Heterogeneity of Inflammatory Processes and Pathways Driving Chronic Obstructive Pulmonary Disease Pathology - European Medical Journal

Heterogeneity of Inflammatory Processes and Pathways Driving Chronic Obstructive Pulmonary Disease Pathology

7 Mins
Respiratory
Download PDF
Chairpeople:
Klaus Rabe,1 Henrik Watz2
Speakers:
Mona Bafadhel,3 Stephanie Christenson,4 Alberto Papi,5 Klaus Rabe,1 Paola Rogliani,6 Henrik Watz2
Disclosure:

Rabe has received grants and research support from the German Federal Ministry for Research and Technology (BMFT); honoraria or consultation fees from AstraZeneca, Berlin-Chemie, Boehringer Ingelheim, Chiesi, GlaxoSmithKline, Menarini, Novartis, Sanofi, Regeneron, and Verona Pharma. Rabe serves as the Director of the Airway Research Centre North (ARCN) within the Deutsche Zentrum für Lungenforschung (DZL). Watz has received consulting fees, payment or honoraria for lectures, presentations, speakers’ bureaus, manuscript writing, or educational events; support for attending meetings and/or travel; and participation on a data safety monitoring board or advisory board, all from AstraZeneca, Boehringer Ingelheim, Chiesi, GlaxoSmithKline, Novartis, and Sanofi. Watz is disease area coordinator of COPD for the German Centre for Lung Research and coordinator of the German COPD guideline. Bafadhel has received grants and research support from AstraZeneca, Roche, Sanofi, and GlaxoSmithKline; honoraria and consulting fees from AstraZeneca, Sanofi, and GlaxoSmithKline; and acted as a scientific advisor for Areteria and AlbusHealth. Christenson has acted as a consultant for AstraZeneca, GlaxoSmithKline, Glenmark, and Sanofi/Regeneron; and has provided writing services for UpToDate. Papi has been a board member and/or received grants, research, consulting, lecture, or travel support from Chiesi Farmaceutici, AstraZeneca, GlaxoSmithKline, Mundipharma Research Limited, Novartis, Sanofi, Menarini, Zambon, Edmond Pharma, and Elpen. Rogliani has received grants and research support from Arcede, AstraZeneca, Boehringer Ingelheim, Chiesi Farmaceutici, GlaxoSmithKline, Menarini Group, Novartis, Sanofi, and Verona Pharma; and honoraria or consultation fees from AstraZeneca, Boehringer Ingelheim, Chiesi Farmaceutici, GlaxoSmithKline, Menarini Group, Novartis, Recipharm, Sanofi, and Zambon.

Acknowledgements:

Medical writing assistance was provided by International Meetings and Science (IMsci), Stamford, Connecticut, USA.

Support:

The symposia were sponsored by Sanofi and by Regeneron Pharmaceuticals Inc., and this article was commissioned and supported by the same companies. Sanofi and Regeneron Pharmaceuticals Inc. have reviewed this article for medical accuracy.

Citation:
EMJ Respir. ;12[1]:63-72. https://doi.org/10.33590/emjrespir/IKHC2212.
Keywords:
Chronic obstructive pulmonary disease (COPD), pathophysiology, exacerbations, IL-4, IL-13, IL-5, IL-33, lung function, Type 2 inflammation.

Each article is made available under the terms of the Creative Commons Attribution-Non Commercial 4.0 License.

Meeting Summary

Two symposia occurred during the European Respiratory Society (ERS) Congress 2024, highlighting the heterogeneity in chronic inflammatory pathways that underlie chronic obstructive pulmonary disease (COPD) pathophysiology. In ‘A Breath of Fresh Air: A Greater Understanding of COPD With Type 2 Inflammation’, Henrik Watz (Chair), German Center for Lung Research, Grosshansdorf, Germany, provided an overview of both the pathophysiology and the burden of disease of COPD. He discussed how exacerbations, which may be increased in those with evidence of Type 2 inflammation, contribute to the cycle of worsening COPD. Mona Bafadhel, King’s College London, UK, provided an examination of the mechanisms and biomarkers of Type 2 inflammation in COPD. Finally, Alberto Papi, University of Ferrara, Italy, summarised the latest research on biological treatments targeting Type 2 inflammation in COPD. The second symposium, ‘Targeting interleukin-33 (IL-33) in COPD: Exploring New Frontiers for COPD Management’, discussed inflammation in COPD, focusing on the central role of IL-33 as a mediator for both Type 2 and Type 1/Type 3 inflammation. Klaus Rabe (Chair), LungenClinic, Grosshansdorf, Germany, reviewed the structure and function of IL-33 and its initial processes that lead to downstream immune responses. Stephanie Christenson, University of California, San Francisco, USA, explored how genetic and environmental factors contribute to IL-33 activity in COPD pathology. Next, Paola Rogliani, University of Rome ‘Tor Vergata’, Italy, presented an examination of IL-33 inflammatory processes and evidence from COPD animal models illustrating the role of IL-33 in airway inflammation and lung function decline. Klaus Rabe concluded with an examination of IL-33 as a target for new COPD treatment approaches.

Introduction

A persistent, often progressive, lung disease, COPD results from chronic inflammation that leads to structural changes resulting in airway remodelling and airflow obstruction.1,2 However, just as individuals with COPD can present with different symptom patterns, the inflammatory pathways involved in COPD pathophysiology are also heterogeneous.1 Although Type 1/Type 3 inflammation drives COPD in most individuals, up to ~30−40% have Type 2 inflammation, and there is also potential overlap in inflammatory mechanisms.3-5

A Breath of Fresh Air: A Greater Understanding of COPD with Type 2 Inflammation

Living with COPD: Linking Inflammation with Patient Burden

Henrik Watz

Chronic inflammation, triggered by irritants and oxidative stress (eg., tobacco smoke, pollutants), is the key factor driving the airway and parenchymal structural changes that characterise COPD and limit airflow in the lungs (Figure 16).1,2,7 Pathophysiological changes in the airway specifically linked to chronic inflammation in COPD include barrier disruption, goblet cell hyperplasia, inflammation and excess mucus, and remodelling. Parenchymal structural changes include breakdown of the alveolar membranes (emphysema) and air trapping.1,2 Smoking, pollutants, environmental or occupational exposure, abnormal lung growth and development, and genetics and early life events all increase the risk of chronic inflammation leading to COPD.1 Even in individuals with COPD who have stopped smoking, the risk of exacerbations, morbidity, and mortality remains elevated for years.8

Figure 1: COPD is driven by chronic inflammation and structural changes.6
COPD: chronic obstructive pulmonary disease.

Traditionally, COPD was informally categorised based on the clinical observation of predominant symptoms, including the ‘pink puffer’ emphysema phenotype, associated with dyspnoea, hyperinflation, air trapping, and cachexia, and the ‘blue bloater’ chronic bronchitis phenotype, associated with chronic productive cough, respiratory infections, exacerbations, and being overweight.1,9 These characterisations were of limited clinical utility, only applicable for describing severe disease, and insufficient to encompass COPD pathophysiological or phenotype heterogeneity.9,10 Currently, COPD categorisation reflects an evolving, broader disease spectrum that includes emphysema- and bronchitis-predominant phenotypes, as well as a frequent exacerbator or rapid decliner phenotype, and two endotypes based on the underlying inflammatory pathway: 1) Type 1/Type 3 and 2) Type 2.1,9,11-14

In addition to increasing morbidity and mortality, COPD places a heavy physical and emotional burden on individuals and contributes to physical debilitation and poor health-related quality of life.1,15,16 In interviews, individuals with persistent COPD symptoms cite exhaustion, breathlessness, feelings of depression and dread, limits on physical activity, and difficulties in lying down at night to sleep.17

More than 80% of individuals with COPD are estimated to have >1 comorbid condition.18 Associated pulmonary conditions include bronchiectasis, asthma, pulmonary arterial hypertension, and an increased risk of lung cancer.1,19,20 Extrapulmonary conditions seen in individuals with COPD include cardiovascular (CV) conditions, such as heart failure, ischaemic heart disease, and arrhythmias; osteoporosis; renal failure; metabolic syndrome; diabetes; gastro-oesophageal reflux disease; and neurologic or psychiatric conditions.1,19-21 Multi-morbidity significantly impacts quality of life and health outcomes and is associated with increased COPD exacerbations, hospitalisations, medical costs, and mortality.22

Exacerbations of COPD increase disease burden, hospitalisations, and re-hospitalisations, and are associated with a poor prognosis. An analysis of 32 studies found 9−26% and 18−39% of COPD hospitalisations resulted in readmission within 30 days and 90 days, respectively. The most common significant risk factors for readmission were comorbidities, previous exacerbations and hospitalisations, and longer initial hospital stay, with frequent COPD exacerbations increasing readmission risk 2.5-fold.23 Patients who experience more severe exacerbations tend to have an increased incidence of CV events.24 Exacerbations also lead to an increased risk of CV death, whether the patient is hospitalised or not.25 Increased frequency and severity of exacerbations have been associated with increased rates of all-cause mortality, COPD-related mortality rate, and future exacerbations.26,27 Importantly, repeated exacerbations lead to progressive and potentially irreversible lung damage. Following a moderate or severe COPD exacerbation, forced expiratory volume in 1 second (FEV1) may not reach a patient’s pre-exacerbation levels, even after ≥1 month.28 Further, individuals who have frequent exacerbations show a significantly faster decline in FEV1 than those with infrequent exacerbations.29 Severe lung function impairment has been shown to be significantly associated with increased 90-day post-discharge mortality.30 In all, low FEV1 is a risk factor for COPD exacerbations and hospitalisations, contributing to a vicious cycle of exacerbations and declining lung function.13

Evidence from studies investigating increased blood eosinophil (EOS) counts, which can correspond to increased lung EOS numbers and elevated markers of Type 2 inflammation in the airways, supports a higher disease burden for individuals with COPD associated with Type 2 inflammation.1,31 An analysis of data from the Genetic Epidemiology of COPD study in 1,553 individuals with COPD found the risk for COPD exacerbations increased with increasing blood EOS counts, particularly for those with blood EOS ≥300 cells/µL.31 The Canadian Cohort of Obstructive Lung Disease study examined blood EOS counts in participants aged ≥40 years with and without COPD. The study found that the annual decline in FEV1 for those with EOS ≥300 cells/µL was significantly greater than for those with lower EOS levels (P≤0.01).32 Elevated EOS count (≥300 cells/µL) has been shown to be a marker for COPD-related rehospitalisation at 30 days up to 12 months and for all-cause rehospitalisation at 90 days up to 12 months.33 In an observational study, individuals with COPD with ≥300 EOS cells/µL were 3.21 times more likely to be readmitted to the hospital for COPD than those with EOS <300 EOS cells/µL.34

In summary, Watz emphasised that worsening disease in COPD contributes to a vicious cycle.35 Increased chronic inflammation and its attendant structural damage worsens symptoms and increases exacerbation risk, leading to a progressive decline in lung function.1,2,7

Unravelling the Role of Type 2 Inflammation in COPD

Mona Bafadhel

The Type 1/Type 3 and Type 2 endotypes of COPD have distinct characteristics.1,14 Type 1/Type 3 inflammation is marked by elevations in sputum neutrophils, Type 1 cytotoxic and T helper (Th) 1 and 17 cells, and innate lymphoid cell (ILC) 3, whereas Type 2 inflammation markers include elevated sputum EOS, Th2 cells, ILC2, and Type 2 cytokines IL-4, IL-13, and IL-5.36-40

Bafadhel underscored the potential roles of Type 2 cytokines in COPD with Type 2 inflammation, including airway remodelling, goblet cell hyperplasia, mucus production, and EOS trafficking to tissues (Figure 26).41-45

Figure 2: Potential roles of Type 2 cytokines in COPD with Type 2 inflammation.6
COPD: chronic obstructive pulmonary disease; IgE: immunoglobulin E; IL: interleukin; ILC2: group 2 innate lymphoid cell; MMP: matrix metalloproteinase; MUC: mucin; PGD2: prostaglandin D2 ; Th2: T helper cell Type 2; TSLP: thymic stromal lymphopoietin.

Blood EOS is recognised by the Global Initiative for Chronic Obstructive Lung Disease (GOLD) as a biomarker for inhaled corticosteroid (ICS) use, with levels ≥300 cells/μL indicating a higher likelihood that ICS may prevent COPD exacerbations.1 However, COPD endotyping remains an emerging concept, and the lack of established biomarkers to differentiate inflammatory profiles complicates the treatment decision-making process.11,46,47 To date, fibrinogen remains the only circulating biomarker approved by the US Food and Drug Administration (FDA) and the European Medicines Agency (EMA) to assess the risk of COPD exacerbations and mortality.48 Other biomarkers that have been investigated in relation to COPD clinical outcomes include blood and sputum EOS count, fractional exhaled nitric oxide (FeNO), and alpha-1 antitrypsin.10,48,49

Bafadhel highlighted the potential of biomarkers to improve COPD understanding and treatment. She emphasised their practicality and ease of use in clinical settings, suggesting that with further research, these biomarkers could play a larger role in personalised COPD therapy. To highlight these points, she summarised several studies illustrating the potential utility of EOS and FeNO in assessing COPD disease and treatment. In a study of 226 individuals with COPD, the annualised rate of moderate and/or severe acute COPD exacerbations was significantly higher in individuals with ≥2 visits where FeNO measured ≥20 parts per billion than in individuals with 0 or 1 visit (P<0.05).50 Elevated EOS was associated with an increased incidence rate ratio of severe and moderate COPD exacerbations in a study of >7,000 individuals with COPD participating in the Copenhagen General Population Study.51 Another analysis of data from this study found that blood EOS ≥300 cells/µL and FeNO ≥20 parts per billion separately and together were significantly associated with FEV1 decline (P≤0.004).52 Lastly, Bafadhel showed data supporting EOS and FeNO as biomarkers to predict ICS responsiveness in COPD. In a post hoc analysis of a randomised trial, annual COPD exacerbation rates were lower with budesonide+formoterol than with formoterol alone in individuals with blood EOS ≥340 cells/μL, but not in individuals with blood EOS <100 cells/μL.53 A separate study showed that ICS plus a long-acting β2 agonist reduced COPD Assessment Test scores by 7.20 points from baseline in individuals with COPD and high baseline FeNO, but was not as effective for improving this measure in those with low baseline FeNO.54

On Target: Perspectives From the Evolving Biologic Landscape in COPD

Alberto Papi

The 2024 GOLD algorithm for therapy escalation after a COPD exacerbation uses blood EOS levels to guide decisions.1 According to GOLD, all individuals experiencing an exacerbation should be treated with a long-acting β-agonist plus a long-acting muscarinic antagonist, but those with EOS ≥300 cells/μL also should receive ICS.1 Despite the optimisation of inhaled treatment with triple therapy, some individuals remain at risk for exacerbations. Papi reviewed the evolving COPD treatment landscape and presented data from clinical studies in COPD for biological therapies targeting steps in the Type 2 inflammatory pathway.

Targeting IL-33 in COPD: Exploring New Frontiers for COPD Management

Understanding the Role of IL-33 Size and Activity in the Pathophysiology of COPD

Klaus Rabe

Rabe reviewed the complexity and diversity of the inflammatory pathways associated with COPD pathology and discussed the structure and function of IL-33. Full-length IL-33 (IL-33FL) secretion from airway epithelial cells is induced in response to inhaled irritants, pathogens, and allergens. In this form, IL-33FL can be cleaved by proteases from inflammatory cells or allergens to form hyperactive, mature IL-33, which has 30- to 60-fold higher activity than IL-33FL, or inactivated by oxidation, sequestration, and proteolytic degradation. Mature, activated IL-33 stimulates ST2+ immune cells, triggering both Type 1 and Type 2 inflammation that can underpin COPD pathology. However, inactivated IL-33 may induce ST2-independent signalling and also promote a COPD phenotype.55,56 Because of its key role in the pathophysiology of different COPD endotypes and phenotypes, IL-33 is a promising therapeutic target.

What Do We Know About IL-33 Genetics?

Stephanie Christenson

Christenson discussed how IL-33 activity in COPD pathology involves a convergence of genetic variations and environmental exposures. To characterise the impact of IL-33 genetics on COPD risk, investigators performed an analysis of IL-33 loss-of-function (LOF) and gain-of-function (GOF) variants known to affect asthma risk among individuals of European ancestry utilising data from the UK Biobank and Geisinger Health System studies in COPD.57 Three variants were selected for analysis: 1) a rare IL33 LOF slice-acceptor allele that reduces total IL33 messenger RNA (mRNA) and decreases asthma risk; 2) a common IL33 intronic GOF variant that increases IL33 mRNA and asthma risk; and 3) a common IL1RL1 (gene that encodes the IL-33 receptor) intronic GOF variant that lowers plasma levels of soluble IL-33 receptor and increases asthma risk. The results showed that the rare LOF variant was associated with a 21% reduction in the risk of COPD and a reduction in blood EOS (P<0.0001), but the two common GOF variants were associated with significantly increased odds of COPD (P<0.05 for each variant).57

Next, Christenson reviewed how smoking influences IL-33 activity in COPD and highlighted the importance of considering the complex effects of smoking on IL-33 activity within the context of COPD. An analysis of IL-33 levels in 62 individuals with COPD showed that smoking pack-years was positively correlated with IL-33 expression in the lungs (R=-0.58). In this study, a larger proportion of individuals with high IL-33 had previously regularly smoked cigarettes than patients with low IL-33; in contrast, those who were currently regularly smoking cigarettes tended to have low IL-33.58 This trend of individuals who currently regularly smoke having lower IL-33 expression than those who formerly regularly smoked has been confirmed in multiple studies of samples collected by bronchial biopsy or bronchial brushings.59 This finding may be due to the shifting of basal cells towards a more differentiated state in those who currently regularly smoke, and may explain the clinical observation of greater efficacy of anti-IL-33 treatment in those with COPD who previously regularly smoked.59 However, even in those with COPD who currently regularly smoke, high IL-33 expression has been associated with lower percent predicted median residual volume and lower percent predicted and percent mean residual volume to total lung capacity.60

The Role of IL-33 in COPD Pathophysiology and Its Impact on Disease Remodelling

Paola Rogliani

Rogliani began with a closer look at the key role IL-33 plays in COPD as an initiator and amplifier of broad inflammatory cascades. Stimulation from smoke, pollutants, infections, or oxidative stress induces IL-33 production and secretion from the airway epithelium and the endothelium of blood vessels in the lungs.55,61 IL-33 then activates immune cells and non-haematopoietic lung cells to promote immune responses and increase inflammation (Figure 362).61 As a central mediator of inflammation in COPD, IL-33 drives both Type 1/Type 3 and Type 2 inflammatory responses, and stimulation of IL-33 can lead to increased neutrophil and blood EOS levels.5,45,63 The chronic inflammation initiated and amplified by IL-33 results in airway remodelling, including airway smooth muscle thickening and fibrosis in small airways.7,64

Figure 3: IL-33 mediation of immune response and airway obstruction and remodelling in COPD.62
ASM: airway smooth muscle; BV: blood vessel; COPD: chronic obstructive pulmonary disease; DC: dendritic cell;
ECM: extracellular matrix.

Evidence from animal models illustrates how IL-33 plays a role in both Type 1/Type 3 and Type 2 inflammatory responses.65,66 In mice, systemic IL-33 overexpression produced through hydrodynamic DNA delivery induced significant neutrophilia, associated with Type 1/Type 3 inflammation, and eosinophilia, associated with Type 2 inflammation, in lung tissue, as well as production of Type 1 and Type 2 inflammatory cytokines.65 A separate study in mice found that prolonged exposure to house dust mites resulting in upregulation of IL-33 induced features of severe airway diseases, such as smooth muscle thickening, increased collagen deposition, and epithelial degeneration.66 Although initial inflammatory responses had Type 2 characteristics, including increased EOS, longer exposures led to a mixed inflammatory phenotype, with increased neutrophils and other markers of Type 1/Type 3 inflammation.66

IL-33: A Promising Target

Klaus Rabe

Rabe reviewed the landscape of available therapies and those in development that target the chronic inflammation underlying COPD pathophysiology. He also presented data supporting IL-33 as a potential target for COPD treatment. Ongoing research is refining the understanding of IL-33 in COPD, focusing on identifying ideal patient subgroups for IL–33–targeted therapies, and evaluating the long-term efficacy and safety of these interventions. In all, Rabe articulated a shared vision of more personalised approaches in medicine, where treatments are tailored to individual patients based on their unique biological mechanisms.

Conclusion

As illustrated by the presentations in these symposia, COPD is a heterogeneous condition with variations in symptomatology, the events that trigger the chronic inflammation driving the disease processes, and the nature of the inflammatory pathways. Understanding the heterogeneity of inflammation in COPD pathophysiology helps to guide diagnosis and treatment. Optimal COPD treatment is critical to arresting the vicious disease cycle of exacerbations and worsening lung function that increases disease morbidity and mortality.

 

MAT-GLB-2406080 – 1.0 – 10/2024

References
Global Initiative for Chronic Obstructive Lung Disease (GOLD). Global strategy for the diagnosis, management, and prevention of chronic obstructive pulmonary disease. Available at: https://goldcopd.org/2024-gold-report/. Last accessed: 26 August 2024. Barnes PJ. Inflammatory mechanisms in patients with chronic obstructive pulmonary disease. J Allergy Clin Immunol. 2016;138(1):16-27. Singh D et al; ECLIPSE investigators. Eosinophilic inflammation in COPD: prevalence and clinical characteristics. Eur Respir J. 2014;44(6):1697-700. Leigh R et al. Stable COPD: predicting benefit from high-dose inhaled corticosteroid treatment. Eur Respir J. 2006;27(5):964-71. Yousuf A et al. T2 biologics for chronic obstructive pulmonary disease. J Allergy Clin Immunol Pract. 2019;7(5):1405-16. Watz H. A breath of fresh air: a greater understanding of COPD with type 2 inflammation. Presentation Session 290. 7-11 September 2024. Linden D et al. Respiratory viral infection: a potential "missing link" in the pathogenesis of COPD. Eur Respir Rev. 2019;28(151):180063. Godtfredsen NS et al. COPD-related morbidity and mortality after smoking cessation: status of the evidence. Eur Respir J. 2008;32(4):844-53. Lan LTT, Dinh-Xuan AT. Pathophysiology updates for chronic obstructive pulmonary disease. Curr Respir Care Rep. 2013;2:139-44. Hersh CP et al. COPDGene and ECLIPSE Investigators. Non-emphysematous chronic obstructive pulmonary disease is associated with diabetes mellitus. BMC Pulm Med. 2014;14:164. Kakavas S et al. Pulmonary function testing in COPD: looking beyond the curtain of FEV1. NPJ Prim Care Respir Med. 2021;31(1):23. Castaldi PJ et al. Cluster analysis in the COPDGene study identifies subtypes of smokers with distinct patterns of airway disease and emphysema. Thorax. 2014;69(5):415-22. Garcia-Aymerich J et al. Lung function impairment, COPD hospitalisations and subsequent mortality. Thorax. 2011;66(7):585-90. Aksoy E et al. Neutrophil to lymphocyte ratio is a better indicator of COPD exacerbation severity in neutrophilic endotypes than eosinophilic endotypes. Int J Chron Obstruct Pulmon Dis. 2018;13:2721-30. Watz H et al. Physical activity in patients with COPD. Eur Respir J. 2009;33(2):262-72. Miravitlles M, Ribera A. Understanding the impact of symptoms on the burden of COPD. Respir Res. 2017;18(1):67. Cook N et al. Impact of cough and mucus on COPD patients: primary insights from an exploratory study with an online patient community. Int J Chron Obstruct Pulm Dis. 2019;14:1365-76. Putcha N et al. Comorbidities and chronic obstructive pulmonary disease: prevalence, influence on outcomes, and management. Semin Respir Crit Care Med. 2015;36(4):575-91. Barnes PJ, Celli BR. Systemic manifestations and comorbidities of COPD. Eur Respir J. 2009;33(5):1165-85. Dal Negro RW et al. Prevalence of different comorbidities in COPD patients by gender and GOLD stage. Multidiscip Respir Med. 2015;10(1):24. Gaddam S et al. Prevalence of chronic kidney disease in patients with chronic obstructive pulmonary disease: a systematic review and meta-analysis. BMC Pulm Med. 2016;16(1):158. Le TT et al. Prevalence and newly diagnosed rates of multimorbidity in older Medicare beneficiaries with COPD. COPD. 2021;18(5):541-8. Alqahtani JS et al. Risk factors for all-cause hospital readmission following exacerbation of COPD: a systematic review and meta-analysis. Eur Respir Rev. 2020;29(156):190166. Graul EL et al. Temporal risk of nonfatal cardiovascular events after chronic obstructive pulmonary disease exacerbation: A population-based study. Am J Respir Crit Care Med. 2024;209(8):960-72. Kunisaki KM et al. Exacerbations of chronic obstructive pulmonary disease and cardiac events. A post hoc cohort analysis from the SUMMIT randomized clinical trial. Am J Respir Crit Care Med. 2018;198(1):51-7. Whittaker H et al. Frequency and severity of exacerbations of COPD associated with future risk of exacerbations and mortality: A UK routine health care data study. Int J Chron Obstruct Pulm Dis. 2022;17:427-37. Rothnie KJ et al. Natural history of chronic obstructive pulmonary disease exacerbations in a general practice-based population with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2018;198(4):464-71. Watz H et al. Spirometric changes during exacerbations of COPD: a post hoc analysis of the WISDOM trial. Respir Res. 2018;19(1):251. Donaldson GC et al. Relationship between exacerbation frequency and lung function decline in chronic obstructive pulmonary disease. Thorax. 2002;57(10):847-52. Hartl S et al. Risk of death and readmission of hospital-admitted COPD exacerbations: European COPD Audit. Eur Respir J. 2016;47(1):113-21. Yun JH et al. Blood eosinophil count thresholds and exacerbations in patients with chronic obstructive pulmonary disease. J Allergy Clin Immunol. 2018;141(6):2037-47.e10. Tan WC et al. High eosinophil counts predict decline in FEV1: results from the CanCOLD study. Eur Respir J. 2021;57(5):2000838. Hegewald MJ et al. Blood eosinophil count and hospital readmission in patients with acute exacerbation of chronic obstructive pulmonary disease. Int J Chron Obstruct Pulm Dis. 2020;15:2629-41. Couillard S et al. Eosinophils in COPD exacerbations are associated with increased readmissions. Chest. 2017;151(2):366-73. Agusti AGN. COPD, a multicomponent disease: implications for management. Respir Med. 2005;99(6):670-82. Barnes PJ. Inflammatory endotypes in COPD. Allergy. 2019;74(7):1249-56. Chen L et al. Imbalance between subsets of CD8(+) peripheral blood T cells in patients with chronic obstructive pulmonary disease. Peer J. 2016;4:e2301. Ghebre MA et al. Biological exacerbation clusters demonstrate asthma and chronic obstructive pulmonary disease overlap with distinct mediator and microbiome profiles. J Allergy Clin Immunol. 2018;141(6):2027-36.e12. Barczyk A et al. Cytokine production by bronchoalveolar lavage T lymphocytes in chronic obstructive pulmonary disease. J Allergy Clin Immunol. 2006;117(6):1484-92. Christenson SA et al. Asthma-COPD overlap. Clinical relevance of genomic signatures of type 2 inflammation in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2015;191(7):758-66. Cooper PR et al. Involvement of IL-13 in tobacco smoke-induced changes in the structure and function of rat intrapulmonary airways. Am J Respir Cell Mol Biol. 2010;43(2):220-6. Doyle AD et al. Eosinophil-derived IL-13 promotes emphysema. Eur Respir J. 2019;53(5):1801291. Zheng T et al. Inducible targeting of IL-13 to the adult lung causes matrix metalloproteinase- and cathepsin-dependent emphysema. J Clin Invest. 2000;106(9):1081-93. Alevy YG et al. IL-13-induced airway mucus production is attenuated by MAPK13 inhibition. J Clin Invest. 2012;122(12):4555-68. Gandhi NA et al. Targeting key proximal drivers of type 2 inflammation in disease. Nat Rev Drug Discov. 2016;15(1):35-50. Petersen H et al. Early endotyping: a chance for intervention in chronic obstructive pulmonary disease. Am J Respir Cell Mol Biol. 2018;59(1):13-7. Stockley RA et al. Chronic obstructive pulmonary disease biomarkers and their interpretation. Am J Respir Crit Care Med. 2019;199(10):1195-204. Cazzola M et al. Evolving concepts in chronic obstructive pulmonary disease blood-based biomarkers. Mol Diagn Ther. 2019;23(5):603-14. Tang B et al. Relationship of blood eosinophils with fractional exhaled nitric oxide and pulmonary function parameters in chronic obstructive pulmonary disease (COPD) exacerbation. Med Sci Monit. 2020;26:e921182. Alcázar-Navarrete B et al. Persistently elevated exhaled nitric oxide fraction is associated with increased risk of exacerbation in COPD. Eur Respir J. 2018;51(1):1701457. Vedel-Krogh S et al. Blood eosinophils and exacerbations in chronic obstructive pulmonary disease. The Copenhagen General Population Study. Am J Respir Crit Care Med. 2016;193(9):965-74. Çolak Y et al. Type-2 inflammation and lung function decline in chronic airway disease in the general population. Thorax. 2024;79(4):349-58. Bafadhel M et al. Predictors of exacerbation risk and response to budesonide in patients with chronic obstructive pulmonary disease: a post-hoc analysis of three randomised trials. Lancet Respir Med. 2018;6(2):117-26. Wu YK et al. Treatment of chronic obstructive pulmonary disease in patients with different fractional exhaled nitric oxide levels. Medicine (Baltimore). 2018;97(47):e11922. Cayrol C, Girard JP. Interleukin-33 (IL-33): a critical review of its biology and the mechanisms involved in its release as a potent extracellular cytokine. Cytokine. 2022;156:155891. Strickson S et al. Oxidised IL-33 drives COPD epithelial pathogenesis via ST2-independent RAGE/EGFR signalling complex. Eur Respir J. 2023;62(3):2202210. Rabe KF et al. Safety and efficacy of itepekimab in patients with moderate-to-severe COPD: a genetic association study and randomised, double-blind, phase 2a trial. Lancet Respir Med. 2021;9(11):1288-98. Session 450. Joo H et al. Association between plasma interleukin-33 level and acute exacerbation of chronic obstructive pulmonary disease. BMC Pulm Med. 2021;21(1):86. Faiz A et al. IL-33 Expression is lower in current smokers at both transcriptomic and protein levels. Am J Respir Crit Care Med. 2023;208(10):1075-87. Kim SW et al. Factors associated with plasma IL-33 levels in patients with chronic obstructive pulmonary disease. Int J Chron Obstruct Pulm Dis. 2017;12:395-402. Donovan C, Hansbro PM. IL-33 in chronic respiratory disease: from preclinical to clinical studies. ACS Pharmacol Transl Sci. 2019;3(1):56-62. Rabe KF. Targeting IL-33 in COPD: exploring new frontiers for COPD management. Presentation Session 450. 7-11 September 2024. Chan BCL et al. IL33: roles in allergic inflammation and therapeutic perspectives. Front Immunol. 2019;10:364. Aghapour M et al. Airway epithelial barrier dysfunction in chronic obstructive pulmonary disease: role of cigarette smoke exposure. Am J Respir Cell Mol Biol. 2018;58(2):157-69. Srivatsan S et al. IL-33 drives type 1 and type 2 inflammation and instructs airway remodeling. Presented at: American Thoracic Society 2023 International Conference; 19-24 May 2023. Allinne J et al. IL-33 blockade affects mediators of persistence and exacerbation in a model of chronic airway inflammation. J Allergy Clin Immunol. 2019;144(6):1624-37.e10.

Rate this content's potential impact on patient outcomes

Thank you!

Please share some more information on the rating you have given