEV 1 in COPD. N Engl J Med 2025; 392:1973-1975. [PMID: 40367384 DOI: 10.1056/nejmc2502456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/16/2025]
Affiliation(s)
| | | | | | | | | | | | | | - Mohd Zahid
- University of Alabama at Birmingham, Birmingham
| | | | | | - Andrew Yen
- University of California, San Diego, La Jolla
| | | | | | - Wei Wang
- Brigham and Women's Hospital, Boston
| | | | | | | | | | | |
Collapse
|
3
|
Götschke J, Walter J, Leuschner G, Gerckens M, Götschke M, Mertsch P, Mümmler C, Lenoir A, Barnikel M, Dinkel J, Behr J, Kneidinger N, Spiro JE, Milger K. Mucus Plug Score Predicts Clinical and Pulmonary Function Response to Biologic Therapy in Patients With Severe Asthma. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2025; 13:1110-1122.e1. [PMID: 39826645 DOI: 10.1016/j.jaip.2025.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 01/05/2025] [Accepted: 01/06/2025] [Indexed: 01/22/2025]
Abstract
BACKGROUND Mucus plugging has been identified as an important feature of severe asthma contributing to airway obstruction and disease severity. Recently, improvement in mucus plugging has been found on treatment with several biologic therapies. OBJECTIVES To analyze associations of baseline characteristic with the mucus plugging score (MPS) and to determine whether the MPS at baseline predicts the clinical and functional response to biologic treatment in patients with severe asthma. METHODS We retrospectively analyzed biologic-naive patients with a suitable computed tomography scan available at baseline. We calculated the MPS and analyzed correlations with baseline parameters and improvements in biomarkers, pulmonary function, and clinical parameters after 4 months of biologic therapy. RESULTS We included 113 patients in the baseline cohort, 101 patients of whom had sufficient data after 4 months of biologic therapy for the follow-up analysis. Computed tomography showed mucus plugging in 77% of patients (median MPS, 4). Multivariate regression analysis showed a correlation of MPS with lower FEV1 (ρ = -0.24; P = .009) and diffusing capacity for carbon monoxide (ρ = -0.26; P = .01), and higher FeNO (ρ = .36; P = .0003) at baseline. Patients received treatment with anti-IgE (8.8%), anti-IL-5 (27.4%), anti-IL-5R (37.2%), anti-IL-4R (25.7%), and anti-thymic stromal lymphopoietin (0.9%) in clinical routine. Baseline MPS correlated with improvements in FEV1 (β = 0.72; P = .01) and Asthma Control Test (β = 0.24; P = .001) in multivariate regression analysis. CONCLUSION Our study suggests that a higher MPS correlates with worse pulmonary function at baseline but also predicts a larger clinical and pulmonary function response to biologic therapies in severe asthma.
Collapse
Affiliation(s)
- Jeremias Götschke
- Department of Medicine V, LMU University Hospital, LMU Munich, Munich, Germany; Comprehensive Pneumology Center Munich, German Center for Lung Research (DZL), Munich, Germany
| | - Julia Walter
- Department of Medicine V, LMU University Hospital, LMU Munich, Munich, Germany; Comprehensive Pneumology Center Munich, German Center for Lung Research (DZL), Munich, Germany
| | - Gabriele Leuschner
- Department of Medicine V, LMU University Hospital, LMU Munich, Munich, Germany; Comprehensive Pneumology Center Munich, German Center for Lung Research (DZL), Munich, Germany
| | - Michael Gerckens
- Department of Medicine V, LMU University Hospital, LMU Munich, Munich, Germany; Comprehensive Pneumology Center Munich, German Center for Lung Research (DZL), Munich, Germany; Institute of Lung Health and Immunity, Comprehensive Pneumology Center, Helmholtz Munich, Member of the German Center of Lung Research (DZL), Munich, Germany
| | - Melanie Götschke
- Department of Medicine V, LMU University Hospital, LMU Munich, Munich, Germany; Comprehensive Pneumology Center Munich, German Center for Lung Research (DZL), Munich, Germany
| | - Pontus Mertsch
- Department of Medicine V, LMU University Hospital, LMU Munich, Munich, Germany; Comprehensive Pneumology Center Munich, German Center for Lung Research (DZL), Munich, Germany
| | - Carlo Mümmler
- Department of Medicine V, LMU University Hospital, LMU Munich, Munich, Germany; Comprehensive Pneumology Center Munich, German Center for Lung Research (DZL), Munich, Germany; Institute of Lung Health and Immunity, Comprehensive Pneumology Center, Helmholtz Munich, Member of the German Center of Lung Research (DZL), Munich, Germany
| | - Alexandra Lenoir
- Department of Medicine V, LMU University Hospital, LMU Munich, Munich, Germany; Comprehensive Pneumology Center Munich, German Center for Lung Research (DZL), Munich, Germany
| | - Michaela Barnikel
- Department of Medicine V, LMU University Hospital, LMU Munich, Munich, Germany; Comprehensive Pneumology Center Munich, German Center for Lung Research (DZL), Munich, Germany
| | - Julien Dinkel
- Department of Radiology, LMU University Hospital, LMU Munich, Munich, Germany, Member of the German Center for Lung Research (DZL)
| | - Jürgen Behr
- Department of Medicine V, LMU University Hospital, LMU Munich, Munich, Germany; Comprehensive Pneumology Center Munich, German Center for Lung Research (DZL), Munich, Germany
| | - Nikolaus Kneidinger
- Department of Medicine V, LMU University Hospital, LMU Munich, Munich, Germany; Comprehensive Pneumology Center Munich, German Center for Lung Research (DZL), Munich, Germany; Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Judith Eva Spiro
- Department of Radiology, LMU University Hospital, LMU Munich, Munich, Germany, Member of the German Center for Lung Research (DZL)
| | - Katrin Milger
- Department of Medicine V, LMU University Hospital, LMU Munich, Munich, Germany; Comprehensive Pneumology Center Munich, German Center for Lung Research (DZL), Munich, Germany; Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, Graz, Austria.
| |
Collapse
|
4
|
Carter C, Torre IB, Blackburn S, Nwankwo L, Semple T, Rawal B, Armstrong-James D, Patel PH, Shah A. Real-World Effectiveness of Biologic Therapy in Allergic Bronchopulmonary Aspergillosis. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2025; 13:1094-1102.e1. [PMID: 40088970 DOI: 10.1016/j.jaip.2025.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 02/28/2025] [Accepted: 03/05/2025] [Indexed: 03/17/2025]
Abstract
BACKGROUND Allergic bronchopulmonary aspergillosis (ABPA) is characterized by a severe hypersensitivity reaction to Aspergillus species. Current treatment relies on oral corticosteroids (OCS) and triazole antifungal therapy, but there is increasing evidence of the benefits of biologic therapies targeting type 2 inflammatory pathways. OBJECTIVE To assess the real-world effectiveness of biologic therapies in patients with ABPA. METHODS We performed a large retrospective single-center analysis of patients with ABPA as defined by the modified International Society for Human and Animal Mycology (ISHAM) criteria between 2014 and 2022. Baseline characteristics were recorded. Clinical outcomes were assessed at 12 months after commencement of a biologic including symptom scores, exacerbation frequency, corticosteroid use, and multidisciplinary team consensus of effectiveness. RESULTS A total of 74 patients received a biologic, of whom 32% (n = 24) received anti-IgE therapy, 65% (n = 48) anti-IL5/5Rα therapy, and 3% (n = 2) anti-IL4-Rα therapy. Of the total, 65% (n = 48) patients were deemed to have a successful response at 12 months with a ≥50% reduction in OCS use and 35% (n = 26) stopped or changed biologic during the follow-up period because of failed clinical response (n = 21), side effects (n = 4), or medical comorbidities (n = 1). There was a significant reduction in the 6-item Asthma Control Questionnaire score (P < .0001), exacerbation rate over 12 months (P < .0001), and maintenance OCS use (P = .0173). Univariate analysis revealed that mucus plugging was associated with nonresponse to biologic therapy (P = .0189). CONCLUSION Biologic therapies are effective in a number of patients with ABPA. However, further prospective clinical trials are required to determine the effectiveness and which phenotypes likely to respond. These data nevertheless increase the evidence base for biologics in ABPA.
Collapse
Affiliation(s)
- Charlotte Carter
- Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Irene Berrar Torre
- Department of Respiratory Medicine, Royal Brompton Hospital, Guy's & St. Thomas' NHS Foundation Trust, London, United Kingdom
| | - Sophia Blackburn
- Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Lisa Nwankwo
- Department of Pharmacy, Royal Brompton Hospital, Guy's & St. Thomas' NHS Foundation Trust, London, United Kingdom
| | - Tom Semple
- Department of Diagnostic Radiology, Royal Brompton Hospital, Guy's & St. Thomas' NHS Foundation Trust, London, United Kingdom; National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Bhavin Rawal
- Department of Diagnostic Radiology, Royal Brompton Hospital, Guy's & St. Thomas' NHS Foundation Trust, London, United Kingdom
| | - Darius Armstrong-James
- Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, United Kingdom; Department of Respiratory Medicine, Royal Brompton Hospital, Guy's & St. Thomas' NHS Foundation Trust, London, United Kingdom
| | - Pujan H Patel
- Department of Respiratory Medicine, Royal Brompton Hospital, Guy's & St. Thomas' NHS Foundation Trust, London, United Kingdom
| | - Anand Shah
- Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, United Kingdom; Department of Respiratory Medicine, Royal Brompton Hospital, Guy's & St. Thomas' NHS Foundation Trust, London, United Kingdom.
| |
Collapse
|
5
|
Kai Y, Hishida Y. Three-dimensional bronchial tree visualization in exercise-induced severe asthma following tezepelumab treatment. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. GLOBAL 2025; 4:100398. [PMID: 39896213 PMCID: PMC11783099 DOI: 10.1016/j.jacig.2024.100398] [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: 08/02/2024] [Revised: 10/04/2024] [Accepted: 10/06/2024] [Indexed: 02/04/2025]
Abstract
Airway hyperresponsiveness, a key feature of asthma, is associated with exercise-induced asthma. Tezepelumab was reported to reduce airway hyperresponsiveness. Tezepelumab was confirmed through 3-dimensional bronchial tree visualization to be effective for exercise-induced asthma, reducing the need for a short-acting β2 agonist.
Collapse
Affiliation(s)
- Yoshiro Kai
- Department of Respiratory Medicine, Minami-Nara General Medical Center, Nara, Japan
- Department of Respiratory Medicine, Nara Medical University, Nara, Japan
| | - Yuichi Hishida
- Department of Radiology, Minami-Nara General Medical Center, Nara, Japan
| |
Collapse
|
6
|
Menzella F, Cottini M, Lombardi C, Senna G, Chan R, Bosi A, Bortoli M, Tonin S, Corsi L, Rastelli A, Marchi MR. A real-world study on tezepelumab effectiveness in severe asthma focusing on small airway dysfunction. Respir Med 2025; 241:108054. [PMID: 40147569 DOI: 10.1016/j.rmed.2025.108054] [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: 02/05/2025] [Revised: 03/23/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND Severe asthma (SA) is a complex condition often involving small airway dysfunction (SAD). Tezepelumab has demonstrated efficacy in clinical trials, but real-world evidence is scarce, and its impact on SAD remains unexplored. OBJECTIVE This prospective study evaluated the effectiveness of tezepelumab in patients with SA, stratified by the SAD presence and asthma phenotype (type 2-high vs type 2-low). METHODS Seventeen SA patients received tezepelumab. A range of clinical and laboratory outcomes were assessed, including annualized asthma exacerbation rate (AAER), lung function, and oral corticosteroids (OCS) use. Respiratory parameters were assessed using spirometry, body plethysmography, and forced oscillation technique (FOT). RESULTS After 6 months of treatment, tezepelumab significantly reduced median AAER (from 5.0 to 0.0, p = 0.001) and OCS dose (from 14.6 mg/day to 0.0 mg/day, p < 0.001), alongside a marked reduction in median blood eosinophil count (from 130 to 60 cells/mm3, p = 0.032). Among respiratory parameters, total resistance, measured by body plethysmography, improved significantly in the overall population (median values from 0.49 to 0.37 KPa L s-1, p = 0.005). Spirometry and FOT measures, including total reactance (p = 0.018) and tidal expiratory flow limitation (p = 0.043), improved only in patients with SAD. CONCLUSION Tezepelumab significantly reduced exacerbations and improved asthma control, positively impacting respiratory parameters and small airway function in patients with SAD. These findings support SAD as a treatable trait, highlighting the importance of integrating advanced diagnostic tools, such as body plethysmography and FOT, into routine clinical practice.
Collapse
Affiliation(s)
- Francesco Menzella
- Pulmonology Unit, S. Valentino Hospital, Montebelluna (TV), AULSS2 Marca Trevigiana, Italy.
| | | | - Carlo Lombardi
- Departmental Unit of Allergology, Immunology and Pulmonary Diseases, Fondazione Poliambulanza, Brescia, Italy
| | - Gianenrico Senna
- Asthma, Allergy and Clinical Immunology Section, University Hospital of Verona, Verona, Italy; Department of Medicine, University of Verona, Verona, Italy
| | - Rory Chan
- University of Dundee School of Medicine, Dundee, UK
| | - Annamaria Bosi
- Pulmonology Unit, S. Valentino Hospital, Montebelluna (TV), AULSS2 Marca Trevigiana, Italy
| | - Michela Bortoli
- Department of Pneumology, Hospital Cittadella (PD), ULSS6 Euganea, Cittadella, Italy
| | - Silvia Tonin
- Pulmonology Unit, S. Valentino Hospital, Montebelluna (TV), AULSS2 Marca Trevigiana, Italy
| | - Lorenzo Corsi
- Pulmonology Unit, S. Valentino Hospital, Montebelluna (TV), AULSS2 Marca Trevigiana, Italy
| | - Andrea Rastelli
- Department of Pneumology, Hospital Cittadella (PD), ULSS6 Euganea, Cittadella, Italy
| | - Maria Rita Marchi
- Department of Pneumology, Hospital Cittadella (PD), ULSS6 Euganea, Cittadella, Italy
| |
Collapse
|
7
|
Fedosenko S, Venegas Garrido C, Nair P. Recent advances in asthma mucus biology and emerging treatment strategies. Curr Opin Pulm Med 2025; 31:251-261. [PMID: 40047213 DOI: 10.1097/mcp.0000000000001167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2025]
Abstract
PURPOSE OF REVIEW To describe the recent advances in the pathobiology and treatment of mucus hypersecretion in asthma, a critical factor contributing to airway obstruction, inflammation, and impaired lung function. RECENT FINDINGS Significant progress has been made in understanding how mucin protein regulation, mucus viscosity, and adhesion are affected by cytokine-driven inflammation, especially interleukin-13, and defects in ion transport mechanisms. Advances in imaging techniques, such as multidetector computed tomography (MDCT) and hyperpolarized gas MRI, allow for a more precise assessment of mucus plugging and associated ventilation defects. Emerging therapies, including biologicals targeting type-2 (T2) inflammation, and novel mucolytics aimed at modifying mucus properties and secretion, offer promising effects in reducing mucus in severe asthmatics. SUMMARY The growing understanding of mucus biology and the development of advanced imaging and therapeutic strategies could significantly improve the management of mucus-related complications in asthma. By targeting mucus characteristics, these findings support future approaches to reduce airway obstruction, enhance lung function, and improve clinical outcomes in patients with severe asthma. A deeper understanding of the glycobiology of mucus is critical to develop new therapies.
Collapse
Affiliation(s)
- Sergey Fedosenko
- Division of Respirology, Department of Medicine, St Joseph's Healthcare and McMaster University, Hamilton, Ontario, Canada
| | | | | |
Collapse
|
8
|
Michils A, Hackx M, Mlynarski L, Haccuria A, Perez-Bogerd S, Malinovschi A, Van Muylem A. How FEV 1 Improvement Induced by Anti-IL-5 in Severe Type-2 Asthma Is Linked to Mucus Plugs Clearance. Allergy 2025; 80:1143-1145. [PMID: 39698780 DOI: 10.1111/all.16441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 11/13/2024] [Accepted: 12/05/2024] [Indexed: 12/20/2024]
Affiliation(s)
- Alain Michils
- Chest Department, Erasme University Hospital, Université libre de Bruxelles, Brussels, Belgium
| | - Maxime Hackx
- Radiology Department, Erasme University Hospital, Université libre de Bruxelles, Brussels, Belgium
| | - Lucas Mlynarski
- Chest Department, Erasme University Hospital, Université libre de Bruxelles, Brussels, Belgium
| | - Amaryllis Haccuria
- Chest Department, Erasme University Hospital, Université libre de Bruxelles, Brussels, Belgium
| | - Silvia Perez-Bogerd
- Chest Department, Erasme University Hospital, Université libre de Bruxelles, Brussels, Belgium
| | - Andreï Malinovschi
- Department of Medical Sciences, Clinical Physiology, Uppsala University, Uppsala, Sweden
| | - Alain Van Muylem
- Chest Department, Erasme University Hospital, Université libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
9
|
Dorscheid D, Gauvreau GM, Georas SN, Hiemstra PS, Varricchi G, Lambrecht BN, Marone G. Airway epithelial cells as drivers of severe asthma pathogenesis. Mucosal Immunol 2025:S1933-0219(25)00029-7. [PMID: 40154790 DOI: 10.1016/j.mucimm.2025.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 01/31/2025] [Accepted: 03/19/2025] [Indexed: 04/01/2025]
Abstract
Our understanding of the airway epithelium's role in driving asthma pathogenesis has evolved over time. From being regarded primarily as a physical barrier that could be damaged via inflammation, the epithelium is now known to actively contribute to asthma development through interactions with the immune system. The airway epithelium contains multiple cell types with specialized functions spanning barrier action, mucociliary clearance, immune cell recruitment, and maintenance of tissue homeostasis. Environmental insults may cause direct or indirect injury to the epithelium leading to impaired barrier function, epithelial remodelling, and increased release of inflammatory mediators. In severe asthma, the epithelial barrier repair process is inhibited and the response to insults is exaggerated, driving downstream inflammation. Genetic and epigenetic mechanisms also maintain dysregulation of the epithelial barrier, adding to disease chronicity. Here, we review the role of the airway epithelium in severe asthma and how targeting the epithelium can contribute to asthma treatment.
Collapse
Affiliation(s)
- Del Dorscheid
- Centre for Heart Lung Innovation, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Gail M Gauvreau
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Steve N Georas
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Pieter S Hiemstra
- Department of Pulmonology, Leiden University Medical Center, Leiden, the Netherlands
| | - Gilda Varricchi
- Department of Translational Medical Sciences (DiSMeT) and Center for Basic and Clinical Immunology Research (CISI), School of Medicine, University of Naples Federico II, Naples, Italy; Institute of Experimental Endocrinology and Oncology (IEOS), National Research Council, Naples, Italy
| | - Bart N Lambrecht
- Center for Inflammation Research, Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium.
| | - Gianni Marone
- Department of Translational Medical Sciences (DiSMeT) and Center for Basic and Clinical Immunology Research (CISI), School of Medicine, University of Naples Federico II, Naples, Italy; Institute of Experimental Endocrinology and Oncology (IEOS), National Research Council, Naples, Italy.
| |
Collapse
|
10
|
Castro M, Papi A, Porsbjerg C, Lugogo NL, Brightling CE, González-Barcala FJ, Bourdin A, Ostrovskyy M, Staevska M, Chou PC, Duca L, Pereira AM, Fogarty C, Nadama R, Zhang M, Rodrigues A, Soler X, Sacks HJ, Deniz Y, Rowe PJ, de Prado Gómez L, Jacob-Nara JA. Effect of dupilumab on exhaled nitric oxide, mucus plugs, and functional respiratory imaging in patients with type 2 asthma (VESTIGE): a randomised, double-blind, placebo-controlled, phase 4 trial. THE LANCET. RESPIRATORY MEDICINE 2025; 13:208-220. [PMID: 39947221 DOI: 10.1016/s2213-2600(24)00362-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 03/08/2025]
Abstract
BACKGROUND Asthma is a respiratory disease characterised by chronic airway inflammation and mucus hypersecretion. VESTIGE used functional respiratory imaging to assess changes in airway structure and function, including mucus plugging, in response to dupilumab. METHODS VESTIGE was a randomised, double-blind, placebo-controlled, phase 4 trial done at 72 research sites or academic centres in 14 countries. We recruited adult patients (aged 18-70 years) with physician-diagnosed, uncontrolled, moderate-to-severe type 2 asthma (blood eosinophil count ≥300 cells/μL and fractional exhaled nitric oxide [FeNO] ≥25 parts per billion [ppb]) being treated with medium-dose to high-dose inhaled corticosteroids combined with other controller medications. Patients were randomly assigned (2:1; block size of 6) via interactive voice-web response technology to receive add-on dupilumab 300 mg subcutaneously once every 2 weeks or volume-matched placebo up to week 24. Randomisation was stratified by inhaled corticosteroids dose level and region (eastern Europe vs the rest of the world). Participants and investigators, including those assessing outcomes, were masked to group assignment. The primary endpoints were the proportion of patients with a FeNO concentration below 25 ppb at week 24, and percentage change from baseline to week 24 in airway volumes (specific regional airway volumes corrected for lung volume, [s]iVaw) at total lung capacity (TLC), both assessed in the intention-to-treat population. Safety was assessed in all patients who received at least one dose of study drug or placebo. The trial is registered with ClinicalTrials.gov, NCT04400318, and is completed. FINDINGS Patient recruitment occurred from July 18, 2020, to Jan 6, 2023. Patients (mean age 50·4 years [SD 12·6]; 68 [62%] female and 41 [38%] male) were randomly assigned to receive dupilumab 300 mg (n=72) or placebo (n=37). At week 24, patients in the dupilumab group were significantly more likely than those in the placebo group to have a FeNO concentration below 25 ppb (41 [57%] of 72 patients vs four [11%] of 37; odds ratio: 9·8 [95% CI 3·1 to 30·8]; p<0·001). Treatment with dupilumab versus placebo led to a numerical increase in (s)iVaw at TLC from baseline to week 24, although the difference was not significant (least squares [LS] mean percentage change from baseline to week 24: 19·7% [SE 8·1] for dupilumab and -2·0% [11·5] for placebo; LS mean difference vs placebo: 21·8% [95% CI -7·7 to 51·3]; p=0·14). Safety was consistent with the reported safety profile for dupilumab. Treatment-emergent adverse events related to study intervention were reported in 11 (15%) of 72 patients who received dupilumab and four (11%) of 37 who received placebo; no deaths occurred during the intervention period. INTERPRETATION The full results of this study indicate that dupilumab reduced airway inflammation and mucus plugging, and improved airway volume and flow, corresponding to improved lung function and asthma control. This study highlights the potential of imaging technology to assess disease burden, monitor progression, and evaluate therapeutic responses, which can provide valuable insights to guide clinical decision making for patients with uncontrolled, moderate-to-severe type 2 asthma. FUNDING Sanofi and Regeneron Pharmaceuticals.
Collapse
Affiliation(s)
- Mario Castro
- Department of Medicine, University of Kansas School of Medicine, Kansas City, KS, USA.
| | - Alberto Papi
- Department of Cardiorespiratory Medicine, Respiratory Medicine Unit, University of Ferrara, S Anna University Hospital, Ferrara, Italy
| | - Celeste Porsbjerg
- Department of Respiratory Medicine and Infectious Diseases, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
| | - Njira L Lugogo
- Michigan Medicine Asthma Program, University of Michigan, Ann Arbor, MI, USA
| | - Christopher E Brightling
- Institute for Lung Health, NIHR Leicester Biomedical Research Centre, Department of Respiratory Sciences, University of Leicester, Leicester, UK
| | - Francisco-Javier González-Barcala
- Neumología, Hospital Clínico Universitario de Santiago de Compostela-Grupo de Investigación TRIAD, Santiago de Compostela, Spain; Universidad de Santiago de Compostela, Santiago de Compostela, Spain
| | - Arnaud Bourdin
- Department of Respiratory Diseases, PhyMedExp, University of Montpellier, INSERM CNRS, Montpellier, France
| | - Mykola Ostrovskyy
- Ivano-Frankivsk National Medical University, Ivano-Frankivsk, Ukraine
| | | | | | | | - Ana Margarida Pereira
- Allergy Unit, Instituto and Hospital CUF, Porto, Portugal; Patient Centered Innovation and Technologies, Center for Health Technology and Services Research, Faculty of Medicine, University of Porto, Porto, Portugal
| | | | - Rufai Nadama
- King Khalid University Hospital, Riyadh, Saudi Arabia
| | | | | | | | | | - Yamo Deniz
- Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | | | | | | |
Collapse
|
11
|
Alladina J, Medoff BD, Cho JL. Innate Immunity and Asthma Exacerbations: Insights From Human Models. Immunol Rev 2025; 330:e70016. [PMID: 40087882 PMCID: PMC11922041 DOI: 10.1111/imr.70016] [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: 01/13/2025] [Revised: 02/14/2025] [Accepted: 02/28/2025] [Indexed: 03/17/2025]
Abstract
Asthma is a common chronic respiratory disease characterized by the presence of airway inflammation, airway hyperresponsiveness, and mucus hypersecretion. Repeated asthma exacerbations can lead to progressive airway remodeling and irreversible airflow obstruction. Thus, understanding and preventing asthma exacerbations are of paramount importance. Although multiple endotypes exist, asthma is most often driven by type 2 airway inflammation. New therapies that target specific type 2 mediators have been shown to reduce the frequency of asthma exacerbations but are incompletely effective in a significant number of asthmatics. Furthermore, it remains unknown whether current treatments lead to sustained changes in the airway or if targeting additional pathways may be necessary to achieve asthma remission. Activation of innate immunity is the initial event in the inflammatory sequence that occurs during an asthma exacerbation. However, there continue to be critical gaps in our understanding of the innate immune response to asthma exacerbating factors. In this review, we summarize the current understanding of the role of innate immunity in asthma exacerbations and the methods used to study them. We also identify potential novel therapeutic targets for asthma and future areas for investigation.
Collapse
Affiliation(s)
- Jehan Alladina
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Benjamin D. Medoff
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Josalyn L. Cho
- Division of Pulmonary, Critical Care and Occupational Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| |
Collapse
|
12
|
Varricchi G, Poto R, Lommatzsch M, Brusselle G, Braido F, Virchow JC, Canonica GW. Biologics and airway remodeling in asthma: early, late, and potential preventive effects. Allergy 2025; 80:408-422. [PMID: 39520155 PMCID: PMC11804314 DOI: 10.1111/all.16382] [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: 08/25/2024] [Revised: 10/02/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024]
Abstract
Although airway remodeling in severe and/or fatal asthma is still considered irreversible, its individual components as a cause of clinical symptoms and/or lung function changes remain largely unknown. While inhaled glucocorticoids have not consistently been shown to affect airway remodeling, biologics targeting specific pathways of airway inflammation have been shown to improve lung function, mucus plugging, and airway structural changes that can exceed those seen with glucocorticoids. This superiority of biologic treatment, which cannot be solely explained by insufficient doses or limited durations of glucocorticoid therapies, needs to be further explored. For this field of research, we propose a novel classification of the potential effects of biologics on airway remodeling into three temporal effects: early effects (days to weeks, primarily modulating inflammatory processes), late effects (months to years, predominantly affecting structural changes), and potential preventive effects (outcomes of early treatment with biologics). For the identification of potential preventive effects of biologics, we call for studies exploring the impact of early biological treatment on airway remodeling in patients with moderate-to-severe asthma, which should be accompanied by a long-term evaluation of clinical parameters, biomarkers, treatment burden, and socioeconomic implications.
Collapse
Affiliation(s)
- G. Varricchi
- Department of Translational Medical SciencesUniversity of Naples Federico IINaplesItaly
- Center for Basic and Clinical Immunology Research (CISI)University of Naples Federico IINaplesItaly
- World Allergy Organization (WAO) Center of ExcellenceNaplesItaly
- Institute of Experimental Endocrinology and Oncology (IEOS)National Research CouncilNaplesItaly
| | - R. Poto
- Department of Translational Medical SciencesUniversity of Naples Federico IINaplesItaly
- Center for Basic and Clinical Immunology Research (CISI)University of Naples Federico IINaplesItaly
- World Allergy Organization (WAO) Center of ExcellenceNaplesItaly
| | - M. Lommatzsch
- Department of Pneumology and Critical Care MedicineUniversity of RostockRostockGermany
| | - G. Brusselle
- Department of Respiratory MedicineGhent University HospitalGhentBelgium
| | - F. Braido
- Respiratory Diseases and Allergy DepartmentIRCCS Polyclinic Hospital San MartinoGenoaItaly
| | - J. C. Virchow
- Department of Pneumology and Critical Care MedicineUniversity of RostockRostockGermany
| | - G. W. Canonica
- Respiratory Diseases and Allergy DepartmentIRCCS Polyclinic Hospital San MartinoGenoaItaly
- Department of Biomedical SciencesHumanitas UniversityMilanItaly
- Asthma & Allergy Unit‐IRCCS Humanitas Research HospitalMilanItaly
| |
Collapse
|
13
|
Sun W, Mou S, Huntington C, Killick H, Scott IC, Kelly A, Gavala M, Larsson J, Vakkalanka MD, Alexis NE, Wiley W, Wheeler A, Shah K, Yuan M, Mylott WR, Contrepois K, Rosenbaum AI. Development and qualification of an LC-MS/MS method for quantification of MUC5AC and MUC5B mucins in spontaneous sputum. Bioanalysis 2025; 17:187-198. [PMID: 39976267 PMCID: PMC11853556 DOI: 10.1080/17576180.2025.2457844] [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: 10/08/2024] [Accepted: 01/21/2025] [Indexed: 02/21/2025] Open
Abstract
AIM Airway mucins in sputum are promising respiratory disease biomarkers, despite posing substantial analytical challenges due to their physicochemical properties and rare and heterogenous nature of the matrix. We aimed to identify a suitable sputum collection and processing method, and qualify a bioanalytical method for MUC5AC and MUC5B quantification in clinical samples. METHOD Mucins were quantified in induced and spontaneous sputum collected from the same COPD patients, following various sample processing procedures. LC-MS/MS method used truncated recombinant mucins as surrogate analytes in surrogate matrix. RESULTS Frozen spontaneous sputum was found to be a suitable and convenient matrix for mucin quantification and fit-for-purpose method qualification was performed. CONCLUSION Our methodology provides accurate and reliable MUC5AC and MUC5B quantification and facilitates multi-site clinical sputum collection.
Collapse
Affiliation(s)
- Weiwen Sun
- Integrated Bioanalysis, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, South San Francisco, CA, USA
| | - Si Mou
- Integrated Bioanalysis, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, South San Francisco, CA, USA
| | | | - Helen Killick
- Translational Science and Experimental Medicine, Research and Early Development Respiratory & Immunology, R&D, AstraZeneca, Cambridge, UK
| | - Ian Christopher Scott
- Translational Science and Experimental Medicine, Research and Early Development Respiratory & Immunology, R&D, AstraZeneca, Cambridge, UK
| | - Aoife Kelly
- Translational Science and Experimental Medicine, Research and Early Development Respiratory & Immunology, R&D, AstraZeneca, Cambridge, UK
| | - Monica Gavala
- Translational Science and Experimental Medicine, Research and Early Development Respiratory & Immunology, R&D, AstraZeneca, Gaithersburg, MA, USA
| | - Jessica Larsson
- Translational Science and Experimental Medicine, Research and Early Development Respiratory & Immunology, R&D, AstraZeneca, Cambridge, UK
| | - Mani Deepika Vakkalanka
- Chromatographic Services – Research & Development Biologics by LC–MS/MS, PPD Laboratory Services (A part of Thermo Fisher Scientific), Richmond, VA, USA
| | - Neil E. Alexis
- Center for Environmental Medicine Asthma and Lung Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Walter Wiley
- Chromatographic Services – Research & Development Biologics by LC–MS/MS, PPD Laboratory Services (A part of Thermo Fisher Scientific), Richmond, VA, USA
| | - Aaron Wheeler
- Chromatographic Services – Research & Development Biologics by LC–MS/MS, PPD Laboratory Services (A part of Thermo Fisher Scientific), Richmond, VA, USA
| | - Kumar Shah
- Chromatographic Services – Research & Development Biologics by LC–MS/MS, PPD Laboratory Services (A part of Thermo Fisher Scientific), Richmond, VA, USA
| | - Moucun Yuan
- Chromatographic Services – Research & Development Biologics by LC–MS/MS, PPD Laboratory Services (A part of Thermo Fisher Scientific), Richmond, VA, USA
| | - William R. Mylott
- Chromatographic Services – Research & Development Biologics by LC–MS/MS, PPD Laboratory Services (A part of Thermo Fisher Scientific), Richmond, VA, USA
| | - Kévin Contrepois
- Integrated Bioanalysis, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, South San Francisco, CA, USA
| | - Anton I. Rosenbaum
- Integrated Bioanalysis, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, South San Francisco, CA, USA
| |
Collapse
|
14
|
Matheson AM, Johnstone J, Niedbalski PJ, Woods JC, Castro M. New frontiers in asthma chest imaging. J Allergy Clin Immunol 2025; 155:241-254.e1. [PMID: 39709032 DOI: 10.1016/j.jaci.2024.12.1067] [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: 08/29/2024] [Revised: 12/11/2024] [Accepted: 12/13/2024] [Indexed: 12/23/2024]
Abstract
Modern pulmonary imaging can reveal underlying pathologic and pathophysiologic changes in the lungs of people with asthma, with important clinical implications. A multitude of imaging modalities, including computed tomography, magnetic resonance imaging, optical coherence tomography, and endobronchial ultrasound, are now being used to examine underlying structure-function relationships. Imaging-based biomarkers from these techniques, including airway dimensions, blood vessel volumes, mucus scores, extent of ventilation defect, and extent of air trapping, often have increased sensitivity compared with that of traditional lung function measurements and are increasingly being used as end points in clinical trials. Imaging has been crucial to recent improvements in our understanding of the relationships between type 2 inflammation, eosinophilia, and mucus extent. With the advent of effective anti-type 2 biologic therapies, computed tomography and magnetic resonance imaging techniques can identify not just which patients benefit from therapy but why they benefit. Clinical trials have begun to assess the utility of imaging to prospectively plan airway therapy targets in bronchial thermoplasty and have potential to direct future bronchoscopic therapies. Together, imaging techniques provide a diverse set of tools to investigate how spatially distributed airway, blood, and parenchymal abnormalities shape disease heterogeneity in patients with asthma.
Collapse
Affiliation(s)
- Alexander M Matheson
- Center for Pulmonary Imaging Research, Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Joseph Johnstone
- Pulmonary, Critical Care, and Sleep Medicine, University of Kansas Medical Center, Kansas City, Kan
| | - Peter J Niedbalski
- Pulmonary, Critical Care, and Sleep Medicine, University of Kansas Medical Center, Kansas City, Kan; Hoglund Biomedical Imaging Center, University of Kansas Medical Center, Kansas City, Kan
| | - Jason C Woods
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio; Cincinnati Bronchopulmonary Dysplasia Center, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Mario Castro
- Pulmonary, Critical Care, and Sleep Medicine, University of Kansas Medical Center, Kansas City, Kan.
| |
Collapse
|
15
|
Domvri K, Tsiouprou I, Bakakos P, Steiropoulos P, Katsoulis K, Kostikas K, Antoniou KM, Papaioannou AI, Rovina N, Katsaounou P, Papamitsou T, Pastelli N, Tryfon S, Fouka E, Papakosta D, Loukides S, Porpodis K. Effect of mepolizumab in airway remodeling in patients with late-onset severe asthma with an eosinophilic phenotype. J Allergy Clin Immunol 2025; 155:425-435. [PMID: 39521278 DOI: 10.1016/j.jaci.2024.10.024] [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: 06/09/2024] [Revised: 10/08/2024] [Accepted: 10/10/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Clinical trials and real-world experience have provided evidence for the clinical benefits of mepolizumab, an anti-IL-5 biologic, in severe asthma. However, limited data exist regarding the impact of mepolizumab on airway remodeling. OBJECTIVE We sought to investigate the effect of mepolizumab on airway structural remodeling in patients treated for severe asthma in routine clinical care. METHODS The MESILICO (Efficacy of Mepolizumab in patients with latE-onset Severe eosInophiLic asthma and fIxed obstruCtiOn) study is a multicenter study involving 8 pulmonology departments in Greece. This study focused on patients who initiated mepolizumab for severe asthma with an eosinophilic phenotype and had late-onset disease with obstructive patterns (impaired reversibility). Forty-seven patients were recruited, of whom 41 were enrolled in the bronchoscopy substudy. The findings were related to clinical outcome. RESULTS After 12 months, mepolizumab treatment was associated with significant improvements in lung function and Asthma Control Test score, along with a significant decrease in severe exacerbation events (P < .001). Thirty-four of the 41 participants (83%) had paired biopsies for comparative analysis. There was a significant reduction from baseline in sub-basement membrane thickness, airway smooth muscle area, airway smooth muscle layer thickness, extent of epithelial damage, and number of tissue eosinophils (all P < .001). The extent of reduction in airway smooth muscle layer thickness positively correlated with the submucosal eosinophil reduction (r = 0.599; P < .001). CONCLUSIONS This study identified that 12 months of mepolizumab treatment in patients with late-onset severe asthma, who are also characterized by eosinophilic and impaired reversibility phenotypes, not only leads to clinical improvement but also reduces indices of airway tissue remodeling suggestive of a disease-modifying effect.
Collapse
Affiliation(s)
- Kalliopi Domvri
- Pulmonary Department, George Papanikolaou Hospital, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece; Laboratory of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece; Department of Pathology, George Papanikolaou General Hospital of Thessaloniki, Thessaloniki, Greece.
| | - Ioanna Tsiouprou
- Pulmonary Department, George Papanikolaou Hospital, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Petros Bakakos
- 1st Respiratory Department, Sotiria Chest Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Paschalis Steiropoulos
- Department of Respiratory Medicine, Medical School, Democritus University of Thrace, University General Hospital Dragana, Alexandroupolis, Greece
| | | | | | - Katerina M Antoniou
- Department of Thoracic Medicine and Laboratory of Cellular and Molecular Pneumonology, Medical School, University of Crete, Crete, Greece
| | - Andriana I Papaioannou
- 1st Respiratory Department, Sotiria Chest Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Nikoletta Rovina
- 1st Respiratory Department, Sotiria Chest Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Paraskevi Katsaounou
- Pulmonary and Respiratory Failure Department, First ICU, Evaggelismos Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Theodora Papamitsou
- Laboratory of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Nicoleta Pastelli
- Department of Pathology, George Papanikolaou General Hospital of Thessaloniki, Thessaloniki, Greece
| | - Stavros Tryfon
- Pulmonary Department (NHS), George Papanikolaou General Hospital of Thessaloniki, Thessaloniki, Greece
| | - Evangelia Fouka
- Pulmonary Department, George Papanikolaou Hospital, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Despoina Papakosta
- Pulmonary Department, George Papanikolaou Hospital, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Stelios Loukides
- Attiko University Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Konstantinos Porpodis
- Pulmonary Department, George Papanikolaou Hospital, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
16
|
Lindsley AW, Lugogo N, Reeh KAG, Spahn J, Parnes JR. Asthma Biologics Across the T2 Spectrum of Inflammation in Severe Asthma: Biomarkers and Mechanism of Action. J Asthma Allergy 2025; 18:33-57. [PMID: 39830595 PMCID: PMC11742565 DOI: 10.2147/jaa.s496630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 12/10/2024] [Indexed: 01/22/2025] Open
Abstract
Airway inflammation, a hallmark feature of asthma, drives many canonical features of the disease, including airflow limitation, mucus plugging, airway remodeling, and hyperresponsiveness. The T2 inflammatory paradigm is firmly established as the dominant mechanism of asthma pathogenesis, largely due to the success of inhaled corticosteroids and biologic therapies targeting components of the T2 pathway, including IL-4, IL-5, IL-13, and thymic stromal lymphopoietin (TSLP). However, up to 30% of patients may lack signatures of meaningful T2 inflammation (ie, T2 low). In T2-low asthma patients, T2 inflammation may be masked due to anti-inflammatory treatments or may be highly variable depending on exposure to common asthma triggers such as allergens, respiratory infections, and smoke or pollution. The epithelium and epithelial cytokines (TSLP, IL-33) are increasingly recognized as upstream drivers of canonical T2 pathways and as modulators of various effector cells, including mast cells, eosinophils, and neutrophils, which impact the pathological manifestations of airway smooth muscle hypertrophy, hypercontractility, and airway hyperresponsiveness. Approved biologics for severe asthma target several distinct mechanisms of action, leading to differential effects on the spectrum of T2 inflammation, inflammatory biomarkers, and treatment efficacy (reducing asthma exacerbations, improving lung function, and diminishing symptoms). The approved anti-asthma biologics primarily target T2 immune pathways, with little evidence suggesting a benefit of targeting non-T2 asthma-associated mediators. Indeed, many negative results challenge current assumptions about the etiology of non-T2 asthma and raise doubts about the viability of targeting popular alternative inflammatory pathways, such as T17. Novel data have emerged from the use of biologics to treat various inflammatory mediators and have furthered our understanding of pathogenic mechanisms that drive asthma. This review discusses inflammatory pathways that contribute to asthma, quantitatively outlines effects of available biologics on biomarkers, and summarizes data and challenges from clinical trials that address non-T2 mechanisms of asthma.
Collapse
Affiliation(s)
| | - Njira Lugogo
- Michigan Medicine Asthma Program, University of Michigan, Ann Arbor, MI, USA
| | | | | | | |
Collapse
|
17
|
Diaz AA. Beyond Bronchodilation and Airway Inflammation: Mucus Plugs as a Therapeutic Target in COPD. Chest 2025; 167:34-36. [PMID: 39794076 DOI: 10.1016/j.chest.2024.08.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/26/2024] [Accepted: 08/02/2024] [Indexed: 01/13/2025] Open
Affiliation(s)
- Alejandro A Diaz
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.
| |
Collapse
|
18
|
Tiotiu A, Steiropoulos P, Novakova S, Nedeva D, Novakova P, Chong-Neto H, Fogelbach GG, Kowal K. Airway Remodeling in Asthma: Mechanisms, Diagnosis, Treatment, and Future Directions. Arch Bronconeumol 2025; 61:31-40. [PMID: 39368875 DOI: 10.1016/j.arbres.2024.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 08/27/2024] [Accepted: 09/20/2024] [Indexed: 10/07/2024]
Abstract
Airway remodeling (AR) with chronic inflammation, are key features in asthma pathogenesis. AR characterized by structural changes in the bronchial wall is associated with a specific asthma phenotype with poor clinical outcomes, impaired lung function and reduced treatment response. Most studies focus on the role of inflammation, while understanding the mechanisms driving AR is crucial for developing disease-modifying therapeutic strategies. This review paper summarizes current knowledge on the mechanisms underlying AR, diagnostic tools, and therapeutic approaches. Mechanisms explored include the role of the resident cells and the inflammatory cascade in AR. Diagnostic methods such as bronchial biopsy, lung function testing, imaging, and possible biomarkers are described. The effectiveness on AR of different treatments of asthma including corticosteroids, leukotriene modifiers, bronchodilators, macrolides, biologics, and bronchial thermoplasty is discussed, as well as other possible therapeutic options. AR poses a significant challenge in asthma management, contributing to disease severity and treatment resistance. Current therapeutic approaches target mostly airway inflammation rather than smooth muscle cell dysfunction and showed limited benefits on AR. Future research should focus more on investigating the mechanisms involved in AR to identify novel therapeutic targets and to develop new effective treatments able to prevent irreversible structural changes and improve long-term asthma outcomes.
Collapse
Affiliation(s)
- Angelica Tiotiu
- Department of Pulmonology, University Hospital Saint-Luc, Brussels, Belgium; Pole Pneumology, ENT, and Dermatology - LUNS, Institute of Experimental and Clinical Research (IREC), UCLouvain, Brussels, Belgium.
| | - Paschalis Steiropoulos
- Department of Pulmonology, Medical School, Democritus University of Thrace, University General Hospital of Alexandroupolis, Alexandroupolis, Greece
| | - Silviya Novakova
- Department of Allergology, University Hospital "Sv. Georgi" Plovdiv, Bulgaria
| | - Denislava Nedeva
- Clinic of Asthma and Allergology, UMBAL Alexandrovska, Medical University Sofia, Sofia, Bulgaria
| | - Plamena Novakova
- Department of Allergy, Medical University Sofia, Sofia, Bulgaria
| | - Herberto Chong-Neto
- Division of Allergy and Immunology, Complexo Hospital de Clínicas, Universidade Federal do Paraná, Curitiba, Brazil
| | | | - Krzysztof Kowal
- Department of Experimental Allergology and Immunology and Department of Allergology and Internal Medicine, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
19
|
Gyawali B, Georas SN, Khurana S. Biologics in severe asthma: a state-of-the-art review. Eur Respir Rev 2025; 34:240088. [PMID: 39778920 PMCID: PMC11707604 DOI: 10.1183/16000617.0088-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 10/24/2024] [Indexed: 01/11/2025] Open
Abstract
Asthma is considered severe if it remains uncontrolled despite optimal conventional therapy, characterised by poor symptom control, frequent exacerbations and increased exposure to systemic corticosteroids. This has a significant impact on morbidity, mortality and healthcare resource utilisation. Recent advances in the understanding of asthma heterogeneity and immunopathogenesis have helped delineate precise disease pathways. The discovery of these pivotal pathways has led to the development of highly effective biologic therapies. Currently available asthma biologics target immunoglobulin E, interleukin (IL)-5/IL-5Rα, IL-4Rα and thymic stromal lymphopoietin. Identification of specific asthma phenotypes, utilising easily measurable biomarkers, has paved the way towards personalised and precision asthma management. Biologic therapies play a significant role in reducing exacerbations, hospitalisations and the need for maintenance systemic steroids, while also improving the quality of life in patients with severe asthma. The evidence for their clinical efficacy comes from randomised controlled trials (RCTs), extension studies, metanalyses and real-world data. This review synthesises findings from early, pivotal RCTs and subsequent studies following the approval of biologics for severe asthma. The safety and efficacy data from these studies, completed in a variety of settings, provide practical perspectives on their application and enhance their generalisability.
Collapse
Affiliation(s)
- Bishal Gyawali
- Division of Pulmonary and Critical Care Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Steve N Georas
- Division of Pulmonary and Critical Care Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
- Mary Parkes Center for Asthma, Allergy and Pulmonary Care, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Sandhya Khurana
- Division of Pulmonary and Critical Care Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
- Mary Parkes Center for Asthma, Allergy and Pulmonary Care, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| |
Collapse
|
20
|
Tashiro H, Kuwahara Y, Kurihara Y, Takahashi K. Molecular mechanisms and clinical impact of biologic therapies in severe asthma. Respir Investig 2025; 63:50-60. [PMID: 39642687 DOI: 10.1016/j.resinv.2024.11.015] [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: 09/30/2024] [Revised: 11/18/2024] [Accepted: 11/27/2024] [Indexed: 12/09/2024]
Abstract
Severe asthma is a critical condition for patients with asthma, characterized by frequent exacerbations, decreased pulmonary function, and unstable symptoms related to asthma. Consequently, the administration of systemic corticosteroids, which cause secondary damage because of their adverse effects, is considered. Recently, several types of molecular-targeted biological therapies have become available for patients with severe asthma, and they have a capacity to improve the pathophysiology of severe asthma. However, several clinical reports indicate that the effects differ depending on the biological targets of asthma in individual patients. In this review, the molecular mechanisms and clinical impact of biologic therapies in severe asthma are described. In addition, molecules targeted by possible future biologics are also addressed. Better understanding of the mechanistic basis for the role of biologics in severe asthma could lead to new therapeutic options for these patients.
Collapse
Affiliation(s)
- Hiroki Tashiro
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Saga Prefecture, 849-8501, Japan
| | - Yuki Kuwahara
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Saga Prefecture, 849-8501, Japan
| | - Yuki Kurihara
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Saga Prefecture, 849-8501, Japan
| | - Koichiro Takahashi
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Saga Prefecture, 849-8501, Japan.
| |
Collapse
|
21
|
Mailhot-Larouche S, Celis-Preciado C, Heaney LG, Couillard S. Identifying super-responders: A review of the road to asthma remission. Ann Allergy Asthma Immunol 2025; 134:31-45. [PMID: 39383944 DOI: 10.1016/j.anai.2024.09.023] [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: 09/17/2024] [Revised: 09/25/2024] [Accepted: 09/27/2024] [Indexed: 10/11/2024]
Abstract
Asthma is a chronic respiratory disease marked by heterogeneity and variable clinical outcomes. Recent therapeutic advances have highlighted patients achieving optimal outcomes, termed "remission" or "super-response." This review evaluates the various definitions of these terms and explores how disease burden impedes the attainment of remission. We assessed multiple studies, including a recent systematic review and meta-analysis, on biologic treatments for asthma remission. Our review highlights that type 2 inflammation may be the strongest predictor of biologic response. Key comorbidities (eg, obesity and mood disorders) and behavioral factors (eg, poor adherence, improper inhalation technique, and smoking) were identified as dominant traits limiting remission. In addition, asthma burden and longer disease duration significantly restrict the potential for remission in patients with severe asthma under the current treatment paradigm. We review the potential for a "predict-and-prevent" approach, which focuses on early identification of high-risk patients with type 2 inflammation and aggressive treatment to improve long-term asthma outcomes. In conclusion, this scoping review highlights the following unmet needs in asthma remission: (1) a harmonized global definition, with better defined lung function parameters; (2) integration of nonbiologic therapies into remission strategies; and (3) a clinical trial of early biologic intervention in patients with remission-prone, very type 2-high, moderately severe asthma with clinical remission as a predefined primary end point.
Collapse
Affiliation(s)
- Samuel Mailhot-Larouche
- Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Carlos Celis-Preciado
- Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Liam G Heaney
- Centre for Experimental Medicine, Queen's University Belfast School of Medicine, Dentistry and Biomedical Sciences, Belfast, United Kingdom
| | - Simon Couillard
- Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Québec, Canada.
| |
Collapse
|
22
|
Tashiro H, Nanri M, Kuwahara Y, Kurihara Y, Kimura S, Takahashi K. Possible Biological Heterogeneity of Airway Mucus Plugs in a Patient with Asthma. J Asthma Allergy 2024; 17:1265-1269. [PMID: 39655337 PMCID: PMC11628187 DOI: 10.2147/jaa.s499026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 11/19/2024] [Indexed: 12/12/2024] Open
Abstract
Background The presence of mucus plugs in the airway is a severe phenotype in patients with asthma; however, the mechanisms and specific treatments are not fully understood. Purpose To clarify the efficacy of biologics and the mechanisms for mucus plug in patients with asthma. Patients and Methods A 79-year-old Japanese asthmatic woman with high blood eosinophil and fractional exhaled nitric oxide (FeNO) was pointed massive mucus plugs in airway on chest CT imaging. She was treated with mepolizumab for 3 months; however, those were augmented without improvement of pulmonary function and FeNO. She was switched to dupilumab and, three months later, the mucus plugs were completely disappeared with improvement in pulmonary function, FeNO and asthma-related symptoms. She continues treatment with dupilumab, and her asthma symptoms are stable. Conclusion Biologics including mepolizumab and dupilumab have been reported to improve mucus plugs in patients with asthma; however, their efficacy might be different depending on the clinical characteristics of patients. Present data remind us of the possibility that, if FeNO levels remain high after treatment with anti-IL-5 antibody in asthma patients with mucus plugs, switching to other biologics, including anti-IL-4Rα antibody, might be considered.
Collapse
Affiliation(s)
- Hiroki Tashiro
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Saga Prefecture, Japan
| | - Mizuki Nanri
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Saga Prefecture, Japan
| | - Yuki Kuwahara
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Saga Prefecture, Japan
| | - Yuki Kurihara
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Saga Prefecture, Japan
| | - Shinya Kimura
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Saga Prefecture, Japan
| | - Koichiro Takahashi
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Saga Prefecture, Japan
| |
Collapse
|
23
|
De Corso E, Hellings PW, Fokkens WJ, Klimek L, Peters AT, Scadding GK, Desrosiers M, Lee SE, Mullol J. Thymic Stromal Lymphopoietin (TSLP): Evidence in Respiratory Epithelial-driven Diseases Including Chronic Rhinosinusitis with Nasal Polyps. Curr Allergy Asthma Rep 2024; 25:7. [PMID: 39636450 DOI: 10.1007/s11882-024-01186-2] [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] [Accepted: 11/04/2024] [Indexed: 12/07/2024]
Abstract
PURPOSE OF THE REVIEW Thymic stromal lymphopoietin (TSLP) is increasingly recognized for its pivotal role in the pathogenesis of various epithelial-driven chronic inflammatory diseases. This review navigates the existing evidence on TSLP, with a particular focus on asthma, before delving into the current understanding of its role in chronic rhinosinusitis with nasal polyps (CRSwNP). We explore the role of TSLP in the pathogenesis of asthma and CRSwNP, two conditions often interconnected and collectively referred to as"Global Airway Disease". Additionally, this review assesses the therapeutic potential of TSLP inhibition as a treatment option for both CRSwNP and asthma. A systematic literature search was conducted; selected publications were used to describe the biology of TSLP, including its expression and diverse effects on inflammation. RECENT FINDINGS The role of TSLP in asthma is well established and supported by the efficacy of tezepelumab, the first anti-TSLP monoclonal antibody approved for both type 2 (T2)-high and T2-low severe asthma. TSLP may be a key contributor to CRSwNP pathogenesis as evidenced by genetic and mechanistic studies in which TSLP has been shown to regulate T2 inflammation and influence non-T2 responses. Preliminary data from the NAVIGATOR trial indicate that tezepelumab may reduce CRSwNP symptoms in patients with comorbid asthma. While further research is required to clarify the extent of TSLP contribution in CRSwNP, this review highlights the potential of anti-TSLP therapies as a novel approach for managing severe, uncontrolled CRSwNP. If these preliminary findings are confirmed, targeting TSLP could become a promising strategy to treat CRSwNP with or without comorbid asthma.
Collapse
Affiliation(s)
- Eugenio De Corso
- UOC Otorinolaringoiatria, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.Go F.Vito 1, Roma, Italy.
| | - Peter W Hellings
- The European Forum for Research and Education in Allergy and Airway Diseases Scientific Expert Team Members, Brussels, Belgium
- Laboratory of Allergy and Clinical Immunology Research Unit, Department of Microbiology, Immunology and Transplantation, KU Leuven, Louvain, Belgium
- Department of Otorhinolaryngology, Head and Neck Surgery, UZ Leuven, Louvain, Belgium
| | - Wytske J Fokkens
- Department of Otorhinolaryngology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Ludger Klimek
- Center for Rhinology and Allergology, Wiesbaden, 2HNO-University Clinic Charité, Berlin, Germany
| | - Anju T Peters
- Department of Otolaryngology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Glenis K Scadding
- Department of Allergy and Rhinology, Royal National ENT Hospital, London, UK
| | | | - Stella E Lee
- Department of Surgery, Division of Otolaryngology-Head & Neck Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Joaquim Mullol
- Rhinology Unit and Smell Clinic, Department of Otorhinolaryngology, Hospital Clinic, FRCB- IDIBAPS, Universitat de Barcelona, CIBERES. Barcelona, Catalonia, Spain
| |
Collapse
|
24
|
Diaz AA, Grumley S, Yen A, Sonavane S, Elalami R, Abdalla M, Kim K, Nardelli P, Brouha S, Manapragada PP, Abozeed M, Aziz MU, Zahid M, Nath HP, Wang W, Ross JC, Pistenmaa CL, San José Estépar R, Cho MH. Eosinophils, mucus plugs and clinical outcomes: findings from two COPD cohorts. Eur Respir J 2024; 64:2401005. [PMID: 39572219 DOI: 10.1183/13993003.01005-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 09/25/2024] [Indexed: 12/07/2024]
Affiliation(s)
- Alejandro A Diaz
- Harvard Medical School, Boston, MA, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Scott Grumley
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Andrew Yen
- Department of Radiology, University of California San Diego, San Diego, CA, USA
| | | | - Rim Elalami
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Maya Abdalla
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Kangjin Kim
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Pietro Nardelli
- Harvard Medical School, Boston, MA, USA
- Department of Radiology, Brigham and Women's Hospital, Boston, MA, USA
| | - Sharon Brouha
- Department of Radiology, University of California San Diego, San Diego, CA, USA
| | - Padma P Manapragada
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Mostafa Abozeed
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Muhammad Usman Aziz
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Mohd Zahid
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hrudaya P Nath
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Wei Wang
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA
| | - James C Ross
- Harvard Medical School, Boston, MA, USA
- Department of Radiology, Brigham and Women's Hospital, Boston, MA, USA
| | - Carrie L Pistenmaa
- Harvard Medical School, Boston, MA, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Raul San José Estépar
- Harvard Medical School, Boston, MA, USA
- Department of Radiology, Brigham and Women's Hospital, Boston, MA, USA
| | - Michael H Cho
- Harvard Medical School, Boston, MA, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
| |
Collapse
|
25
|
Wechsler ME, Brusselle G, Virchow JC, Bourdin A, Kostikas K, Llanos JP, Roseti SL, Ambrose CS, Hunter G, Jackson DJ, Castro M, Lugogo N, Pavord ID, Martin N, Brightling CE. Clinical response and on-treatment clinical remission with tezepelumab in a broad population of patients with severe, uncontrolled asthma: results over 2 years from the NAVIGATOR and DESTINATION studies. Eur Respir J 2024; 64:2400316. [PMID: 39326921 PMCID: PMC11618813 DOI: 10.1183/13993003.00316-2024] [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: 02/15/2024] [Accepted: 08/23/2024] [Indexed: 09/28/2024]
Abstract
BACKGROUND In asthma, clinical response is characterised by disease improvement with treatment, whereas clinical remission is characterised by long-term disease stabilisation with or without ongoing treatment. The proportions of patients receiving tezepelumab who responded to treatment and who achieved on-treatment clinical remission were assessed in the NAVIGATOR (ClinicalTrials.gov identifier NCT03347279) and DESTINATION (ClinicalTrials.gov identifier NCT03706079) studies of severe, uncontrolled asthma. METHODS NAVIGATOR and DESTINATION were phase 3, randomised, double-blind, placebo-controlled studies; DESTINATION was an extension of NAVIGATOR. Complete clinical response was defined as achieving all of the following: ≥50% reduction in exacerbations versus the previous year, improvements in pre-bronchodilator (BD) forced expiratory volume in 1 s (FEV1) of ≥100 mL or ≥5%, improvements in Asthma Control Questionnaire (ACQ)-6 score of ≥0.5 and physician's assessment of asthma improvement. On-treatment clinical remission was defined as an ACQ-6 total score ≤1.5, stable lung function (pre-BD FEV1 >95% of baseline) and no exacerbations or use of oral corticosteroids during the time periods assessed. RESULTS Higher proportions of tezepelumab than placebo recipients achieved complete clinical response over weeks 0-52 (46% versus 24%; OR 2.83, 95% CI 2.10-3.82) and on-treatment clinical remission over weeks 0-52 (28.5% versus 21.9%; OR 1.44, 95% CI 0.95-2.19) and weeks >52-104 (33.5% versus 26.7%; OR 1.44, 95% CI 0.97-2.14). Tezepelumab recipients who achieved on-treatment clinical remission versus complete clinical response at week 52 had better preserved lung function and lower inflammatory biomarker levels at baseline, and fewer exacerbations in the 12 months before the study. CONCLUSIONS Among patients with severe, uncontrolled asthma, tezepelumab treatment was associated with an increased likelihood of achieving complete clinical response and on-treatment clinical remission compared with placebo. Both are clinically important outcomes, but may be driven by different patient characteristics.
Collapse
Affiliation(s)
- Michael E. Wechsler
- Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO, USA
| | - Guy Brusselle
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - J. Christian Virchow
- Department of Pneumology and Department of Intensive Care Medicine, University of Rostock, Rostock, Germany
| | - Arnaud Bourdin
- PhyMedExp, University of Montpellier, CNRS, INSERM, CHU Montpellier, Montpellier, France
| | | | | | - Stephanie L. Roseti
- Late-stage Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Christopher S. Ambrose
- Respiratory and Immunology, BioPharmaceuticals Medical, AstraZeneca, Gaithersburg, MD, USA
| | - Gillian Hunter
- Biometrics, Late-stage Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - David J. Jackson
- Guy's Severe Asthma Centre, Guy's and St Thomas’ NHS Foundation Trust, London, UK
- School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Mario Castro
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Kansas School of Medicine, Kansas City, KS, USA
| | - Njira Lugogo
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Ian D. Pavord
- Respiratory Medicine, National Institute for Health and Care Research, Oxford Biomedical Research Centre, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Neil Martin
- Institute for Lung Health, National Institute for Health and Care Research, Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
- Respiratory and Immunology, BioPharmaceuticals Medical, AstraZeneca, Cambridge, UK
| | - Christopher E. Brightling
- Institute for Lung Health, National Institute for Health and Care Research, Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
| |
Collapse
|
26
|
Lombardi C, Marcello C, Bosi A, Francesco M. Positioning tezepelumab for patients with severe asthma: from evidence to unmet needs. J Int Med Res 2024; 52:3000605241297532. [PMID: 39552062 PMCID: PMC11571243 DOI: 10.1177/03000605241297532] [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: 07/16/2024] [Accepted: 10/11/2024] [Indexed: 11/19/2024] Open
Abstract
Several endotypes of severe asthma with predominantly type 2 inflammation can be distinguished by the immune pathways driving the inflammatory processes. However, in the absence of type 2 inflammation, asthma is less clearly defined and is generally associated with poor responses to conventional anti-asthmatic therapies. Studies have shown that disruption of the epithelial barrier triggers inflammatory responses and increases epithelial permeability. A key aspect of this process is that epithelial cells release alarmin cytokines, including interleukin (IL)-25, IL-33, and thymic stromal lymphopoietin (TSLP), in response to allergens and infections. Among these cytokines, TSLP has been identified as a potential therapeutic target for severe asthma, leading to the development of a new biologic, tezepelumab (TZP). By blocking TSLP, TZP may produce wide-ranging effects. Based on positive clinical trial results, TZP appears to offer a promising, safe, and effective treatment approach. This narrative review examines the evidence for treating severe asthma with TZP, analyses clinical trial findings, and provides clinicians with practical insights into identifying patients who may respond best to this novel biologic therapy.
Collapse
Affiliation(s)
- Carlo Lombardi
- Departmental Unit of Allergology, Clinical Immunology & Pneumology, Istituto Ospedaliero Fondazione Poliambulanza, Brescia, Italy
| | | | - Annamaria Bosi
- Pulmonology Unit, S. Valentino Hospital, Montebelluna (TV), AULSS2 Marca Trevigiana, Italy
| | - Menzella Francesco
- Pulmonology Unit, S. Valentino Hospital, Montebelluna (TV), AULSS2 Marca Trevigiana, Italy
| |
Collapse
|
27
|
Tanabe N, Nakagawa H, Sakao S, Ohno Y, Shimizu K, Nakamura H, Hanaoka M, Nakano Y, Hirai T. Lung imaging in COPD and asthma. Respir Investig 2024; 62:995-1005. [PMID: 39213987 DOI: 10.1016/j.resinv.2024.08.014] [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: 03/21/2024] [Revised: 08/04/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024]
Abstract
Chronic obstructive pulmonary disease (COPD) and asthma are common lung diseases with heterogeneous clinical presentations. Lung imaging allows evaluations of underlying pathophysiological changes and provides additional personalized approaches for disease management. This narrative review provides an overview of recent advances in chest imaging analysis using various modalities, such as computed tomography (CT), dynamic chest radiography, and magnetic resonance imaging (MRI). Visual CT assessment localizes emphysema subtypes and mucus plugging in the airways. Dedicated software quantifies the severity and spatial distribution of emphysema and the airway tree structure, including the central airway wall thickness, branch count and fractal dimension of the tree, and airway-to-lung size ratio. Nonrigid registration of inspiratory and expiratory CT scans quantifies small airway dysfunction, local volume changes and shape deformations in specific regions. Lung ventilation and diaphragm movement are also evaluated on dynamic chest radiography. Functional MRI detects regional oxygen transfer across the alveolus using inhaled oxygen and ventilation defects and gas diffusion into the alveolar-capillary barrier tissue and red blood cells using inhaled hyperpolarized 129Xe gas. These methods have the potential to determine local functional properties in the lungs that cannot be detected by lung function tests in patients with COPD and asthma. Further studies are needed to apply these technologies in clinical practice, particularly for early disease detection and tailor-made interventions, such as the efficient selection of patients likely to respond to biologics. Moreover, research should focus on the extension of healthy life expectancy in patients at higher risk and with established diseases.
Collapse
Affiliation(s)
- Naoya Tanabe
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, 54 Shogo-in Kawahara-cho, Sakyo-ku, Kyoto, Kyoto, 606-8507, Japan.
| | - Hiroaki Nakagawa
- Division of Respiratory Medicine, Department of Internal Medicine, Shiga University of Medical Science, Setatsukinowa-cho, Otsu, Shiga 520-2192, Japan
| | - Seiichiro Sakao
- Department of Pulmonary Medicine, School of Medicine, International University of Health and Welfare, 4-3 Kozunomori, Narita, Chiba, 286-8686 Japan
| | - Yoshiharu Ohno
- Department of Diagnostic Radiology, Fujita Health University School of Medicine, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, Japan
| | - Kaoruko Shimizu
- Division of Emergent Respiratory and Cardiovascular medicine, Hokkaido University Hospital, Hokkaido University Hospital, Kita14, Nishi5, Kita-Ku, Sapporo, Hokkaido, 060-8648, Japan
| | - Hidetoshi Nakamura
- Department of Respiratory Medicine, Saitama Medical University, 38 Morohongo, Moroyama-machi, Iruma-gun, Saitama, 350-0495, Japan
| | - Masayuki Hanaoka
- First Department of Internal Medicine, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, 390-8621, Japan
| | - Yasutaka Nakano
- Division of Respiratory Medicine, Department of Internal Medicine, Shiga University of Medical Science, Setatsukinowa-cho, Otsu, Shiga 520-2192, Japan
| | - Toyohiro Hirai
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, 54 Shogo-in Kawahara-cho, Sakyo-ku, Kyoto, Kyoto, 606-8507, Japan
| |
Collapse
|
28
|
Flynn C, Brightling C. Picture this: The future of imaging biomarkers in COPD. Respirology 2024; 29:932-934. [PMID: 39089710 DOI: 10.1111/resp.14808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 07/25/2024] [Indexed: 08/04/2024]
Abstract
See related article
Collapse
Affiliation(s)
- Cara Flynn
- Institute for Lung Health, NIHR Biomedical Research Centre, University of Leicester, Leicester, UK
| | - Chris Brightling
- Institute for Lung Health, NIHR Biomedical Research Centre, University of Leicester, Leicester, UK
| |
Collapse
|
29
|
Israel E, Castro M, Ambrose CS, Llanos JP, Molfino NA, Martin NL, Ponnarambil SS, Martin N. Efficacy of tezepelumab in patients with severe asthma and persistent airflow obstruction. ERJ Open Res 2024; 10:00164-2024. [PMID: 39588080 PMCID: PMC11587167 DOI: 10.1183/23120541.00164-2024] [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: 02/19/2024] [Accepted: 05/31/2024] [Indexed: 11/27/2024] Open
Abstract
Background Persistent airflow obstruction (PAO) in patients with asthma can be difficult to treat. Tezepelumab blocks thymic stromal lymphopoietin, an epithelial cytokine implicated in asthma pathogenesis. This analysis evaluated the efficacy of tezepelumab in patients with severe, uncontrolled asthma and PAO. Methods PATHWAY (phase 2b) and NAVIGATOR (phase 3) were multicentre, randomised, double-blind, placebo-controlled studies. This post hoc analysis included PATHWAY and NAVIGATOR patients who received tezepelumab 210 mg or placebo every 4 weeks for 52 weeks. Change from baseline to week 52 in pre-bronchodilator forced expiratory volume in 1 s (FEV1) and the annualised asthma exacerbation rate (AAER) over 52 weeks were assessed in patients with and without PAO (post-bronchodilator FEV1/forced vital capacity ratio <0.7) at baseline. Results Of the 1334 included patients, 782 (58.6%) had PAO at baseline. At week 52, greater improvements in pre-bronchodilator FEV1 from baseline were observed in tezepelumab versus placebo recipients with PAO (least-squares (LS) mean 0.24 versus 0.07 L; difference 0.17 L, 95% confidence interval (CI): 0.11-0.23) and without PAO (LS mean 0.20 versus 0.12 L; difference 0.08 L, 95% CI: 0.01-0.15). Tezepelumab reduced the AAER versus placebo by 61% (95% CI: 51-69) and 56% (95% CI: 42-67) in patients with and without PAO, respectively. For patients with PAO at baseline, the proportion without PAO at week 52 was higher with tezepelumab (12.1%) than placebo (6.6%) (odds ratio 1.96, 95% CI: 1.30-2.94). Conclusion Tezepelumab improved lung function and reduced exacerbations versus placebo in patients with severe, uncontrolled asthma with and without PAO.
Collapse
Affiliation(s)
- Elliot Israel
- Pulmonary and Critical Care Medicine, Allergy and Immunology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Mario Castro
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Kansas School of Medicine, Kansas City, KS, USA
| | - Christopher S. Ambrose
- Respiratory and Immunology, BioPharmaceuticals Medical, AstraZeneca, Gaithersburg, MD, USA
| | | | | | - Nicole L. Martin
- Biometrics, Late-stage Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Waltham, MA, USA
- Cytel Inc., Waltham, MA, USA
| | - Sandhia S. Ponnarambil
- Late-stage Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Neil Martin
- Respiratory and Immunology, BioPharmaceuticals Medical, AstraZeneca, Cambridge, UK
- University of Leicester, Leicester, UK
| |
Collapse
|
30
|
Wan E, Yen A, Elalami R, Grumley S, Nath HP, Wang W, Brouha S, Manapragada PP, Abozeed M, Aziz MU, Zahid M, Ahmed AN, Terry NL, Nardelli P, Ross JC, Kim V, Sonavane S, Kligerman SJ, Vestbo J, Agusti A, Kim K, San José Estépar R, Silverman EK, Cho MH, Diaz AA. Airway Mucus Plugs on Chest Computed Tomography Are Associated with Exacerbations in COPD. Am J Respir Crit Care Med 2024; 211:814-822. [PMID: 39470402 DOI: 10.1164/rccm.202403-0632oc] [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: 03/25/2024] [Accepted: 10/23/2024] [Indexed: 10/30/2024] Open
Abstract
Rationale/Objective: Acute exacerbations (AEs) of chronic obstructive pulmonary disease (COPD) are associated with significant morbidity and mortality. Whether mucus plugs are associated with prospective exacerbations has not been examined extensively. METHODS Mucus plugs were visually-identified on baseline chest computed tomography (CT) scans from smokers with Global Initiative for Chronic Obstructive Lung Disease (GOLD) grades 2-4 COPD enrolled in two multicenter cohort studies: Evaluation of COPD Longitudinally to Identify Predictive Surrogate Endpoints (ECLIPSE) and COPDGene. Associations between ordinal mucus plug score categories (0/1-2/≥3) and prospectively-ascertained AEs, defined as worsening respiratory symptoms requiring systemic steroids and/or antibiotics (moderate-to-severe) and/or ER/hospitalization (severe), were assessed using multivariable-adjusted zero-inflated Poisson regression; subjects were exacerbation-free at enrollment. RESULTS Among 3,250 participants in COPDGene (mean±SD age 63.7±8.4 years, FEV1 50.6%±17.8% predicted, 45.1% female) and 1,716 participants in ECLIPSE (age 63.3±7.1 years, FEV1 48.3%±15.8% predicted, 36.2% female), 44.4% and 46.0% had mucus plugs, respectively. The incidence rates of AEs were 61.0 (COPDGene) and 125.7 (ECLIPSE) per 100 person-years. Relative to those without mucus plugs, the presence of 1-2 and ≥3 mucus plugs was associated with increased risk (adjusted rate ratio, aRR [95%CI]=1.07[1.05-1.09] and 1.15[1.1-1.2] in COPDGene; aRR=1.06[1.02-1.09] and 1.12[1.04-1.2] in ECLIPSE, respectively) for prospective moderate-to-severe AEs. The presence of 1-2 and ≥3 mucus plugs was also associated with increased risk for severe AEs during follow-up (aRR=1.05[1.01-1.08] and 1.09[1.02-1.18] in COPDGene; aRR=1.17[1.07-1.27] and 1.37[1.15-1.62] in ECLIPSE, respectively). CONCLUSION CT-based mucus plugs are associated with an increased risk for future COPD AEs.
Collapse
Affiliation(s)
- Emily Wan
- Brigham and Women's Hospital, Channing Division of Network Medicine, Boston, Massachusetts, United States
- VA Boston Health Care System Jamaica Plain Campus, Boston, Massachusetts, United States;
| | - Andrew Yen
- University of California San Diego, Department of Radiology, La Jolla, California, United States
| | - Rim Elalami
- Brigham and Women's Hospital, Boston, Massachusetts, United States
| | - Scott Grumley
- University of Alabama at Birmingham, Radiology, Birmingham, Alabama, United States
| | - Hrudaya P Nath
- University of Alabama at Birmingham, Department of Medicine, Birmingham, Alabama, United States
| | - Wei Wang
- 7. Division of Sleep Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Sharon Brouha
- University of California San Diego, La Jolla, California, United States
| | - Padma P Manapragada
- The University of Alabama at Birmingham Heersink School of Medicine, Radiology, Birmingham, Alabama, United States
| | - Mostafa Abozeed
- University of Alabama at Birmingham HCOP, Birmingham, Alabama, United States
| | - Muhammad Usman Aziz
- University of Alabama at Birmingham HCOP, Radiology, Birmingham, Alabama, United States
| | - Mohd Zahid
- The University of Alabama at Birmingham Hospital, Birmingham, Alabama, United States
| | - Asmaa N Ahmed
- University of Alabama at Birmingham HCOP, Birmingham, Alabama, United States
| | - Nina L Terry
- University of Alabama at Birmingham, Pulmonary, Allergy and Critical Care Medicine, Birmingham, Alabama, United States
| | - Pietro Nardelli
- Brigham and Women's Hospital, Radiology, Boston, Massachusetts, United States
| | - James C Ross
- Brigham and Womens Hospital, Boston, Massachusetts, United States
| | - Victor Kim
- Temple University, Pulmonary and Critical Care Medicine, Philadelphia, Pennsylvania, United States
| | - Sushilkumar Sonavane
- Mayo Clinic Hospital Jacksonville, Radiology, Jacksonville, Florida, United States
| | | | - Jørgen Vestbo
- The University of Manchester, Division of Infection, Immunity and Respiratory Medicine, Manchester, United Kingdom of Great Britain and Northern Ireland
- Manchester University NHS Foundation Trust, North West Lung Centre, Manchester, United Kingdom of Great Britain and Northern Ireland
| | - Alvar Agusti
- University of Barcelona Faculty of Medicine and Health Sciences, Barcelona, Spain
| | - Kangjin Kim
- Brigham and Women's Hospital and Harvard Medical School, Channing Division of Network Medicine, Department of Medicine, Boston, Massachusetts, United States
| | | | - Edwin K Silverman
- Brigham and Women's Hospital Channing Division of Network Medicine, Boston, Massachusetts, United States
| | - Michael H Cho
- Harvard Medical School, Channing Division of Respiratory Medicine, Boston, Massachusetts, United States
| | - Alejandro A Diaz
- Brigham and Women's Hospital, Medicine, Boston, Massachusetts, United States
| |
Collapse
|
31
|
Brightling CE, Marone G, Aegerter H, Chanez P, Heffler E, Pavord ID, Rabe KF, Uller L, Dorscheid D. The epithelial era of asthma research: knowledge gaps and future direction for patient care. Eur Respir Rev 2024; 33:240221. [PMID: 39694589 PMCID: PMC11653196 DOI: 10.1183/16000617.0221-2024] [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: 09/30/2024] [Accepted: 10/11/2024] [Indexed: 12/20/2024] Open
Abstract
The Epithelial Science Expert Group convened on 18-19 October 2023, in Naples, Italy, to discuss the current understanding of the fundamental role of the airway epithelium in asthma and other respiratory diseases and to explore the future direction of patient care. This review summarises the key concepts and research questions that were raised. As an introduction to the epithelial era of research, the evolution of asthma management throughout the ages was discussed and the role of the epithelium as an immune-functioning organ was elucidated. The role of the bronchial epithelial cells in lower airway diseases beyond severe asthma was considered, as well as the role of the epithelium in upper airway diseases such as chronic rhinosinusitis. The biology and application of biomarkers in patient care was also discussed. The Epithelial Science Expert Group also explored future research needs by identifying the current knowledge and research gaps in asthma management and ranking them by priority. It was identified that there is a need to define and support early assessment of asthma to characterise patients at high risk of severe asthma. Furthermore, a better understanding of asthma progression is required. The development of new treatments and diagnostic tests as well as the identification of new biomarkers will also be required to address the current unmet needs. Finally, an increased understanding of epithelial dysfunction will determine if we can alter disease progression and achieve clinical remission.
Collapse
Affiliation(s)
- Christopher E Brightling
- Institute for Lung Health, National Institute for Health and Care Research Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
- Joint first authors
| | - Gianni Marone
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research, School of Medicine, University of Naples Federico II, Naples, Italy
- Institute of Experimental Endocrinology and Oncology, National Research Council of Italy, Naples, Italy
- Joint first authors
| | - Helena Aegerter
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Pascal Chanez
- Department of Respiratory Diseases, Aix-Marseille University, Marseille, France
| | - Enrico Heffler
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy
- Personalized Medicine, Asthma and Allergy, IRCCS Humanitas Research Hospital, Rozzano (MI), Italy
| | - Ian D Pavord
- Respiratory Medicine, National Institute for Health and Care Research Oxford Biomedical Research Centre, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Klaus F Rabe
- LungenClinic Grosshansdorf, Member of the German Center for Lung Research (DZL), Grosshansdorf, Germany
- Chirstian-Alrechts University Kiel, Member of the German Center for Lung Research (DZL), Kiel, Germany
| | - Lena Uller
- Unit of Respiratory Immunopharmacology, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Del Dorscheid
- Center for Heart Lung Innovation, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
32
|
Yan S, Yang B, Qin H, Du C, Liu H, Jin T. Exploring the therapeutic potential of monoclonal antibodies targeting TSLP and IgE in asthma management. Inflamm Res 2024; 73:1425-1434. [PMID: 38907743 DOI: 10.1007/s00011-024-01908-2] [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: 03/15/2024] [Revised: 06/06/2024] [Accepted: 06/15/2024] [Indexed: 06/24/2024] Open
Abstract
BACKGROUND In recent years, there has been a growing interest in the utilization of biologic therapies for the management of asthma. Both TSLP and IgE are important immune molecules in the development of asthma, and they are involved in the occurrence and regulation of inflammatory response. METHODS A comprehensive search of PubMed and Web of Science was conducted to gather information on anti-TSLP antibody and anti-IgE antibody. RESULTS This investigation elucidates the distinct mechanistic roles of Thymic Stromal Lymphopoietin (TSLP) and Immunoglobulin E (IgE) in the pathogenesis of asthma, with a particular emphasis on delineating the therapeutic mechanisms and pharmacological properties of monoclonal antibodies targeting IgE and TSLP. Through a meticulous examination of clinical trials involving paradigmatic agents such as omalizumab and tezepelumab, we offer valuable insights into the potential treatment modalities for diseases with shared immunopathogenic pathways involving IgE and TSLP. CONCLUSION The overarching objective of this comprehensive study is to delve into the latest advancements in asthma therapeutics and to provide guidance for future investigations in this domain.
Collapse
Affiliation(s)
- Shuang Yan
- Sichuan University of Arts and Science, DaZhou, 635000, China.
- Key Laboratory of Exploitation and Study of Distinctive Plants in Education Department of Sichuan Province, Sichuan Institute of Arts and Science, DaZhou, 635000, China.
- Key Laboratory of Green Chemistry of Sichuan Institutes of Higher Education, ZiGong, 643000, China.
| | - Bowen Yang
- Unit for Drug and Instrument Supervision and Inspection of Wuxi Joint Logistic Support Center, Nanjing, 210000, China
| | - Haichuan Qin
- Sichuan University of Arts and Science, DaZhou, 635000, China
| | - Chengzhen Du
- Sichuan University of Arts and Science, DaZhou, 635000, China
| | - Hua Liu
- Sichuan University of Arts and Science, DaZhou, 635000, China
| | - Tengchuan Jin
- Department of Obstetrics and Gynecology, Division of Life Sciences and Medicine, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, Anhui, P.R. China.
- Laboratory of Structural Immunology, Key Laboratory of Immune Response and Immunotherapy, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, Anhui, China.
- Biomedical Sciences and Health Laboratory of Anhui Province, University of Science & Technology of China, Hefei, 230027, China.
- Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, 230001, China.
| |
Collapse
|
33
|
Wechsler ME, Wells JM. What every clinician should know about inflammation in COPD. ERJ Open Res 2024; 10:00177-2024. [PMID: 39319045 PMCID: PMC11417604 DOI: 10.1183/23120541.00177-2024] [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: 02/23/2024] [Accepted: 04/22/2024] [Indexed: 09/26/2024] Open
Abstract
Inflammation drives COPD pathogenesis and exacerbations. Although the conceptual framework and major players in the inflammatory milieu of COPD have been long established, the nuances of cellular interactions and the etiological differences that create heterogeneity in inflammatory profiles and treatment response continue to be revealed. This wealth of data and understanding is not only a boon to the researcher but also provides guidance to the clinician, moving the field closer to precision medicine. It is through this lens that this review seeks to describe the inflammatory processes at play in COPD, relating inflammation to pathological and functional changes, identifying patient-specific and disease-related factors that may influence clinical observations, and providing current insights on existing and emerging anti-inflammatory treatments and treatment targets, including biological therapies and phosphodiesterase (PDE) inhibitors.
Collapse
Affiliation(s)
- Michael E. Wechsler
- Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO, USA
| | - J. Michael Wells
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
34
|
Bourdin A, Bardin P, Chanez P. Imagining the severe asthma decision trees of the future. Expert Rev Respir Med 2024; 18:561-567. [PMID: 39120156 DOI: 10.1080/17476348.2024.2390987] [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: 05/24/2024] [Revised: 07/12/2024] [Accepted: 08/07/2024] [Indexed: 08/10/2024]
Abstract
INTRODUCTION There are no validated decision-making algorithms concerning severe asthma (SA) management. Future risks are crucial factors and can be derived from SA trajectories. AREAS COVERED The future severe asthma-decision trees should revisit current knowledge and gaps. A focused literature search has been conducted. EXPERT OPINION Asthma severity is currently defined a priori, thereby precluding a role for early interventions aiming to prevent outcomes such as exacerbations (systemic corticosteroids exposure) and lung function decline. Asthma 'at-risk' might represent the ultimate paradigm but merits longitudinal studies considering modern interventions. Real exacerbations, severe airway hyperresponsiveness, excessive T2-related biomarkers, noxious environments and patient behaviors, harms of OCS and high-doses inhaled corticosteroids (ICS), and low adherence-to-effectiveness ratios of ICS-containing inhalers are predictors of future risks. New tools such as imaging, genetic, and epigenetic signatures should be used. Logical and numerical artificial intelligence may be used to generate a consistent risk score. A pragmatic definition of response to treatments will allow development of a validated and applicable algorithm. Biologics have the best potential to minimize the risks, but cost remains an issue. We propose a simplified six-step algorithm for decision-making that is ultimately aiming to achieve asthma remission.
Collapse
Affiliation(s)
- Arnaud Bourdin
- Département de Pneumologie et Addictologie, PhyMedExp, University of Montpellier, INSERM U1046, CNRS UMR 9214, Montpellier, France
| | - Phil Bardin
- Monash Lung and Sleep Allergy Immunology, Monash Hospital, Monash Health and University, Hudson Institute, Melbourne, Victoria, Australia
| | - Pascal Chanez
- APHM, Clinique des bronches allergies et sommeil, Marseille, France
- Aix Marseille Univ, INSERM U1263, INRA 1260 (C2VN), Marseille, France
| |
Collapse
|
35
|
Hayashi Y, Tanabe N, Matsumoto H, Shimizu K, Sakamoto R, Oguma T, Sunadome H, Sato A, Sato S, Hirai T. Associations of fractional exhaled nitric oxide with airway dimension and mucus plugs on ultra-high-resolution computed tomography in former smokers and nonsmokers with asthma. Allergol Int 2024; 73:397-405. [PMID: 38403524 DOI: 10.1016/j.alit.2024.01.013] [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: 12/14/2023] [Revised: 01/21/2024] [Accepted: 01/30/2024] [Indexed: 02/27/2024] Open
Abstract
BACKGROUND Associations of fractional exhaled nitric oxide (FeNO) with airway wall remodeling and mucus plugs remain to be explored in smokers and nonsmokers with asthma. Ultra-high-resolution computed tomography (U-HRCT), which allows accurate structural quantification of airways >1 mm in diameter, was used in this study to examine whether higher FeNO was associated with thicker walls of the 3rd to 6th generation airways and mucus plugging in patients with asthma. METHODS The retrospective analyses included consecutive former smokers and nonsmokers with asthma who underwent U-HRCT in a hospital. The ratio of wall area to summed lumen and wall area was calculated as the wall area percent (WA%). Mucus plugging was visually scored. RESULTS Ninety-seven patients with asthma (including 59 former smokers) were classified into low (<20 ppb), middle (20-35 ppb), and high (>35 ppb) FeNO groups (n = 24, 26, and 47). In analysis including all patients and subanalysis including nonsmokers or former smokers, WA% in the 6th generation airways was consistently higher in the high FeNO group than in the low FeNO group, whereas WA% in the 3rd to 5th generation airways was not. In multivariable models, WA% in the 6th generation airways and the rate of mucus plugging were higher in the high FeNO group than in the low FeNO group after adjusting for age, sex, body mass index, smoking status, lung volume, and allergic rhinitis presence. CONCLUSIONS Higher FeNO may reflect the inflammation and remodeling of relatively peripheral airways in asthma in both former smokers and nonsmokers.
Collapse
Affiliation(s)
- Yusuke Hayashi
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Naoya Tanabe
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | - Hisako Matsumoto
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan; Department of Respiratory Medicine & Allergology, Kindai University Faculty of Medicine, Osakasayama, Japan
| | - Kaoruko Shimizu
- Division of Emergent Respiratory and Cardiovascular Medicine, Hokkaido University Hospital, Sapporo, Japan
| | - Ryo Sakamoto
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tsuyoshi Oguma
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan; Department of Respiratory Medicine, Kyoto City Hospital, Kyoto, Japan
| | - Hironobu Sunadome
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan; Department of Respiratory Care and Sleep Control Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Atsuyasu Sato
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Susumu Sato
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan; Department of Respiratory Care and Sleep Control Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Toyohiro Hirai
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
36
|
Venegas Garrido C, Mukherjee M, Svenningsen S, Nair P. Eosinophil-mucus interplay in severe asthma: Implications for treatment with biologicals. Allergol Int 2024; 73:351-361. [PMID: 38485545 DOI: 10.1016/j.alit.2024.03.001] [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: 02/21/2024] [Accepted: 02/22/2024] [Indexed: 06/23/2024] Open
Abstract
Airway mucus is a hydrogel with unique biophysical properties due to its primary water composition and a small proportion of large anionic glycoproteins or mucins. The predominant mucins in human mucus, MUC5AC and MUC5B, are secreted by specialized cells within the airway epithelium both in normal conditions and in response to various stimuli. Their relative proportions are correlated with specific inflammatory responses and disease mechanisms. The dysregulation of mucin expression is implicated in numerous respiratory diseases, including asthma, COPD, and cystic fibrosis, where the pathogenic role of mucus has been extensively described yet often overlooked. In airway diseases, excessive mucus production or impaired mucus clearance leads to mucus plugging, with secondary airway occlusion that contribute to airflow obstruction, asthma severity and poor control. Eosinophils and Charcot Leyden crystals in sputum contribute to the mucus burden and tenacity. Mucin may also contribute to eosinophil survival. Other mechanisms, including eosinophil-independent IL-13 release, mast-cell activation and non-type-2 (T2) cytokines, are also likely to participate in mucus pathobiology. An accurate assessment of mucus and its clinical and functional consequences require a thorough approach that includes evaluation of cellular predominance in sputum, airway cytokines and other inflammatory markers, mucus characteristics and composition and structural and functional impact measured by advanced lung imaging. This review, illustrated with clinical scenarios, provides an overview of current methods to assess mucus and its relevance to the choice of biologics to treat patients with severe asthma.
Collapse
Affiliation(s)
- Carmen Venegas Garrido
- Division of Respirology, Department of Medicine, St Joseph's Healthcare & McMaster University, Hamilton, Ontario, Canada
| | - Manali Mukherjee
- Division of Respirology, Department of Medicine, St Joseph's Healthcare & McMaster University, Hamilton, Ontario, Canada
| | - Sarah Svenningsen
- Division of Respirology, Department of Medicine, St Joseph's Healthcare & McMaster University, Hamilton, Ontario, Canada
| | - Parameswaran Nair
- Division of Respirology, Department of Medicine, St Joseph's Healthcare & McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
37
|
Asano K. Mucus plugs in severe asthma and related airway diseases. Allergol Int 2024; 73:349-350. [PMID: 38906642 DOI: 10.1016/j.alit.2024.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Indexed: 06/23/2024] Open
Affiliation(s)
- Koichiro Asano
- Division of Pulmonary Medicine, Department of Medicine, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa 259-1193, Japan.
| |
Collapse
|
38
|
Pelaia C, Melhorn J, Hinks TS, Couillard S, Vatrella A, Pelaia G, Pavord ID. Type 2 severe asthma: pathophysiology and treatment with biologics. Expert Rev Respir Med 2024; 18:485-498. [PMID: 38994712 DOI: 10.1080/17476348.2024.2380072] [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: 05/05/2024] [Accepted: 07/10/2024] [Indexed: 07/13/2024]
Abstract
INTRODUCTION The hallmark of most patients with severe asthma is type 2 inflammation, driven by innate and adaptive immune responses leading to either allergic or non-allergic eosinophilic infiltration of airways. The cellular and molecular pathways underlying severe type 2 asthma can be successfully targeted by specific monoclonal antibodies. AREAS COVERED This review article provides a concise overview of the pathophysiology of type 2 asthma, followed by an updated appraisal of the mechanisms of action and therapeutic efficacy of currently available biologic treatments used for management of severe type 2 asthma. Therefore, all reported information arises from a wide literature search performed on PubMed. EXPERT OPINION The main result of the recent advances in the field of anti-asthma biologic therapies is the implementation of a personalized medicine approach, aimed to achieve clinical remission of severe asthma. Today this accomplishment is made possible by the right choice of the most beneficial biologic drug for the pathologic traits characterizing each patient, including type 2 severe asthma and its comorbidities.
Collapse
Affiliation(s)
- Corrado Pelaia
- Department of Medical and Surgical Sciences, University "Magna Græcia" of Catanzaro, Catanzaro, Calabria, Italy
| | - James Melhorn
- Nuffield Department of Medicine, Respiratory Medicine Unit, University of Oxford, Oxford, UK
| | - Timothy Sc Hinks
- Nuffield Department of Medicine, Respiratory Medicine Unit, University of Oxford, Oxford, UK
| | - Simon Couillard
- Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC, Canada
| | - Alessandro Vatrella
- Department of Medicine, Surgery and Dentistry, University of Salerno, Salerno, Italy
| | - Girolamo Pelaia
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, Catanzaro, Italy
| | - Ian D Pavord
- Nuffield Department of Medicine, Respiratory Medicine Unit, University of Oxford, Oxford, UK
| |
Collapse
|
39
|
Caminati M, Buhl R, Corren J, Hanania NA, Kim H, Korn S, Lommatzsch M, Martin N, Matucci A, Nasser SM, Pavord ID, Domingo C. Tezepelumab in patients with allergic and eosinophilic asthma. Allergy 2024; 79:1134-1145. [PMID: 38146651 DOI: 10.1111/all.15986] [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: 08/11/2023] [Revised: 12/05/2023] [Accepted: 12/10/2023] [Indexed: 12/27/2023]
Abstract
Asthma is a heterogeneous disease commonly driven by allergic and/or eosinophilic inflammation, both of which may be present in severe disease. Most approved biologics for severe asthma are indicated for specific phenotypes and target individual downstream type 2 components of the inflammatory cascade. Tezepelumab, a human monoclonal antibody (immunoglobulin G2λ), binds specifically to thymic stromal lymphopoietin (TSLP), an epithelial cytokine that initiates and sustains allergic and eosinophilic inflammation in asthma. By blocking TSLP, tezepelumab has demonstrated efficacy across known asthma phenotypes and acts upstream of all current clinically used biomarkers. In a pooled analysis of the phase 2b PATHWAY (NCT02054130) and phase 3 NAVIGATOR (NCT03347279) studies, compared with placebo, tezepelumab reduced the annualized asthma exacerbation rate over 52 weeks by 62% (95% confidence interval [CI]: 53, 70) in patients with perennial aeroallergen sensitization (allergic asthma); by 71% (95% CI: 62, 78) in patients with a baseline blood eosinophil count ≥300 cells/μL; and by 71% (95% CI: 59, 79) in patients with allergic asthma and a baseline blood eosinophil count ≥300 cells/μL. This review examines the efficacy and mode of action of tezepelumab in patients with allergic asthma, eosinophilic asthma and coexisting allergic and eosinophilic phenotypes.
Collapse
Affiliation(s)
- Marco Caminati
- Asthma Center and Allergy Unit, Verona Integrated University Hospital & Department of Medicine, University of Verona, Verona, Italy
| | - Roland Buhl
- Pulmonary Department, Mainz University Hospital, Mainz, Germany
| | - Jonathan Corren
- David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Nicola A Hanania
- Section of Pulmonary and Critical Care Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Harold Kim
- Department of Medicine, Western University, London, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Stephanie Korn
- IKF Pneumologie Mainz, Mainz, Germany
- Thoraxklinik Heidelberg, Heidelberg, Germany
| | - Marek Lommatzsch
- Department of Pneumology and Critical Care Medicine, University of Rostock, Rostock, Germany
| | - Neil Martin
- Respiratory and Immunology, BioPharmaceuticals Medical, AstraZeneca, Cambridge, UK
- University of Leicester, Leicester, UK
| | - Andrea Matucci
- Immunoallergology Unit, Careggi University Hospital, Florence, Italy
| | - Shuaib M Nasser
- Department of Allergy, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Ian D Pavord
- Respiratory Medicine, NIHR Oxford Biomedical Research Centre, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Christian Domingo
- Servei de Pneumologia, Corporació Sanitària Parc Taulí, Sabadell, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| |
Collapse
|
40
|
Barbas CSV. Editorial: Asthma: physiology and pathophysiology. Front Physiol 2024; 15:1403211. [PMID: 38651041 PMCID: PMC11033497 DOI: 10.3389/fphys.2024.1403211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 03/21/2024] [Indexed: 04/25/2024] Open
|
41
|
Varricchi G, Brightling CE, Grainge C, Lambrecht BN, Chanez P. Airway remodelling in asthma and the epithelium: on the edge of a new era. Eur Respir J 2024; 63:2301619. [PMID: 38609094 PMCID: PMC11024394 DOI: 10.1183/13993003.01619-2023] [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: 09/22/2023] [Accepted: 02/15/2024] [Indexed: 04/14/2024]
Abstract
Asthma is a chronic, heterogeneous disease of the airways, often characterised by structural changes known collectively as airway remodelling. In response to environmental insults, including pathogens, allergens and pollutants, the epithelium can initiate remodelling via an inflammatory cascade involving a variety of mediators that have downstream effects on both structural and immune cells. These mediators include the epithelial cytokines thymic stromal lymphopoietin, interleukin (IL)-33 and IL-25, which facilitate airway remodelling through cross-talk between epithelial cells and fibroblasts, and between mast cells and airway smooth muscle cells, as well as through signalling with immune cells such as macrophages. The epithelium can also initiate airway remodelling independently of inflammation in response to the mechanical stress present during bronchoconstriction. Furthermore, genetic and epigenetic alterations to epithelial components are believed to influence remodelling. Here, we review recent advances in our understanding of the roles of the epithelium and epithelial cytokines in driving airway remodelling, facilitated by developments in genetic sequencing and imaging techniques. We also explore how new and existing therapeutics that target the epithelium and epithelial cytokines could modify airway remodelling.
Collapse
Affiliation(s)
- Gilda Varricchi
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), School of Medicine, University of Naples Federico II, WAO Center of Excellence, Naples, Italy
- Institute of Experimental Endocrinology and Oncology (IEOS), National Research Council, Naples, Italy
- G. Varricchi and C.E. Brightling contributed equally
| | - Christopher E. Brightling
- Institute for Lung Health, NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
- G. Varricchi and C.E. Brightling contributed equally
| | - Christopher Grainge
- School of Medicine and Public Health, University of Newcastle, Callaghan, Australia
| | - Bart N. Lambrecht
- Center for Inflammation Research, Laboratory of Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, Ghent, Belgium
| | - Pascal Chanez
- Department of Respiratory Diseases, Aix-Marseille University, Marseille, France
| |
Collapse
|
42
|
Panettieri Jr R, Lugogo N, Corren J, Ambrose CS. Tezepelumab for Severe Asthma: One Drug Targeting Multiple Disease Pathways and Patient Types. J Asthma Allergy 2024; 17:219-236. [PMID: 38524099 PMCID: PMC10960583 DOI: 10.2147/jaa.s342391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 02/23/2024] [Indexed: 03/26/2024] Open
Abstract
Asthma is a heterogeneous inflammatory disease of the airways, affecting many children, adolescents, and adults worldwide. Up to 10% of people with asthma have severe disease, associated with a higher risk of hospitalizations, greater healthcare costs, and poorer outcomes. Patients with severe asthma generally require high-dose inhaled corticosteroids and additional controller medications to achieve disease control; however, many patients remain uncontrolled despite this intensive treatment. The treatment of severe uncontrolled asthma has improved with greater understanding of asthma pathways and phenotypes as well as the advent of targeted biologic therapies. Tezepelumab, a monoclonal antibody, blocks thymic stromal lymphopoietin, an epithelial cytokine that has multifaceted effects on the initiation and persistence of asthma inflammation and pathophysiology. Unlike other biologic treatments, tezepelumab has demonstrated efficacy across severe asthma phenotypes, with the magnitude of effects varying by phenotype. Here we describe the anti-inflammatory effects and efficacy of tezepelumab across the most relevant phenotypes of severe asthma. Across clinical studies, tezepelumab reduced annualized asthma exacerbation rates versus placebo by 63-71% in eosinophilic severe asthma, by 58-68% in allergic severe asthma, by 67-71% in allergic and eosinophilic severe asthma, by 34-49% in type 2-low asthma, and by 31-41% in oral corticosteroid-dependent asthma. Furthermore, in all these asthma phenotypes, tezepelumab demonstrated higher efficacy in reducing exacerbations requiring hospitalizations or emergency department visits versus placebo. In patients with severe uncontrolled asthma, who commonly have multiple drivers of inflammation and disease, tezepelumab may modulate airway inflammation more extensively, as other available biologics block only specific downstream components of the inflammatory cascade.
Collapse
Affiliation(s)
- Reynold Panettieri Jr
- Rutgers Institute for Translational Medicine and Science, Rutgers University, New Brunswick, NJ, USA
| | - Njira Lugogo
- Michigan Medicine Asthma Program, University of Michigan, Ann Arbor, MI, USA
| | - Jonathan Corren
- Departments of Medicine and Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | | |
Collapse
|
43
|
Krings JG, Gierada DS. Do Biologic Therapies Decrease Mucus Plugging in Asthma? NEJM EVIDENCE 2023; 2:EVIDe2300179. [PMID: 38320185 DOI: 10.1056/evide2300179] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2024]
Abstract
Asthma researchers have long recognized that abnormal mucus production and clearance play a role in the pathophysiology of asthma.1 Mucus plugs are known to be common in patients with severe asthma, and mucus plug scores, for which higher scores indicate more severe plugging, are directly correlated with airflow obstruction and markers of eosinophilic airway inflammation (i.e., higher scores or marker levels are associated with more severe obstruction). Other work has shown that mucus plugs were associated with distal deficits in regional ventilation as delineated by hyperpolarized gas magnetic resonance imaging.2,3.
Collapse
Affiliation(s)
- James G Krings
- Division of Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis
| | - David S Gierada
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis
| |
Collapse
|