Next Article in Journal
Electrocardiographic Predictors of High-Risk Patent Foramen Ovale Anatomy Defined by Transesophageal Echocardiography
Previous Article in Journal
Psychological Effects of Hemodialysis on Patients with Renal Failure: A Cross-Sectional Study
Previous Article in Special Issue
Oxidative Stress and Risk Factors in Adult Patients with Bronchial Asthma: A Clinical Analysis of Representative Biomarkers
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Neutrophilic Asthma—From Mechanisms to New Perspectives of Therapy

Department of Allergology, Medical University of Gdansk, 80-210 Gdansk, Poland
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2025, 14(20), 7137; https://doi.org/10.3390/jcm14207137
Submission received: 25 August 2025 / Revised: 26 September 2025 / Accepted: 7 October 2025 / Published: 10 October 2025
(This article belongs to the Special Issue Advances in Asthma: 2nd Edition)

Abstract

Neutrophilic asthma (NA) is an inflammatory phenotype of asthma, characterized by predominantly neutrophilic infiltrations in bronchial mucosa. It is usually diagnosed on the basis of high neutrophil count in induced sputum (from >40% to >76%) with low eosinophils (<2%). The prevalence of NA ranges from 16% to 28% of the adult asthma population depending on the definitions and study methods applied. A clinical picture of NA is characterized by late onset of symptoms, higher exacerbation rate, lower level of symptoms control, and poorer response to steroids compared to eosinophilic phenotype. Comorbidities such as obesity and GERD as well as the influence of environmental factors (air pollution, smoking, bacterial infections) contribute to the development and severe course of the disease. NA is T2-low disease with predominantly Th1/Th17-type inflammation. Neutrophils are key cells responsible for initiating and sustaining inflammation. In addition to their primary functions like phagocytosis, degranulation, and NETosis, neutrophils release several pro-inflammatory cytokines (IL-1α, IL-1β, IL-6, TNF) and chemokines (CXCL-1, -2, -8, -9, -10) responsible for the recruitment of other neutrophils or T cells. Increasing knowledge about the biology of neutrophiles and their role in asthma results in new potential therapies that could improve control of NA, particularly new biologicals targeting Th1/Th17-related cytokines. In this review, we discuss the prevalence, mechanisms, and clinical features of neutrophilic asthma. Furthermore, current therapeutic options and some promising perspectives for the near future are presented.

1. Introduction

Asthma is characterized by chronic airway inflammation leading to bronchial hyperreactivity and, as a consequence, recurrent episodes of wheezing, shortness of breath, chest tightness, and cough which vary in time and intensity [1]. Asthma is a heterogeneous disease as patients vary in terms of inflammation patterns, concomitant diseases, exacerbating factors, as well as response to therapy. Eosinophilic asthma (EA) is the most common inflammatory phenotype. Chronic T2-high inflammation is characterized by overexpression of pro-inflammatory cytokines (IL-4, IL-5, IL-13) and eosinophilic infiltrations in bronchial mucosa [2]. Clinical characteristics of EA include early onset of symptoms, high prevalence of rhinitis/sinusitis, as well as a significant influence of airborne allergens or non-steroidal anti-inflammatory drugs (NSAIDs). However, patients with this phenotype usually have good response to ICS treatment [1]. Moreover, in recent years they have benefited from modern biological therapies targeting IgE, IL-5, IL-5R, or IL-4/-13R.
Contrary to EA, phenotypes related to T2-low inflammation (neutrophilic and paucigranulocytic asthma) are not well defined. Moreover, in clinical practice, no biomarkers are routinely used to diagnose them, assess severity of symptoms, or predict prognosis. In clinics, they are characterized by later onset, coexisting obesity, tobacco smoking, as well as a worse response to ICS compared to EA [3]. So far no biological drugs targeting neutrophils have been registered. As a result, patients suffering from T2-low asthma achieve control of symptoms less often and have more exacerbations compared to EA counterparts.
Considering the significant progress in the therapy of EA that followed investigating its mechanisms, it can be speculated that better understanding of non-EA would also lead to defining new drug targets and biomarkers. Consecutively, personalization of management would ameliorate its efficacy and safety.
This review discusses prevalence, mechanism, and clinical features of neutrophilic asthma. Furthermore, current therapeutic options and new perspectives for the future are presented.

2. Definition and Prevalence of Neutrophilic Asthma

2.1. Diagnosing Neutrophilic Asthma

Cross-sectional cluster analyses have identified four major asthma subgroups based on the sputum cellular patterns: eosinophilic, neutrophilic, mixed, and paucigranulocytic. Eosinophilic phenotype is the best defined one with proposed thresholds of blood eosinophilia (>0.3 × 109/L or >0.15 × 109/L); sputum eosinophilia ≥ 2% [1]. In contrast, neutrophilic asthma has not been well defined yet. Cluster analyses demonstrate that clinical features distinct from eosinophilic asthma are present in study groups characterized by a high neutrophil count in induced sputum (from >40% to >76%) with low eosinophils (<2%) [4,5,6,7].
There is no consensus on a specific cut-off point for neutrophilic asthma within BAL. However, the findings of certain studies suggest that values of neutrophil rate ≥5% may prove to be useful in the identification of a neutrophil-predominant phenotype, especially for children [8,9].
As far as neutrophil blood count is concerned, a cut-off of 5.0 × 109/L has been used in several studies [10]. Contrary to sputum or BAL neutrophilia, it is not associated with clear clinical phenotypes so it probably does not define neutrophilic asthma well.
So far there are no recommended, easily available and reliable biomarkers that could facilitate the diagnosing of NA.
In conclusion, diagnosing neutrophilic asthma is challenging. There are no tests that can be easily implemented in everyday practice. Cut-offs for induced sputum neutrophil count vary significantly which may stem from heterogeneity of the population with NA.
Detailed data regarding proposed cut-offs for diagnosing neutrophils-predominant asthma are shown in Table 1.

2.2. Estimated Prevalence of Neutrophilic Asthma

As there are no explicit criteria based on cut-offs for cell concentration in BAL or sputum, the prevalence of neutrophilic asthma (NA) mostly ranges from 16% to 28% of the adult asthma population depending on the definitions and study methods applied [7,17,18]. However, some studies demonstrate much lower rates (4–5%), while other studies found neutrophilic asthma to be the predominant phenotype (57%) [17,19,20]. In a large study of 995 patients tested repeatedly over time, 47% of patients did not have eosinophilia at any time and another 20% only sporadically suggesting a significant role of non-eosinophilic pathways in majority of the cases [21].
Among infants and children (6 months–3 years of age) with recurrent wheezing, neutrophilia was found in 65% sputum samples. Interestingly, neutrophils prevailed in infants with mild-to-moderate course of the disease while severe patients had predominantly eosinophilic inflammation [22]. Among 2800 children with poorly controlled asthma, 16% had neutrophilic phenotype confirmed by means of BAL [23].

2.3. Is Neutrophilic Asthma a Phenotype or a Lab Artifact?

The prevalence of neutrophilic asthma is diverse partly due to the fact that cellular patterns of inflammation vary over time. It has been demonstrated that patients with EA experience exacerbations [24]. Moreover, sputum eosinophil and neutrophil counts seem to be significantly affected by the therapy of asthma. Cowan et al. compared induced sputum of 94 patients with asthma in two time points: steroid-naïve and upon treatment. In the former, 67% of patients were eosinophilic, 31% paucigranulocytic and 2% mixed; there were no neutrophilic subjects. After the introduction of ICS, 39% were eosinophilic, 46% paucigranulocytic, 3% mixed, and 5% neutrophilic. Sputum neutrophils rate increased upon treatment from 19.3% to 27.7% [25]. In another study, neutrophils were found in bronchial biopsies in patients with typical clinical pattern of EA with atopy and elevated serum IgE [14].
Moreover, blood neutrophilia is affected by several concomitant conditions. It is well known that neutrophilia accompanies hematologic and non-hematologic malignancies, chronic inflammatory diseases (e.g., colitis ulcerosa, Crohn’s disease, hepatitis, rheumatoid arthritis, etc.), thrombosis, asplenia, and several endocrinopathies. In some cases, it is induced by therapy with beta-blockers, lithium, or systemic glucocorticosteroids, including, often forgotten by patients, joint injections of slow-release formulas [26]. It has been demonstrated that smokers have higher neutrophilia compared to non-smokers [27].
The variability of airway neutrophilia, concomitant influence of eosinophilic inflammation, and possible effects of coexisting pathologies impede the interpretation of several studies in this field. As a consequence, there is no generally accepted diagnostic criterion for neutrophilic asthma. Moreover, its very existence is sometimes questioned. Some researchers believe that this is COPD rather than asthma. Others dismiss airway neutrophilia as a marker of corticosteroid exposure or tobacco smoking without causative relation to asthma itself [28]. In our view, diagnosis should not be based solely on neutrophils level. Instead, clinical characteristics of the patient should be thoroughly analyzed.
Furthermore, in everyday practice as well as in a large body of the literature, the division into eosinophilic (i.e., mostly T2-high) and non-eosinophilic (i.e., T2-low) asthma is often used. In this approach, non-eosinophilic asthma includes neutrophilic and paucigranulocytic phenotypes. From the practical point of view, such simplification is quite useful as it allows for a determination of a major endotype in a given patient and allocation of proper biological therapy. However, it often leads to using the terms ‘non-eosinophilic’ and ‘neutrophilic’ asthma interchangeably. As a result, it is difficult to ascertain how many non-eosinophilic cases had primary paucigranulocytic phenotype and how big the proportion of predominantly neutrophilic asthma truly is in this entity. Some authors have demonstrated that paucigranulocytic asthma is twice as common as neutrophilic (38% vs. 16%) [7].

3. Clinical Characteristics of Neutrophilic Asthma

One of the earliest studies linking neutrophilic inflammation with distinct clinical features of asthma was a report from the Mayo clinic presenting cases of fatal asthma attacks. The fatalities had prevailing neutrophilic infiltrations in the airway submucosa in contrast to eosinophilic inflammation usually found in patients who had not had acute events [29].

3.1. Asthma Onset

Cluster analyses have demonstrated that groups with predominantly neutrophilic asthma are relatively older and most of the participants were diagnosed with asthma in adulthood [5,17]. In the study by Moore, half of the group was diagnosed with asthma at the age > 12 years [6]. Another study demonstrates that the median age at diagnosis was 56 years [4]. In most of the studies, women prevailed over men [4].
Studies on asthma in elderly have shown that the proportion of patients with sputum neutrophilia is increasing with age [30]. In contrast, an allergic component is also found to be less frequent with advanced age [30]. On the other hand, neutrophilic profiles are often found in patients reporting allergen-induced exacerbations over time or individuals with elevated total and specific IgE [11].

3.2. Clinical Course of the Disease

Cluster analyses have demonstrated that neutrophilic inflammation is associated with poorer control of asthma with higher incidence of daily symptoms and awakenings as well as higher usage of rescue treatment [17,31,32]. Unsurprisingly, neutrophilic asthma is related to lower disease-related quality of life [17].
Sputum and bronchial neutrophils as well as the enhanced expression of Th17-related cytokines are strongly associated with the severity of asthma [33,34]. Moore et al. have demonstrated that 38% of NA patients have severe asthma [6]. In the Chinese population, although neutrophilic asthma was found to be uncommon (4.3% of participants), 60% of its cases were assessed to be severe [17].
Population with neutrophilic asthma is also characterized by a higher exacerbation rate. In a large Dannish study analyzing the course of asthma over 8 years, blood neutrophilia > 4.85 G/l doubled the risk of moderate exacerbations of asthma, defined as those requiring OCS intake [35]. Higher exacerbation rate was also found to be related to the expression of cytokines associated with neutrophilic inflammation [33]. Bullone et al. have shown that 17% of the NA group had more than one exacerbation per year compared to 9% in non-neutrophilic cases [11]. In another study, the NA group had the highest rate of emergency department visits as well as hospitalizations due to asthma attacks [6]. In contrast, in a study by Welsh et al. based on repeated induced sputum assessment, persistent neutrophilia was not associated with number of exacerbations or shorter time to first exacerbation [36]. Inconsistencies between the studies may result from variability of airway neutrophilia and heterogeneity of the population with NA.
Neutrophilic phenotype is characterized by a poorer response to first-line asthma treatment—ICS [4]. Some authors claim that neutrophilic inflammation is responsible for about 50% of corticosteroid resistant/insensitive asthma cases [37]. Usually, to achieve comparable clinical outcomes, higher dose of ICS should be used than in EA. In cluster analyses, NA patients are treated, on average, with higher doses of ICS, and up to 40% require high doses of ICS [6]. Some studies show that the dose of ICS is about twice as high compared to that for other phenotypes [11].
The severity of asthma and relatively frequent exacerbations lead to higher use of systemic steroids either continuously or upon exacerbations [6]. The prevention of exacerbations with biological drugs is not as available as that for the more common EA. This is due to the fact that the majority of currently used biological drugs are indicated for the treatment of T2-high inflammation, so patients with the neutrophilic phenotype do not benefit from them [1]. Again, elevated blood neutrophil count was associated with uncontrolled asthma in patients receiving biologics [32].

3.3. Pulmonary Function

Patients with neutrophilic asthma have significantly lower FEV1% and FEV1/VC and smaller reversibility after SABA compared to the eosinophilic group [11]. Substantial proportion of the group has fixed obstruction. Choi et el. found that NA patients constitute majority of the fixed obstruction group with remodeling [38]. A 10-fold increase in neutrophil count was associated with a reduction in postbronchodilator FEV1 of 92 mL [39]. Some authors do not confirm that, while others indicate that the neutrophilic group has more severe obstruction compared to the paucigranulocytic, but not the eosinophilic group [4,6,7].
Some authors have demonstrated functional markers of air trapping (decreased FVC%, increased FRC%). The highest numerical value of RV was observed in severe neutrophilic asthma, and this group had the highest proportion of air trappers [11].

3.4. Resistance to ICS

Neutrophilic asthma is associated with treatment with higher doses of ICS, more frequent oral steroid use, and more frequent hospital admissions (65% do 28%) [6]. It is estimated that 50% corticosteroid-resistant/insensitive asthma cases are characterized by neutrophilic inflammation [37]. The limited response to such treatment can be explained by several reasons. Firstly, CS has been shown to inhibit the inflammatory pathway characterized by the predominance of T2 cytokines such as IL-4 and IL-5. The consequence of this pathway being inhibited is a reduction in eosinophil production and survival, with a resultant alleviation of inflammation in the airways [40,41]. While ICS promote apoptosis of eosinophils, they have been demonstrated to inhibit apoptosis of neutrophils [42,43]. The cytokines IL-8, IL-17, and TNF-α, which are present in Th1/Th17 inflammation, contribute to neutrophil recruitment and activation in mechanisms not related to ICS. IL-17 induces the expression and activity of transcription factor CEBPB which, in turn, enhances the production of lipocalin-2 (LCN-2) and serum amyloid A (SAA), contributing to resistance to ICS [44,45].

3.5. Concomitant Diseases

Patients with neutrophilic asthma are characterized by elevated levels of CRP and fibrinogen, suggesting that systemic inflammation may be hallmark of this phenotype [7,46]. Inflammation may also stem from concomitant diseases. Cluster analyses have demonstrated that NA is often accompanied by obesity and GERD. A substantial proportion of patients are current smokers or ex-smokers [47].

3.5.1. Obesity

Obesity is a chronic disease of complex etiology, defined as abnormal or excessive fat accumulation. Worldwide, 500 million people are obese; this is equivalent to 11% of adult men and 15% of women [48]. Patients with asthma are even more affected, having a prevalence exceeding 20% [49]. Obesity elevates the risk of asthma and obese patients tend to have uncontrolled symptoms, more frequent and severe exacerbations, as well as a worse response to ICS.
In obesity, adipose tissue propagates chronic inflammation both locally and systemically [50]. Fat cells produce several adipokines such as leptin, resistin, lipocain 2, IL-6, TNF, and IL-1β, which recruit and activate cells of the innate immune system and induce systemic inflammation [51,52,53,54]. Leptin has pro-inflammatory properties, improves neutrophils’ function, and increases their recruitment into organs, including the lungs [50,55,56]. This explains correlations that have been shown between blood neutrophil counts and BMI [57,58]. In the female asthmatic population, 1.0% increase in neutrophil blood counts was associated with one unit increase in BMI [59].
In cluster analyses, obesity-related asthma has shown predominantly non-Th2 endotype, with later onset and severe symptoms, more prevalent in females. In a study assessing the severity of adult-onset asthma, obese patients made up 16% of the uncontrolled cases and only 8% of the controlled asthma [60]. In a cohort of 233 subjects with asthma, the highest neutrophil counts were observed in a group with severe obesity. A total of 83.3% of obese subjects had blood neutrophils ≥ 4.0 × 109/L compared to 31.5% in the normal weight subgroup. Compared to the reference group (BMI < 30 kg/m2, blood neutrophils < 5 × 109/L), BMI ≥ 30 kg/m2 with blood neutrophils ≥ 5 × 109/L was associated with uncontrolled asthma [61].
Weight loss interventions have been demonstrated to be effective in improving both asthma control and lung function [62].

3.5.2. GERD

GERD is a very common comorbidity in asthma. Most of the studies indicate that 25–35% of patients are affected with a much higher incidence in difficult-to-treat and severe asthma [63,64,65]. Simpson et al. have demonstrated that GERD is more frequent in neutrophilic than in eosinophilic asthma [66]. Relations between asthma and GERD are complex as they may interact in a vicious cycle.
First, GERD affects the respiratory system in mechanisms of micro-aspirations (‘reflux theory’) as well as bronchoconstriction related to common vagal innervation of bronchi and esophagus (‘reflex theory’). In healthy individuals, GERD is one of the most common causes of chronic cough and vocal cord dysfunction manifested by hoarseness [67]. GERD-related cough is related to elevated sputum neutrophil counts and overexpression of pro-inflammatory cytokines including IL-8 [35]. These effects are reversible upon treatment with proton pump inhibitors (PPI) [68].
On the other hand, asthma can aggravate GERD by lung hyperinflation and raising the pressure gradient between the chest and the abdomen. Moreover, bronchodilators used in the therapy of asthma lead to the relaxation of lower esophageal sphincter [67]. These effects seem to be particularly important in obese patients with asthma. The triangle of asthma, GERD, and obesity correlates with neutrophilic inflammation in lower airways [69].

3.5.3. Cigarette Smoking

It is estimated that 20–25% of the asthma population have some smoking habits [47]. It has been well documented that exposure to smoke substantially increases risk of asthma in children. Smoking patients with asthma have a higher percentage of neutrophils in induced sputum and a poorer response to treatment with inhaled corticosteroids compared to non-smoking counterparts [7,70]. Combustible cigarette smoking increases levels of several pro-inflammatory cytokines including IL-17a, IL-6, and IL-8. Concomitant exposure of human epithelial cells to IL-17a, smoke, and airborne allergens leads to secretion of IL-6 and IL-8 associated with neutrophilic inflammation [71]. This phenomenon has also been confirmed in an animal model. Response to ovalbumin in sensitized rats was increased by cigarette smoke exposure and led to neutrophilic infiltrations of the pulmonary tissue [72].
Smoking patients with asthma and low FEV1 have also been shown to have an increased number of IRF5+ macrophages (associated with Th1 responses) and a reduced number of IL10+ macrophages (with anti-inflammatory effects). It has been suggested that this may be the result of the combined effects of smoking and chronic asthma [73].
Comparison of the most clinical characteristics of EA and NA is presented in Table 2.
NA shares some clinical features with paucigranulocytic asthma. Analysis by Deng et al. demonstrated three distinct clinical clusters of paucigranulocytic asthma. One of them was smoking-associated and characterized by older age, later asthma onset, and increased risk of exacerbations. Another two had some features of EA with rhinitis and mild symptoms [74]. Due to several clinical similarities, non-eosinophilic asthma phenotypes are often perceived as one entity. Some authors propose using more practical, ‘clinically-centered’ phenotypes based on concomitant disease (‘obesity-related asthma’ or ‘smokers’ asthma’) instead of cellular assessment [75].
However, according to most authors, paucigranylocytic asthma represents “benign” phenotype with good response to treatment [76]. Ntontsi et al. demonstrated that only 15% of patients with paucigranulocytic asthma have uncontrolled symptoms. Pulmonary function has been found to be significantly better compared to that of patients with eosinophilic or neutrophilic asthma [19].

4. Neutrophils in Asthma

Neutrophils are the most abundant class of white blood cells in human circulation and are a crucial component of the immune system, particularly in antibacterial and antifungal responses [77,78]. Their best-known functions include phagocytosis, degranulation, and release of neutrophil extracellular traps (NETosis) [79,80,81]. Neutrophils secrete several pro-inflammatory cytokines (IL-1α, IL-1β, IL-6, TNF) and chemokines responsible for the recruitment of other neutrophils (CXCL-1, -2, -8) or T cells (CXCL-9, -10) [82,83]. On the other hand, they also regulate tissue repair and remodeling. Recent studies have shown heterogeneity of neutrophils and their functional compartmentalization [84,85,86]. Moreover, neutrophils populations in tissues change their phenotype (N1 or N2-predominant) depending on tissue microenvironment with either prevailing pro-inflammatory activity or promoting of regeneration and growth. Such functional plasticity of neutrophils enables their adaptation to the characteristics of tissue and the current situation.
In the course of asthma, neutrophils migrate into the airways in the early phases of the disease and subsequently release various inflammatory mediators. Their recruitment may be initiated by viral or bacterial infections, air pollution, tobacco smoke, or other irritants. Epithelial injury triggers the activation of epithelial cells and tissue macrophages to produce cytokines and chemokines including IL-1α, Il-1β, IL-6, IL-8, IL-17, and IL-23. IL-8 is believed to be the most potent neutrophil chemoattractant in the lungs, promoting gradient-dependent migration, i.e., from lower to higher concentration of the cytokine [87]. Neutrophils respond to IL-8 signaling via CXCR1/CXCR2 receptors on the cell surface. The binding leads to directed cell movement across the endothelium and epithelium into the airway lumen. Furthermore, the capacity of neutrophils to secrete IL-8 has been demonstrated. Thus, initial stimulation by IL-8 is consecutively enhanced by a feedback loop that promotes neutrophil recruitment to the airways by neutrophils [88].

4.1. Direct Effects of Neutrophils on Bronchial Mucosa

Degranulation of neutrophils is one of the main mechanisms stimulating inflammatory bronchial response in patients with asthma. Contents of neutrophilic granules such as NE, MPO, cathepsin G, and proteinase 3 damage the epithelial barrier, destroy cells, and induce the secretion of pro-inflammatory cytokines by epithelial cells (Figure 1) [89,90,91]. MPO, a cationic enzyme located in primary azurophilic granules, has been shown to catalyze ROS production and to be involved in NET formation and stabilization [92,93,94].
Increased production of reactive oxygen species (ROS) in neutrophilic asthma contributes to chronic oxidative stress, which in turn promotes lung tissue remodeling and impairs bronchial epithelial regeneration [95,96,97,98]. The excess of ROS leads to a number of detrimental effects, including lipid peroxidation of cell membranes, denaturation of proteins and enzymes, DNA damage, and activation of inflammatory factors such as NF-κB [99,100,101,102,103].
NETs formed by neutrophils in the context of asthma disrupt the tight junctions of the bronchial epithelium, leading to the efflux of intracellular components [104,105]. NETs can trigger inflammatory responses by damaging airway epithelial cells, inducing IL-8 production and activating eosinophils to release their granules [106]. The protein HMGB1, a component of NET, has been demonstrated to enhance the expression and secretion of pro-inflammatory cytokines, alarmins, and growth factors (e.g., TNF-α, TSLP, MMP-9, and VEGF). These mediators play a pivotal role in the course of neutrophilic asthma [107].

4.2. Neutrophils-Associated Cytokines in Asthma

Neutrophils also secrete cytokines and chemokines, such as IL-1β, TNF-α, CXCL8 (IL-8), and GM-CSF, which further enhance the inflammatory response leading to chronic inflammation, airway hyperresponsiveness and, in a longer perspective, airway remodeling (Figure 2) [89,108,109,110,111].

4.2.1. TSLP

Thymic stromal lymphopoietin (TSLP) is an alarmin produced by epithelial cells in response to irritants (e.g., allergens, viruses, bacteria, pollutants, and smoke) [110]. Compared to healthy controls, TSLP expression is increased in the airways of patients with asthma [111,112]. It mainly drives the Th2 type of inflammatory response, by inducing dendritic cells to express OX40 ligand (OX40L) which is required for triggering naive CD4(+) T cells to produce IL-4, IL-5, and IL-13 [113,114]. In addition, CD8+ cytotoxic T lymphocytes also produce IL-5, IL-13, and IFN-γ as a result of additional stimulation by CD40L [115]. Furthermore, TSLP also contributes to the development of non-eosinophilic inflammation. It is suggested that human TSLP and TLR3 ligands may promote Th17 cell differentiation through activation of dendritic cells [116]. It has also been documented that bronchial epithelial cells increase neutrophil recruitment through IL-8 and GM-CSF production [117].

4.2.2. IL-17

Interleukin 17, a key cytokine in neutrophilic asthma, is produced by Th17 cells. It elevates expression of cytokines and factors which propagate neutrophil recruitment and activation into the airways via IL-6, IL-8, and TNF- α [118]. Furthermore, it induces bronchial remodeling by stimulating fibroblasts to produce collagen and growth factors, such as transforming growth factor-beta (TGF-β). IL-17 is overexpressed in bronchial mucosa of patients with neutrophilic asthma, particularly in cases with more pronounced neutrophilic inflammation and frequent exacerbations [11]. In another study, patients with severe asthma present increased levels of IL-17 in bronchial biopsy as well as in sputum [119,120]. A correlation has been demonstrated between IL-17 mRNA levels and IL-8 mRNA levels, as well as neutrophil counts, indicating an association between IL-17 and neutrophilic inflammation [120]. IL-17 may be associated with glucocorticoid resistance as it induces glucocorticoid receptor (GR) β in epithelial cells in asthma [121].

4.2.3. IL-8

Interleukin 8 (CXCL8/IL-8), secreted by bronchial epithelial cells in response to the pro-inflammatory cytokines IL-17, TNF-α, and IFN-γ, functions as a chemoattractant of neutrophil accumulation, promoting their survival in the airways [122,123]. Several studies have confirmed that IL-8 levels are significantly higher in patients with asthma than in healthy subjects [124,125,126]. In asthma, levels of IL-8 are correlated with the percentage of neutrophils in BAL, which points to the role of IL-8 in the recruitment and activation of neutrophils in the airways [127]. The highest IL-8 levels have been found to be associated with the non-eosinophilic asthma phenotype [16]. Furthermore, a significant increase in IL-8 levels was observed in induced sputum children during an asthma exacerbation, in comparison to the stable period of the disease. This finding is in line with what is known about the role of neutrophil recruitment during airway infections [128].

4.2.4. IL-1β

Interleukin-1β (IL-1β) is a potent pro-inflammatory cytokine which is produced and secreted by a variety of cell types, particularly by innate immune system cells, such as monocytes and macrophages. It is produced as an inactive precursor, termed pro-IL-1β, in response to PAMPs [129]. Consecutively, pro-IL-1β is cleaved by the pro-inflammatory protease caspase-1 [130]. Chlamydia spp. and Haemophilus spp. infections have been shown to induce responses with increased IL-1β, NLRP3, and caspase-1, that drive steroid-resistant neutrophilic inflammation and airway hyperresponsiveness.
It has been demonstrated that IL-1β promotes Th17 differentiation and IL-17 production. Therefore, an increased level of IL-1β is a marker of more severe asthma, as it induces steroid-resistant neutrophilic inflammation and airway hyperreactivity [131,132]. This finding was also confirmed in a mouse model of asthma. Neutrophilic airway inflammation, disease severity, and steroid resistance have been shown to correlate with NLRP3 and IL-1β expression. Furthermore, treatment with anti-IL-1β antibodies resulted in both inhibition of the IL-1β response and remission of clinical characteristics of steroid-resistant asthma, while IL-1β administration led to the restoration of these characteristics [131].

4.2.5. IL-33

IL-33 is passively released as an ‘alarmin’ upon tissue injury or necrosis, predominantly secreted by structural cells at barrier tissues [133]. Although it is a cytokine that mainly drives T2-high inflammation through the activation of immune cells such as mast cells, eosinophils, and ILC2, there are also studies demonstrating its role in non-eosinophilic asthma [134]. IL-33 acts through the ST2 receptor, which is present in conventional and regulatory T cells, ILC2, M2 macrophages, mast cells, eosinophils, basophils, neutrophils, NK cells, and iNKT cells [135]. Following IL-33 stimulation, peripheral neutrophils show increased MPO expression and phosphorylation of ERK and MAPK [136]. It has been reported that neutrophil inflammatory proteases produce a shorter form of IL-33, which has 10–30 times more potent effects than full-length IL-33 in activating target cells [137,138]. This suggests that IL-33 can be more efficient in neutrophilic inflammation.
It has been found that levels of IL-33 and ST2 receptor were elevated in groups characterized by uncontrolled asthma symptoms and low eosinophilia. In a mouse model, blockade of the IL-33/ST2 axis resulted in reduction in neutrophilic inflammation, IL-17A, MPO, and NET marker (SA100A9) and also reduced the severity of symptoms [136]. In another study on a mouse model of rhinovirus-induced chronic asthma, it was observed that early neutrophilic inflammation with NET formation was found to be IL-33-dependent. In nasal samples from rhinovirus-infected patients with asthma, IL-33 levels correlated with neutrophil elastase and dsDNA, which was not found in healthy controls [139].

4.3. Smooth Muscles in Neutrophilic Asthma

In asthma, pro-inflammatory environment affects the structure and functions of bronchial smooth muscles (BSM) cells. They are major effectors of exaggerated airway narrowing and hyperresponsiveness. Their contraction occurs in response to several stimuli, e.g., allergens, pollution, cold air, or physical exercise. Bronchial hyperresponsiveness has been described to be controlled by T2-related cytokines such as IL-5 and IL-13. In neutrophilic asthma, pro-inflammatory cytokines IL-1β, IL-17, and IL-23 may also further increase this process [140]. Moreover, BSM stimulated with them secrete several chemokines (CXCL1, CXCL10, CCL10), recruiting immune cells and prolonging inflammation.
The development of long-standing asthma is characterized by airway remodeling, which encompasses the hypertrophy and proliferation of smooth muscle cells, culminating in the thickening of the bronchial walls [141]. This process is promoted by neutrophil-derived matrix metalloprotease-9 (MMP-9) and elastase [142,143]. The levels of elastase inversely correlate with pulmonary function [144]. Moreover, Th17-related cytokines such as IL-17A, IL-17F, and IL-22 have been demonstrated to possess the capacity to induce BSM cell migration in a receptor-selective manner. Furthermore, the presence of Th17 cytokine receptors (IL-17RA, IL-17RC, and IL-22R1) in these cells has been confirmed. It is noteworthy that IL-17A and IL-17F exert their effects through signaling pathways other than the IL-22 pathway, suggesting the existence of more than one mechanism in the pathogenesis of remodeling [145]. Moreover, IL-23 prolongs the expression of the listed Th17 cytokines that induce tissue pathology and chronic inflammatory diseases, giving prominence to the IL-23/IL-17 immune axis [146].
One of the most significant contributors to airway remodeling is transforming growth factor (TGF-β). In bronchi, it is secreted by several types of cells, including neutrophils. TGF-β is profibrotic cytokine [147]. Although neutrophils constitutively express TGF-β, those derived from the asthma population secrete higher amounts of this cytokine [148].

5. Management of Neutrophilic Asthma—Present and Future

Current asthma treatment focuses on a personalized approach, integrating pharmacological and non-pharmacological strategies to optimize disease control. The latter include lifestyle modifications, allergen avoidance, and smoking cessation in order to improve long-term outcomes. According to the GINA 2025 guidelines, ICS remain the cornerstone of therapy, effectively reducing airway inflammation and preventing exacerbations [1]. For patients with moderate-to-severe asthma, combination therapies with addition of LABA, LAMA, and LTRA are recommended. The evolving understanding of asthma phenotypes allows for more tailored treatments, ensuring better disease management and quality of life for patients. Thus, in cases of severe treatment-resistant asthma, biologic therapies targeting key inflammatory pathways (e.g., IL-4, IL-5) are widely used. They have shown significant benefits in reducing exacerbation rates and steroid dependency [1]. However, the vast majority of biologicals used in today’s practice target only T2-high inflammation (anti-IL-5, anti-IL-5R, anti-IgE, etc.). The only biological drug registered for use in asthma irrespective of the inflammatory pathway involved is tezepelumab. In neutrophilic asthma it has been demonstrated that, by binding TSLP, tezepelumab inhibits the TH17/neutrophil cascade at the beginning of the immune pathway, limiting inflammation and preventing the airway remodeling typical for steroid-resistant neutrophilic asthma [149].
Increasing knowledge about the biology of neutrophils and their role in asthma results in new potential therapies that could ameliorate control of neutrophilic asthma in the near future. Below, we present some therapies targeting neutrophilic inflammation that have been investigated recently.

5.1. Anti-IL-17: Brodalumab

Brodalumab (AMG 827) is a human anti-IL-17RA immunoglobulin G2 (IgG2) monoclonal antibody which binds with high affinity to human IL-17RA. This blocks the biological activity of IL-17A, IL-17F, IL-17A/F heterodimer, and 17E (IL-25).
In the randomized, double-blind, placebo-controlled trial, 302 patients with inadequately controlled moderate-to-severe asthma were treated with brodalumab at doses of 140 mg, 210 mg, 280 mg, or placebo. Significant improvement in ACQ was only seen in the subgroup characterized by high obstruction reversibility (≥20% improvement in FEV1 after β- agonists), and treated with a dose of 210 mg. There was no effect in the 280 mg group [150]. Another study (NCT 01902290) was terminated earlier due to the lack of efficacy observed following interim analysis. Therefore, brodalumab was deemed ineffective, and it was not considered to have a therapeutic role in the treatment of asthma, including asthma with a neutrophilic phenotype [151].

5.2. Anti- IL-1β

The activity of IL-1β, which is released by alveolar macrophages, is significantly higher in patients with asthma than in healthy subjects [152]. IL-1ra is a natural anti-inflammatory cytokine that competes with IL-1β for binding to the type I IL-1 receptor [153]. It has been shown that IL-1ra inhibits neutrophilic inflammation induced by both LPS and IL-1 [154]. Anakinra is a recombinant form of human IL-1ra, the use of which in patients with asthma has been studied by Hernandez et al. [153]. Patients who received two subcutaneous doses of anakinra (1 mg/kg, maximum dose 100 mg) were subjected to an inhaled LPS challenge, and the number of neutrophils in their sputum was assessed. Compared with the placebo group, those receiving anakinra experienced a significant reduction in airway neutrophilia. Furthermore, LPS-induced IL-1β, IL-6, and IL-8 concentrations were reduced by 39%, 83%, and 150%, respectively, during the anakinra treatment period compared with placebo [153]. These data may be relevant for the treatment of non-T2 asthma.
There were several studies planned to evaluate anakinra as a rescue treatment for acute allergic reactions (NCT 03513471, NCT 04035109). However, they were abandoned during COVID-19 pandemic. An ongoing trial (NCT 04278404) aims at assessing the safety and pharmacokinetics of anakinra in children.

5.3. Targeting IL-8 (CXCL-8)

IL-8 is one of the main chemotactic factors induced by Th17, and it works via the chemokine receptor 2 (CXCR2), along with the leukotriene B4 [155]. CXCR2 is a key regulator of neutrophil migration and, therefore, has been the most studied target in preclinical studies.
In a randomized, double-blinded study, SCH 527123 resulted in a mean reduction of 36% in sputum neutrophil count in 22 patients with severe asthma and sputum total cell count < 10 × 106/g and neutrophils > 40% compared with a 6.7% increase in the placebo group [156]. The mean absolute blood neutrophil count initially decreased but returned to normal at week 5. There were fewer mild exacerbations of asthma (1.3 versus 2.25, p = 0.05) and an improvement in the ACQ score (by an average of 0.42 points). No statistically significant changes in FEV1 were observed. The drug was deemed safe for use in patients with severe asthma [157].
Another CXCR2 antagonist that was tested was AZD8309. Twenty healthy subjects were given a placebo or AZD8309 (300 mg) orally twice daily for three days, after which they were given inhaled LPS and their induced sputum was collected. AZD8309 was found to result in an average 77% reduction in sputum total cell count and 79% reduction in neutrophils compared with placebo after the LPS challenge [156].
These studies led to a clinical trial of AZD5069, which is another selective CXCR2 receptor antagonist. Of the four groups of adult patients with uncontrolled asthma, three received AZD5069 at different doses, while one was given a placebo. While treatment with this agent was well tolerated, none of the AZD5069 doses reduced the incidence of severe exacerbations compared with the placebo. According to the authors, these results call into question the role of CXCR2-mediated neutrophil recruitment in exacerbations of severe refractory asthma [158].

5.4. Targeting IL-6

Several human and animal model studies have demonstrated pleiotropic activity of IL-6 in terms of airway inflammation. Overexpression of IL-6 was associated with a lower FEV1 in patients with mixed eosinophilic–neutrophilic bronchitis. On the contrary, the loss of IL-6 signaling abolished the increase in eosinophil- and neutrophil-recruiting cytokines and chemokines, as well as allergen-induced airway inflammation in mice. These findings imply that asthma exacerbations, particularly those involving eosinophil–neutrophil inflammation, may respond to therapies targeting the IL-6 pathway [159].
Tocilizumab is a human monoclonal antibody that blocks IL-6 signaling. A small proof-of-concept study investigated whether a single dose of tocilizumab could prevent bronchospasm induced by inhaled allergens in patients with mild asthma but found no evidence to support this. However, effective blockade of IL-6 was demonstrated by a decrease in levels of CRP, IL-6, and soluble IL-6R. The presence of a biological effect but a lack of clinical effects means that this type of therapy requires further and more thorough testing on different patient groups [160]. One such study is being conducted now in the USA and Canada (NCT 04278404) to assess the safety and pharmacokinetics of tocilizumab in pediatric asthma.
Another option of IL-6 targeting treatment includes clazakizumab being tested in adults and in teenagers with severe asthma (phase 2 PRECISE study) and FB704A being tested in adults with severe asthma (phase 2 study NCT 05018299). Results of these trials have not been published so far [161].

5.5. Biologics Targeting the IL-33 Pathway

Astegolimab, a human IgG2 antibody, selectively inhibits the IL-33 receptor, ST2. In a randomized clinical trial, astegolimab reduced annual asthma exacerbation rate in a broad population of patients, including those with eosinophil-low phenotype, with inadequately controlled, severe asthma [162]. The drug remains a promising treatment option, but no phase III trial has yet been registered, despite the publication of a phase IIb study in 2021.
Itepekimab, an IgG4P monoclonal antibody targeting IL-33, has been shown to improve asthma control and lung function when administered subcutaneously at a dose of 300 mg every two weeks [163]. Current clinical trials are mainly conducted for COPD (AERIFY-1 and AERIFY-2) [164]. Clinical trials in patients with atopic dermatitis have been terminated due to the drug’s insufficient efficacy, as determined by an unblinded evaluation of proof-of-concept data [165].
Torozakimab, another anti-IL-33 antibody, was assessed in FRONTIER-3 study. The primary endpoint was changed from baseline to week 16 in pre-bronchodilator FEV1 in the group of patients with moderate-to-severe asthma. This effect was not seen in a study group as a whole. However, it was confirmed in a subgroup of frequent exacerbators (at least two episodes in the previous 12 months) [166].
To conclude, there are some emerging therapies that can ameliorate prognosis in patients with severe NA in the near future. Surprisingly, the most promising options are drugs with targets that are not specific for neutrophilic inflammation (e.g., anti-TSLP). On the contrary, drugs targeting cytokines strongly associated with neutrophils’ function (IL-1, IL-17) have not been proven highly effective. This raises concerns about the inclusion criteria applied in clinical trials. Effectively, in most of them, researchers recruited a general asthmatic population characterized by moderate-to-severe symptoms, high exacerbation rates, and poor response to ICS irrespective of inflammatory phenotype. Thus, poor responses acquired in several studies should not discourage the conducting of clinical trials in well-defined cohorts with confirmed NA.

6. Unmet Goals and Directions of Future Studies

Despite the better understanding of neutrophilic inflammation regulation, there are still important unmet needs in the field of neutrophilic asthma.
First, in terms of diagnosis of NA, sputum or blood neutrophil counts do not seem sufficient. There is a need for reliable biomarkers characterized by both good performance (sensitivity, specificity) and wide availability. The ideal biomarker of NA should correlate with the intensity of neutrophilic inflammation so that it can facilitate the diagnosis of NA and objective monitoring of the clinical course of the disease. Potential candidates include cytokines related to function of neutrophils (IL-1, IL-17); chemokines (CCL-4, CXCL-8); substances secreted by neutrophils (MMP-9, MPO); chitinase-like protein YKL-40; serum amyloid A [167,168,169,170,171]. Expression of the aforementioned biomarkers is significantly higher in NA compared to other phenotypes. However, their correlation with clinical outcomes, predictive value in relation to exacerbations, or response to biologicals remain unclear.
Another aspect that requires further investigation is the clinical relevance of neutrophilia in patients with asthma. As neutrophilia is not specific and may stem from several acute conditions (infections), comorbidities (obesity, endocrinopathies, etc.), therapies (systemic steroids), or lifestyle (e.g., smoking), it should be interpreted cautiously. In the future, we will need longitudinal studies on the stability of neutrophilic inflammation in asthma and relations between neutrophilia, asthma, ICS therapy, and comorbidities. In our view, the proper defining of neutrophilic phenotype and phenotyping of patients in daily practice will only be possible if we better understand the divisions between ‘primary neutrophilic asthma’ and ‘asthma with concomitant neutrophilic pathology’.
Finally, better understanding of neutrophilic inflammation in asthma may result in the development of tailored therapies. Despite finding potential targets important for neutrophilic inflammation, so far no neutrophil-specific drug has been found to be highly effective. As a matter of fact, we lacked clinical trials with clearly defined subgroups with NA. The poor results of the studies mentioned in this paper may stem from the enrolment of diverse populations with severe asthma instead of patients with confirmed NA who would benefit the most from neutrophils-targeted interventions.

7. Conclusions

Neutrophilic asthma is related to higher exacerbation rate, lower level of symptoms control, and poorer response to steroids compared to eosinophilic phenotype. It is further complicated by comorbidities such as obesity or GERD that may contribute to the development and severe course of the disease. The most important directions for future studies include the formulation of clear and applicable diagnostic criteria, choosing biomarkers that facilitate diagnosis and monitoring, and development of drugs targeting neutrophilic inflammation.

Author Contributions

The initial draft of the manuscript was written by I.I. and K.S. The critical review and editing of the manuscript was done by K.S., M.C. and M.N. All authors have read and agreed to the published version of the manuscript.

Funding

Publication was funded by Medical University of Gdańsk (programme ‘Excellence Initiative–Research University’; no. 61-00206).

Data Availability Statement

Data availability is not applicable to this article.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

BALbrochoalveolar lavage
BMIbody mass index
BSMbronchial smooth muscles
BTKBruton tyrosine kinase
CXCL1chemokine ligand 1
COPDchronic obstructive pulmonary disease
EAeosinophilic asthma
FEV1forced expiratory volume in 1 s
FVCforced vital capacity
GERDgastroesophageal reflux disease
ICSinhaled corticosteroids
IFNinterferon
ILinterleukin
ILCinnate lymphoid cell
LABAlong-acting beta-agonists
LAMAlong-acting muscarinic antagonists
LCN-2lipocalin 2
LTB4leukotriene B4
LTRAleukotriene receptor antagonists
MPOmyeloperoxidase
NAneutrophilic asthma
NEneutrophil elastase
NETneutrophil extracellular traps
NETosisformation of neutrophil extracellular traps
NSAIDsnon-steroidal anti-inflammatory drugs
PDEphosphodiesterase
PDGFRplatelet-derived growth factor receptor
PRRpattern recognition receptors
ROSreactive oxygen species
SAAserum amyloid A
SABAshort-acting beta-agonists
TGFtransforming growth factor
TLRtoll-like receptors
TNFtumor necrosis factor
TSLPthymic stromal lymphopoietin
VEGFvascular endothelial growth factor

References

  1. Global Initiative for Asthma. Global Strategy for Asthma Management and Prevention. GINA 2025 Report; Global Initiative for Asthma: Fontana, WI, USA, 2025; Available online: https://ginasthma.org/2025-gina-strategy-report/ (accessed on 6 May 2025).
  2. Domingo, C.; Sicras-Mainar, A.; Sicras-Navarro, A.; Sogo, A.; Mirapeix, R.M.; Engroba, C. Prevalence, T2 Biomarkers, and Cost of Severe Asthma in the Era of Biologics: The BRAVO-1 Study. J. Investig. Allergol. Clin. Immunol. 2024, 34, 97–105. [Google Scholar] [CrossRef]
  3. Nabe, T. Steroid-Resistant Asthma and Neutrophils. Biol. Pharm. Bull. 2020, 43, 31–35. [Google Scholar] [CrossRef] [PubMed]
  4. Green, R.H.; Brightling, C.E.; Woltmann, G.; Parker, D.; Wardlaw, A.J.; Pavord, I.D. Analysis of induced sputum in adults with asthma: Identification of subgroup with isolated sputum neutrophilia and poor response to inhaled corticosteroids. Thorax 2002, 57, 875–879. [Google Scholar] [CrossRef]
  5. Simpson, J.L.; Scott, R.; Boyle, M.J.; Gibson, P.G. Inflammatory subtypes in asthma: Assessment and identification using induced sputum. Respirology 2006, 11, 54–61. [Google Scholar] [CrossRef]
  6. Moore, W.C.; Hastie, A.T.; Li, X.; Li, H.; Busse, W.W.; Jarjour, N.N.; Wenzel, S.E.; Peters, S.P.; Meyers, D.A.; Bleecker, E.R.; et al. Sputum neutrophil counts are associated with more severe asthma phenotypes using cluster analysis. J. Allergy Clin. Immunol. 2014, 133, 1557–1563.e5. [Google Scholar] [CrossRef]
  7. Demarche, S.; Schleich, F.; Henket, M.; Paulus, V.; Van Hees, T.; Louis, R. Detailed analysis of sputum and systemic inflammation in asthma phenotypes: Are paucigranulocytic asthmatics really non-inflammatory? BMC Pulm. Med. 2016, 5, 16–46. [Google Scholar] [CrossRef] [PubMed]
  8. Busse, W.W. What Are Those Neutrophils Doing in Severe Asthma Anyway? J. Allergy Clin. Immunol. Pract. 2019, 7, 526–528. [Google Scholar] [CrossRef] [PubMed]
  9. Grunwell, J.R.; Stephenson, S.T.; Tirouvanziam, R.; Brown, L.A.S.; Brown, M.R.; Fitzpatrick, A.M. Children with Neutrophil-Predominant Severe Asthma Have Proinflammatory Neutrophils with Enhanced Survival and Impaired Clearance. J. Allergy Clin. Immunol. Pract. 2019, 7, 516–525.e6. [Google Scholar] [CrossRef]
  10. Syabbalo, N. Clinical Features and Management of Neutrophilic Asthma. J. Pulm. Med. Respir. Res. 2020, 6, 036. [Google Scholar] [CrossRef]
  11. Bullone, M.; Carriero, V.; Bertolini, F.; Folino, A.; Mannelli, A.; Di Stefano, A.; Gnemmi, I.; Torchio, R.; Ricciardolo, F.L.M. Elevated serum IgE, oral corticosteroid dependence and IL-17/22 expression in highly neutrophilic asthma. Eur. Respir. J. 2019, 54, 1900068. [Google Scholar] [CrossRef]
  12. Belda, J.; Leigh, R.; Parameswaran, K.; O’Byrne, P.M.; Sears, M.R.; Hargreave, F.E. Induced sputum cell counts in healthy adults. Am. J. Respir. Crit. Care Med. 2000, 161 Pt 1, 475–478. [Google Scholar] [CrossRef]
  13. Brooks, C.R.; Gibson, P.G.; Douwes, J.; Van Dalen, C.J.; Simpson, J.L. Relationship between airway neutrophilia and ageing in asthmatics and non-asthmatics. Respirology 2013, 18, 857–865. [Google Scholar] [CrossRef]
  14. Schleich, F.N.; Manise, M.; Sele, J.; Henket, M.; Seidel, L.; Louis, R. Distribution of sputum cellular phenotype in a large asthma cohort: Predicting factors for eosinophilic vs neutrophilic inflammation. BMC Pulm. Med. 2013, 13, 11. [Google Scholar] [CrossRef]
  15. Gibson, P.G.; Simpson, J.L.; Saltos, N. Heterogeneity of airway inflammation in persistent asthma: Evidence of neutrophilic inflammation and increased sputum interleukin-8. Chest 2001, 119, 1329–1336. [Google Scholar] [CrossRef] [PubMed]
  16. Shi, B.; Li, W.; Dong, H.; Xu, M.; Hao, Y.; Gao, P. Distribution of inflammatory phenotypes among patients with asthma in Jilin Province, China: A cross-sectional study. BMC Pulm. Med. 2021, 21, 364. [Google Scholar] [CrossRef]
  17. Wang, F.; He, X.Y.; Baines, K.J.; Gunawardhana, L.P.; Simpson, J.L.; Li, F.; Gibson, P.G. Different inflammatory phenotypes in adults and children with acute asthma. Eur. Respir. J. 2011, 38, 567–574. [Google Scholar] [CrossRef]
  18. Nair, P.; Aziz-Ur-Rehman, A.; Radford, K. Therapeutic implications of ‘neutrophilic asthma’. Curr. Opin. Pulm. Med. 2015, 21, 33–38. [Google Scholar] [CrossRef] [PubMed]
  19. Ntontsi, P.; Loukides, S.; Bakakos, P.; Kostikas, K.; Papatheodorou, G.; Papathanassiou, E.; Hillas, G.; Koulouris, N.; Papiris, S.; Papaioannou, A.I. Clinical, functional and inflammatory characteristics in patients with paucigranulocytic stable asthma: Comparison with different sputum phenotypes. Allergy 2017, 72, 1761–1767. [Google Scholar] [CrossRef] [PubMed]
  20. Dasgupta, A.; Ade, V.; Dutta, J.; Dasgupta, G. Inflammatory phenotypes of severe asthma in India. Lung India 2019, 36, 267–268. [Google Scholar]
  21. McGrath, K.W.; Icitovic, N.; Boushey, H.A.; Lazarus, S.C.; Sutherland, E.R.; Chinchilli, V.M.; Fahy, J.V.; Asthma Clinical Research Network of the National Heart, Lung, and Blood Institute. A large subgroup of mild-to-moderate asthma is persistently noneosinophilic. Am. J. Respir. Crit. Care Med. 2012, 185, 612–619. [Google Scholar] [CrossRef]
  22. Guo, Y.; Zou, Y.; Zhai, J.; Li, J.; Liu, J.; Ma, C.; Jin, X.; Zhao, L. Phenotypes of the inflammatory cells in the induced sputum from young children or infants with recurrent wheezing. Pediatr. Res. 2019, 85, 489–493. [Google Scholar] [CrossRef] [PubMed]
  23. Teague, W.G.; Lawrence, M.G.; Shirley, D.T.; Garrod, A.S.; Early, S.V.; Payne, J.B.; Wisniewski, J.A.; Heymann, P.W.; Daniero, J.J.; Steinke, J.W.; et al. Lung Lavage Granulocyte Patterns and Clinical Phenotypes in Children with Severe, Therapy-Resistant Asthma. J. Allergy Clin. Immunol. Pract. 2019, 7, 1803–1812.e10. [Google Scholar] [CrossRef]
  24. Jayaram, L.; Pizzichini, M.M.; Cook, R.J.; Boulet, L.P.; Lemière, C.; Pizzichini, E.; Cartier, A.; Hussack, P.; Goldsmith, C.H.; Laviolette, M.; et al. Determining asthma treatment by monitoring sputum cell counts: Effect on exacerbations. Eur. Respir. J. 2006, 27, 483–494. [Google Scholar] [CrossRef]
  25. Cowan, D.C.; Cowan, J.O.; Palmay, R.; Williamson, A.; Taylor, D.R. Effects of steroid therapy on inflammatory cell subtypes in asthma. Thorax 2010, 65, 384–390. [Google Scholar] [CrossRef]
  26. Otieno, S.; Altaham, A.; Karri, S.; Kaweeta, F.; Lands, L.; Wier, A. CIN or not: An approach to the evaluation and management of chronic idiopathic neutrophilia. Blood Rev. 2021, 46, 100739. [Google Scholar] [CrossRef]
  27. Van der Haart, H.; Postma, D.; Timens, W.; Hacken, N. Acute effects of cigarette smoke on inflammation and oxidative stress: A review. Thorax 2004, 59, 713–721. [Google Scholar] [CrossRef]
  28. Gibson, P.G.; Foster, P.S. Neutrophilic asthma: Welcome back! Eur. Respir. J. 2019, 54, 1901846. [Google Scholar] [CrossRef]
  29. Sur, S.; Crotty, T.B.; Kephart, G.M.; Hyma, B.A.; Colby, T.V.; Reed, C.E.; Hunt, L.W.; Gleich, G.J. Sudden-onset fatal asthma. A distinct entity with few eosinophils and relatively more neutrophils in the airway submucosa? Am. Rev. Respir. Dis. 1993, 148, 713–719. [Google Scholar] [CrossRef] [PubMed]
  30. Schleich, F.; Graff, S.; Guissard, F.; Henket, M.; Paulus, V.; Louis, R. Asthma in elderly is characterized by increased sputum neutrophils, lower airway caliber variability and air trapping. Respir. Res. 2021, 22, 15. [Google Scholar] [CrossRef] [PubMed]
  31. Rönnebjerg, L.; Axelsson, M.; Kankaanranta, H.; Backman, H.; Rådinger, M.; Lundbäck, B.; Ekerljung, L. Severe Asthma in a General Population Study: Prevalence and Clinical Characteristics. J. Asthma Allergy 2021, 14, 1105–1115. [Google Scholar] [CrossRef]
  32. Yasui, H.; Oishi, K.; Nihashi, F.; Furuhashi, K.; Fujisawa, T.; Inoue, Y.; Karayama, M.; Hozumi, H.; Suzuki, Y.; Enomoto, N.; et al. Factors associated with uncontrolled severe asthma in the biologic era. Respir. Med. 2025, 236, 107881. [Google Scholar] [CrossRef]
  33. Ricciardolo, F.L.M.; Sorbello, V.; Folino, A.; Gallo, F.; Massaglia, G.M.; Favatà, G.; Conticello, S.; Vallese, D.; Gani, F.; Malerba, M.; et al. Identification of IL-17F/frequent exacerbator endotype in asthma. J. Allergy Clin. Immunol. 2017, 140, 395–406. [Google Scholar] [CrossRef] [PubMed]
  34. Jatakanon, A.; Uasuf, C.; Maziak, W.; Lim, S.; Chung, K.F.; Barnes, P.J. Neutrophilic inflammation in severe persistent asthma. Am. J. Respir. Crit. Care Med. 1999, 160 Pt 1, 1532–1539. [Google Scholar] [CrossRef] [PubMed]
  35. Vedel-Krogh, S.; Fallgaard Nielsen, S.; Lange, P.; Vestbo, J.; Nordestgaard, B.G. Association of Blood Eosinophil and Blood Neutrophil Counts with Asthma Exacerbations in the Copenhagen General Population Study. Clin. Chem. 2017, 63, 823–832. [Google Scholar] [CrossRef] [PubMed]
  36. Walsh, C.J.; Zaihra, T.; Benedetti, A.; Fugère, C.; Olivenstein, R.; Lemière, C.; Hamid, Q.; Martin, J.G. Exacerbation risk in severe asthma is stratified by inflammatory phenotype using longitudinal measures of sputum eosinophils. Clin. Exp. Allergy 2016, 46, 1291–1302. [Google Scholar] [CrossRef]
  37. Ray, A.; Kolls, J.K. Neutrophilic Inflammation in Asthma and Association with Disease Severity. Trends Immunol. 2017, 38, 942–954. [Google Scholar] [CrossRef]
  38. Choi, J.S.; Jang, A.S.; Park, J.S.; Park, S.W.; Paik, S.H.; Park, J.S.; Uh, S.T.; Kim, Y.H.; Park, C.S. Role of neutrophils in persistent airway obstruction due to refractory asthma. Respirology 2012, 17, 322–329. [Google Scholar] [CrossRef]
  39. Shaw, D.E.; Berry, M.A.; Hargadon, B.; McKenna, S.; Shelley, M.J.; Green, R.H.; Brightling, C.E.; Wardlaw, A.J.; Pavord, I.D. Association between neutrophilic airway inflammation and airflow limitation in adults with asthma. Chest 2007, 132, 1871–1875. [Google Scholar] [CrossRef]
  40. Schleimer, R.; Bochner, B. The effects of glucocorticoids on human eosinophils. J. Allergy Clin. Immunol. 1994, 94 Pt 2, 1202–1213. [Google Scholar] [CrossRef]
  41. Tao, Z.; Zhu, H.; Zhang, J.; Huang, Z.; Xiang, Z.; Hong, T. Recent advances of eosinophils and its correlated diseases. Front. Public Health 2022, 10, 954721. [Google Scholar] [CrossRef]
  42. Cox, G. Glucocorticoid treatment inhibits apoptosis in human neutrophils. Separation of survival and activation outcomes. J. Immunol. 1995, 154, 4719–4725. [Google Scholar] [CrossRef] [PubMed]
  43. Nguyen, L.; Lim, S.; Oates, T.; Chung, K.F. Increase in airway neutrophils after oral but not inhaled corticosteroid therapy in mild asthma. Respir. Med. 2005, 99, 200–207. [Google Scholar] [CrossRef]
  44. Hong, L.; Herjan, T.; Bulek, K.; Xiao, J.; Comhair, S.A.A.; Erzurum, S.C.; Li, X.; Liu, C. Mechanisms of Corticosteroid Resistance in Type 17 Asthma. J. Immunol. 2022, 209, 1860–1869. [Google Scholar] [CrossRef]
  45. Lee, J.; Hall, J.A.; Kroehling, L.; Wu, L.; Najar, T.; Nguyen, H.H.; Lin, W.Y.; Yeung, S.T.; Silva, H.M.; Li, D.; et al. Serum Amyloid A Proteins Induce Pathogenic Th17 Cells and Promote Inflammatory Disease. Cell 2020, 183, 2036–2039. [Google Scholar] [CrossRef]
  46. Wood, L.G.; Baines, K.J.; Fu, J.; Scott, H.A.; Gibson, P.G. The neutrophilic inflammatory phenotype is associated with systemic inflammation in asthma. Chest 2012, 142, 86–93. [Google Scholar] [CrossRef]
  47. Thomson, N.C.; Polosa, R.; Sin, D.D. Cigarette Smoking and Asthma. J. Allergy Clin. Immunol. Pract. 2022, 10, 2783–2797. [Google Scholar] [CrossRef] [PubMed]
  48. World Health Organization. Obesity and Overweight. Fact Sheet; World Health Organization: Geneva, Switzerland, 2025; Available online: http://www.who.int/mediacentre/factsheets/fs311/en/ (accessed on 1 March 2024).
  49. Nathell, L.; Jensen, I.; Larsson, K. High prevalence of obesity in asthmatic patients on sick leave. Respir. Med. 2002, 96, 642–650. [Google Scholar] [CrossRef]
  50. Uribe-Querol, E.; Rosales, C. Neutrophils Actively Contribute to Obesity-Associated Inflammation and Pathological Complications. Cells 2022, 11, 1883. [Google Scholar] [CrossRef]
  51. Loffreda, S.; Yang, S.Q.; Lin, H.Z.; Karp, C.L.; Brengman, M.L.; Wang, D.J.; Klein, A.S.; Bulkley, G.B.; Bao, C.; Noble, P.W.; et al. Leptin regulates proinflammatory immune responses. FASEB J. 1998, 12, 57–65. [Google Scholar] [CrossRef] [PubMed]
  52. Komakula, S.; Khatri, S.; Mermis, J.; Savill, S.; Haque, S.; Rojas, M.; Brown, L.; Teague, G.W.; Holguin, F. Body mass index is associated with reduced exhaled nitric oxide and higher exhaled 8-isoprostanes in asthmatics. Respir. Res. 2007, 8, 32. [Google Scholar] [CrossRef]
  53. Miethe, S.; Guarino, M.; Alhamdan, F.; Simon, H.U.; Renz, H.; Dufour, J.F.; Potaczek, D.P.; Garn, H. Effects of obesity on asthma: Immunometabolic links. Pol. Arch. Intern. Med. 2018, 128, 469–477. [Google Scholar] [CrossRef] [PubMed]
  54. Kawai, T.; Autieri, M.V.; Scalia, R. Adipose tissue inflammation and metabolic dysfunction in obesity. Am. J. Physiol. Cell Physiol. 2021, 320, C375–C391. [Google Scholar] [CrossRef] [PubMed]
  55. Bouloumie, A.; Marumo, T.; Lafontan, M.; Busse, R. Leptin induces oxidative stress in human endothelial cells. FASEB J. 1999, 13, 1231–1238. [Google Scholar] [CrossRef]
  56. Caldefie-Chezet, F.; Poulin, A.; Tridon, A.; Sion, B.; Vasson, M.P. Leptin: A potential regulator of polymorphonuclear neutrophil bactericidal action? J. Leukoc. Biol. 2001, 69, 414–418. [Google Scholar] [CrossRef]
  57. Kim, J.A.; Park, H.S. White blood cell count and abdominal fat distribution in female obese adolescents. Metabolism 2008, 57, 1375–1379. [Google Scholar] [CrossRef]
  58. Rhee, H.; Love, T.; Harrington, D. Blood Neutrophil Count is Associated with Body Mass Index in Adolescents with Asthma. JSM Allergy Asthma 2018, 3, 1019. [Google Scholar]
  59. Scott, H.A.; Gibson, P.G.; Garg, M.L.; Wood, L.G. Airway inflammation is augmented by obesity and fatty acids in asthma. Eur. Respir. J. 2011, 38, 594–602. [Google Scholar] [CrossRef]
  60. Winsa-Lindmark, S.; Stridsman, C.; Sahlin, A.; Hedman, L.; Stenfors, N.; Myrberg, T.; Lindberg, A.; Rönmark, E.; Backman, H. Severity of adult-onset asthma—A matter of blood neutrophils and severe obesity. Respir. Med. 2023, 219, 107418. [Google Scholar] [CrossRef] [PubMed]
  61. Backman, H.; Winsa Lindmark, S.; Hedman, L.; Kankaanranta, H.; Warm, K.; Lindberg, A.; Bossios, A.; Rönmark, E.; Stridsman, C. The interplay between obesity and blood neutrophils in adult-onset asthma. Respir. Med. 2024, 222, 107529. [Google Scholar] [CrossRef]
  62. Tay, T.R.; Hew, M. Comorbid “treatable traits” in difficult asthma: Current evidence and clinical evaluation. Allergy 2018, 73, 1369–1382. [Google Scholar] [CrossRef]
  63. Kiljander, T.O.; Salomaa, E.R.; Hietanen, E.K.; Terho, E.O. Gastroesophageal reflux in asthmatics: A double-blind, placebo-controlled crossover study with omeprazole. Chest 1999, 116, 1257–1264. [Google Scholar] [CrossRef]
  64. Bor, S.; Kitapcioglu, G.; Solak, Z.A.; Ertilav, M.; Erdinc, M. Prevalence of gastroesophageal reflux disease in patients with asthma and chronic obstructive pulmonary disease. J. Gastroenterol. Hepatol. 2010, 25, 309–313. [Google Scholar] [CrossRef]
  65. Leggett, J.J.; Johnston, B.T.; Mills, M.; Gamble, J.; Heaney, L.G. Prevalence of gastroesophageal reflux in difficult asthma: Relationship to asthma outcome. Chest 2005, 127, 1227–1231. [Google Scholar] [CrossRef]
  66. Simpson, J.L.; Baines, K.J.; Ryan, N.; Gibson, P.G. Neutrophilic asthma is characterised by increased rhinosinusitis with sleep disturbance and GERD. Asian Pac. J. Allergy Immunol. 2014, 32, 66–74. [Google Scholar] [CrossRef]
  67. Paoletti, G.; Melone, G.; Ferri, S.; Puggioni, F.; Baiardini, I.; Racca, F.; Canonica, G.W.; Heffler, E.; Malipiero, G. Gastroesophageal reflux and asthma: When, how, and why. Curr. Opin. Allergy Clin. Immunol. 2021, 21, 52–58. [Google Scholar] [CrossRef]
  68. Takeda, N.; Takemura, M.; Kanemitsu, Y.; Hijikata, H.; Fukumitsu, K.; Asano, T.; Yamaba, Y.; Suzuki, M.; Kubota, E.; Kamiya, T.; et al. Effect of anti-reflux treatment on gastroesophageal reflux-associated chronic cough: Implications of neurogenic and neutrophilic inflammation. J. Asthma 2020, 57, 1202–1210. [Google Scholar] [CrossRef] [PubMed]
  69. Liu, T.; Woodruff, P.G.; Zhou, X. Advances in non-type 2 severe asthma: From molecular insights to novel treatment strategies. Eur. Respir. J. 2024, 64, 2300826. [Google Scholar] [CrossRef]
  70. Shimoda, T.; Obase, Y.; Kishikawa, R.; Iwanaga, T. Influence of cigarette smoking on airway inflammation and inhaled corticosteroid treatment in patients with asthma. Allergy Asthma Proc. 2016, 37, 50–58. [Google Scholar] [CrossRef] [PubMed]
  71. Siew, L.Q.C.; Wu, S.Y.; Ying, S.; Corrigan, C.J. Cigarette smoking increases bronchial mucosal IL-17A expression in asthmatics, which acts in concert with environmental aeroallergens to engender neutrophilic inflammation. Clin. Exp. Allergy 2017, 47, 740–750. [Google Scholar] [CrossRef] [PubMed]
  72. Belvisi, M.G.; Baker, K.; Malloy, N.; Raemdonck, K.; Dekkak, B.; Pieper, M.; Nials, A.T.; Birrell, M.A. Modelling the asthma phenotype: Impact of cigarette smoke exposure. Respir. Res. 2018, 19, 89. [Google Scholar] [CrossRef]
  73. Turan, N.; van der Veen, T.A.; Draijer, C.; Fattahi, F.; Ten Hacken, N.H.; Timens, W.; van Oosterhout, A.J.; van den Berge, M.; Melgert, B.N. Neutrophilic Asthma Is Associated With Smoking, High Numbers of IRF5+, and Low Numbers of IL10+ Macrophages. Front. Allergy 2021, 2, 676930. [Google Scholar] [CrossRef]
  74. Deng, K.; Zhang, X.; Liu, Y.; Zhang, L.; Wang, G.; Feng, M.; Oliver, B.G.; Wang, L.; Hansbro, P.M.; Qin, L.; et al. Heterogeneity of Paucigranulocytic Asthma: A Prospective Cohort Study with Hierarchical Cluster Analysis. J. Allergy Clin. Immunol. Pract. 2021, 9, 2344–2355. [Google Scholar] [CrossRef]
  75. Kuruvilla, M.E.; Lee, F.E.; Lee, G.B. Understanding Asthma Phenotypes, Endotypes, and Mechanisms of Disease. Clin. Rev. Allergy Immunol. 2019, 56, 219–233. [Google Scholar] [CrossRef]
  76. Papaioannou, A.I.; Fouka, E.; Ntontsi, P.; Stratakos, G.; Papiris, S. Paucigranulocytic Asthma: Potential Pathogenetic Mechanisms, Clinical Features and Therapeutic Management. J. Pers. Med. 2022, 12, 850. [Google Scholar] [CrossRef]
  77. Siwicki, M.; Kubes, P. Neutrophils in host defense, healing, and hypersensitivity: Dynamic cells within a dynamic host. J. Allergy Clin. Immunol. 2023, 151, 634–655. [Google Scholar] [CrossRef] [PubMed]
  78. Nathan, C. Neutrophils and immunity: Challenges and opportunities. Nat. Rev. Immunol. 2006, 6, 173–182. [Google Scholar] [CrossRef] [PubMed]
  79. Segal, A.W. How neutrophils kill microbes. Annu. Rev. Immunol. 2005, 23, 197–223. [Google Scholar] [CrossRef] [PubMed]
  80. Rosazza, T.; Warner, J.; Sollberger, G. NET formation—Mechanisms and how they relate to other cell death pathways. FEBS J. 2021, 288, 3334–3350. [Google Scholar] [CrossRef]
  81. Brinkmann, V.; Reichard, U.; Goosmann, C.; Fauler, B.; Uhlemann, Y.; Weiss, D.S.; Weinrauch, Y.; Zychlinsky, A. Neutrophil extracellular traps kill bacteria. Science 2004, 303, 1532–1535. [Google Scholar] [CrossRef]
  82. Lämmermann, T.; Afonso, P.V.; Angermann, B.R.; Wang, J.M.; Kastenmüller, W.; Parent, C.A.; Germain, R.N. Neutrophil swarms require LTB4 and integrins at sites of cell death in vivo. Nature 2013, 498, 371–375. [Google Scholar] [CrossRef]
  83. Uderhardt, S.; Martins, A.J.; Tsang, J.S.; Lämmermann, T.; Germain, R.N. Resident Macrophages Cloak Tissue Microlesions to Prevent Neutrophil-Driven Inflammatory Damage. Cell 2019, 177, 541–555.e17. [Google Scholar] [CrossRef]
  84. Christoffersson, G.; Vågesjö, E.; Vandooren, J.; Lidén, M.; Massena, S.; Reinert, R.B.; Brissova, M.; Powers, A.C.; Opdenakker, G.; Phillipson, M. VEGF-A recruits a proangiogenic MMP-9-delivering neutrophil subset that induces angiogenesis in transplanted hypoxic tissue. Blood 2012, 120, 4653–4662. [Google Scholar] [CrossRef] [PubMed]
  85. Ballesteros, I.; Rubio-Ponce, A.; Genua, M.; Lusito, E.; Kwok, I.; Fernández-Calvo, G.; Khoyratty, T.E.; van Grinsven, E.; González-Hernández, S.; Nicolás-Ávila, J.Á.; et al. Co-option of neutrophil fates by tissue environments. Cell 2020, 183, 1282–1297.e18. [Google Scholar] [CrossRef]
  86. Kurimoto, T.; Yin, Y.; Habboub, G.; Gilbert, H.Y.; Li, Y.; Nakao, S.; Hafezi-Moghadam, A.; Benowitz, L.I. Neutrophils express oncomodulin and promote optic nerve regeneration. J. Neurosci. 2013, 33, 14816–14824. [Google Scholar] [CrossRef]
  87. Kunkel, S.L.; Standiford, T.; Kasahara, K.; Strieter, R.M. Interleukin-8 (IL-8): The major neutrophil chemotactic factor in the lung. Exp. Lung Res. 1991, 17, 17–23. [Google Scholar] [CrossRef]
  88. Chapman, R.W.; Phillips, J.E.; Hipkin, R.W.; Curran, A.K.; Lundell, D.; Fine, J.S. CXCR2 antagonists for the treatment of pulmonary disease. Pharmacol. Ther. 2009, 121, 55–68. [Google Scholar] [CrossRef]
  89. Varricchi, G.; Modestino, L.; Poto, R.; Cristinziano, L.; Gentile, L.; Postiglione, L.; Spadaro, G.; Galdiero, M.R. Neutrophil extracellular traps and neutrophil-derived mediators as possible biomarkers in bronchial asthma. Clin. Exp. Med. 2022, 22, 285–300. [Google Scholar] [CrossRef]
  90. Crisford, H.; Sapey, E.; Stockley, R.A. Proteinase 3; a potential target in chronic obstructive pulmonary disease and other chronic inflammatory diseases. Respir. Res. 2018, 19, 180. [Google Scholar] [CrossRef] [PubMed]
  91. Gu, W.; Huang, C.; Chen, G.; Kong, W.; Zhao, L.; Jie, H.; Zhen, G. The role of extracellular traps released by neutrophils, eosinophils, and macrophages in asthma. Respir. Res. 2024, 25, 290. [Google Scholar] [CrossRef] [PubMed]
  92. Papayannopoulos, V.; Metzler, K.D.; Hakkim, A.; Zychlinsky, A. Neutrophil elastase and myeloperoxidase regulate the formation of neutrophil extracellular traps. J. Cell Biol. 2010, 191, 677–691. [Google Scholar] [CrossRef]
  93. Lewis, B.W.; Ford, M.L.; Rogers, L.K.; Britt, R.D., Jr. Oxidative Stress Promotes Corticosteroid Insensitivity in Asthma and COPD. Antioxidants 2021, 10, 1335. [Google Scholar] [CrossRef]
  94. Metzler, K.D.; Fuchs, T.A.; Nauseef, W.M.; Reumaux, D.; Roesler, J.; Schulze, I.; Wahn, V.; Papayannopoulos, V.; Zychlinsky, A. Myeloperoxidase is required for neutrophil extracellular trap formation: Implications for innate immunity. Blood 2011, 117, 953–959. [Google Scholar] [CrossRef] [PubMed]
  95. Postma, D.S.; Renkema, T.E.; Noordhoek, J.A.; Faber, H.; Sluiter, H.J.; Kauffman, H. Association between nonspecific bronchial hyperreactivity and superoxide anion production by polymorphonuclear leukocytes in chronic air-flow obstruction. Am. Rev. Respir. Dis. 1988, 137, 57–61. [Google Scholar] [CrossRef]
  96. Sanders, S.P. Nitric oxide in asthma. Pathogenic, therapeutic, or diagnostic? Am. J. Respir. Cell Mol. Biol. 1999, 21, 147–149. [Google Scholar] [CrossRef] [PubMed]
  97. Comhair, S.A.; Bhathena, P.R.; Farver, C.; Thunnissen, F.B.; Erzurum, S.C. Extracellular glutathione peroxidase induction in asthmatic lungs: Evidence for redox regulation of expression in human airway epithelial cells. FASEB J. 2001, 15, 70–78. [Google Scholar] [CrossRef]
  98. Makena, P.; Kikalova, T.; Prasad, G.L.; Baxter, S.A. Oxidative Stress and Lung Fibrosis: Towards an Adverse Outcome Pathway. Int. J. Mol. Sci. 2023, 24, 12490. [Google Scholar] [CrossRef] [PubMed]
  99. Gentile, F.; Arcaro, A.; Pizzimenti, S.; Daga, M.; Cetrangolo, G.P.; Dianzani, C.; Lepore, A.; Graf, M.; Ames, P.R.J.; Barrera, G. DNA damage by lipid peroxidation products: Implications in cancer, inflammation and autoimmunity. AIMS Genet. 2017, 4, 103–137. [Google Scholar] [CrossRef]
  100. Hawkins, C.L.; Davies, M.J. Detection, identification, and quantification of oxidative protein modifications. J. Biol. Chem. 2019, 294, 19683–19708. [Google Scholar] [CrossRef]
  101. Juan, C.A.; Pérez de la Lastra, J.M.; Plou, F.J.; Pérez-Lebeña, E. The Chemistry of Reactive Oxygen Species (ROS) Revisited: Outlining Their Role in Biological Macromolecules (DNA, Lipids and Proteins) and Induced Pathologies. Int. J. Mol. Sci. 2021, 22, 4642. [Google Scholar] [CrossRef]
  102. Kettritz, R.; Choi, M.; Rolle, S.; Wellner, M.; Luft, F.C. Integrins and cytokines activate nuclear transcription factor-kappaB in human neutrophils. J. Biol. Chem. 2004, 279, 2657–2665. [Google Scholar] [CrossRef]
  103. Barnabas, M.; Awakan, O.J.; Rotimi, D.E.; Akanji, M.A.; Adeyemi, O.S. Exploring redox imbalance and inflammation for asthma therapy. Mol. Biol. Rep. 2023, 50, 7851–7865. [Google Scholar] [CrossRef] [PubMed]
  104. Wright, T.K.; Gibson, P.G.; Simpson, J.L.; McDonald, V.M.; Wood, L.G.; Baines, K.J. Neutrophil extracellular traps are associated with inflammation in chronic airway disease. Respirology 2016, 21, 467–475. [Google Scholar] [CrossRef]
  105. Li, Y.; Yang, Y.; Gan, T.; Zhou, J.; Hu, F.; Hao, N.; Yuan, B.; Chen, Y.; Zhang, M. Extracellular RNAs from lung cancer cells activate epithelial cells and induce neutrophil extracellular traps. Int. J. Oncol. 2019, 55, 69–80. [Google Scholar] [CrossRef]
  106. Pham, D.L.; Ban, G.Y.; Kim, S.H.; Shin, Y.S.; Ye, Y.M.; Chwae, Y.J.; Park, H.S. Neutrophil autophagy and extracellular DNA traps contribute to airway inflammation in severe asthma. Clin. Exp. Allergy 2017, 47, 57–70. [Google Scholar] [CrossRef]
  107. Liang, Y.; Hou, C.; Kong, J.; Wen, H.; Zheng, X.; Wu, L.; Huang, H.; Chen, Y. HMGB1 binding to receptor for advanced glycation end products enhances inflammatory responses of human bronchial epithelial cells by activating p38 MAPK and ERK1/2. Mol. Cell Biochem. 2015, 405, 63–71. [Google Scholar] [CrossRef]
  108. Hosoki, K.; Ying, S.; Corrigan, C.; Qi, H.; Kurosky, A.; Jennings, K.; Sun, Q.; Boldogh, I.; Sur, S. Analysis of a Panel of 48 Cytokines in BAL Fluids Specifically Identifies IL-8 Levels as the Only Cytokine that Distinguishes Controlled Asthma from Uncontrolled Asthma, and Correlates Inversely with FEV1. PLoS ONE 2015, 10, e0126035. [Google Scholar] [CrossRef]
  109. Huang, A.X.; Lu, L.W.; Liu, W.J.; Huang, M. Plasma Inflammatory Cytokine IL-4, IL-8, IL-10, and TNF-α Levels Correlate with Pulmonary Function in Patients with Asthma-Chronic Obstructive Pulmonary Disease (COPD) Overlap Syndrome. Med. Sci. Monit. 2016, 22, 2800–2808. [Google Scholar] [CrossRef]
  110. Parnes, J.R.; Molfino, N.A.; Colice, G.; Martin, U.; Corren, J.; Menzies-Gow, A. Targeting TSLP in Asthma. J. Asthma Allergy 2022, 15, 749–765. [Google Scholar] [CrossRef]
  111. Ying, S.; O’Connor, B.; Ratoff, J.; Meng, Q.; Mallett, K.; Cousins, D.; Robinson, D.; Zhang, G.; Zhao, J.; Lee, T.H.; et al. Thymic stromal lymphopoietin expression is increased in asthmatic airways and correlates with expression of Th2-attracting chemokines and disease severity. J. Immunol. 2005, 174, 8183–8190. [Google Scholar] [CrossRef] [PubMed]
  112. Ying, S.; O’Connor, B.; Ratoff, J.; Meng, Q.; Fang, C.; Cousins, D.; Zhang, G.; Gu, S.; Gao, Z.; Shamji, B.; et al. Expression and cellular provenance of thymic stromal lymphopoietin and chemokines in patients with severe asthma and chronic obstructive pulmonary disease. J. Immunol. 2008, 181, 2790–2798. [Google Scholar] [CrossRef] [PubMed]
  113. Allakhverdi, Z.; Comeau, M.R.; Jessup, H.K.; Yoon, B.R.; Brewer, A.; Chartier, S.; Paquette, N.; Ziegler, S.F.; Sarfati, M.; Delespesse, G. Thymic stromal lymphopoietin is released by human epithelial cells in response to microbes, trauma, or inflammation and potently activates mast cells. J. Exp. Med. 2007, 204, 253–258. [Google Scholar] [CrossRef]
  114. Ito, T.; Wang, Y.H.; Duramad, O.; Hori, T.; Delespesse, G.J.; Watanabe, N.; Qin, F.X.; Yao, Z.; Cao, W.; Liu, Y.J. TSLP-activated dendritic cells induce an inflammatory T helper type 2 cell response through OX40 ligand. J. Exp. Med. 2005, 202, 1213–1223. [Google Scholar] [CrossRef]
  115. Gilliet, M.; Soumelis, V.; Watanabe, N.; Hanabuchi, S.; Antonenko, S.; de Waal-Malefyt, R.; Liu, Y.J. Human dendritic cells activated by TSLP and CD40L induce proallergic cytotoxic T cells. J. Exp. Med. 2003, 197, 1059–1063. [Google Scholar] [CrossRef]
  116. Tanaka, J.; Watanabe, N.; Kido, M.; Saga, K.; Akamatsu, T.; Nishio, A.; Chiba, T. Human TSLP and TLR3 ligands promote differentiation of Th17 cells with a central memory phenotype under Th2-polarizing conditions. Clin. Exp. Allergy 2009, 39, 89–100. [Google Scholar] [CrossRef]
  117. Uddin, M.; Seumois, G.; Lau, L.C.; Rytila, P.; Davies, D.E.; Djukanovic, R. Enhancement of neutrophil function by the bronchial epithelium stimulated by epidermal growth factor. Eur. Respir. J. 2008, 31, 714–724. [Google Scholar] [CrossRef]
  118. Yamasaki, A.; Okazaki, R.; Harada, T. Neutrophils and Asthma. Diagnostics 2022, 12, 1175. [Google Scholar] [CrossRef] [PubMed]
  119. Al-Ramli, W.; Préfontaine, D.; Chouiali, F.; Martin, J.G.; Olivenstein, R.; Lemière, C.; Hamid, Q. T(H)17-associated cytokines (IL-17A and IL-17F) in severe asthma. J. Allergy Clin. Immunol. 2009, 123, 1185–1187. [Google Scholar] [CrossRef] [PubMed]
  120. Bullens, D.M.; Truyen, E.; Coteur, L.; Dilissen, E.; Hellings, P.W.; Dupont, L.J.; Ceuppens, J.L. IL-17 mRNA in sputum of asthmatic patients: Linking T cell driven inflammation and granulocytic influx? Respir. Res. 2006, 7, 135. [Google Scholar] [CrossRef] [PubMed]
  121. Vazquez-Tello, A.; Semlali, A.; Chakir, J.; Martin, J.G.; Leung, D.Y.; Eidelman, D.H.; Hamid, Q. Induction of glucocorticoid receptor-beta expression in epithelial cells of asthmatic airways by T-helper type 17 cytokines. Clin. Exp. Allergy 2010, 40, 1312–1322. [Google Scholar] [CrossRef]
  122. Altieri, A.; Piyadasa, H.; Hemshekhar, M.; Osawa, N.; Recksiedler, B.; Spicer, V.; Hiemstra, P.S.; Halayko, A.J.; Mookherjee, N. Combination of IL-17A/F and TNF-α uniquely alters the bronchial epithelial cell proteome to enhance proteins that augment neutrophil migration. J. Inflamm. 2022, 19, 26. [Google Scholar] [CrossRef]
  123. Mukaida, N. Interleukin-8: An expanding universe beyond neutrophil chemotaxis and activation. Int. J. Hematol. 2000, 72, 391–398. [Google Scholar] [PubMed]
  124. Charrad, R.; Kaabachi, W.; Rafrafi, A.; Berraies, A.; Hamzaoui, K.; Hamzaoui, A. IL-8 Gene Variants and Expression in Childhood Asthma. Lung 2017, 195, 749–757. [Google Scholar] [CrossRef]
  125. Dimitrova, D.; Youroukova, V.; Ivanova-Todorova, E.; Tumangelova-Yuzeir, K.; Velikova, T. Serum levels of IL-5, IL-6, IL-8, IL-13 and IL-17A in pre-defined groups of adult patients with moderate and severe bronchial asthma. Respir. Med. 2019, 154, 144–154. [Google Scholar] [CrossRef]
  126. Zhang, J.; Bai, C. Elevated Serum Interleukin-8 Level as a Preferable Biomarker for Identifying Uncontrolled Asthma and Glucocorticosteroid Responsiveness. Tanaffos 2017, 16, 260–269. [Google Scholar]
  127. Nocker, R.E.; Schoonbrood, D.F.; van de Graaf, E.A.; Hack, C.E.; Lutter, R.; Jansen, H.M.; Out, T.A. Interleukin-8 in airway inflammation in patients with asthma and chronic obstructive pulmonary disease. Int. Arch. Allergy Immunol. 1996, 109, 183–191. [Google Scholar] [CrossRef]
  128. Norzila, M.Z.; Fakes, K.; Henry, R.L.; Simpson, J.; Gibson, P.G. Interleukin-8 secretion and neutrophil recruitment accompanies induced sputum eosinophil activation in children with acute asthma. Am. J. Respir. Crit. Care Med. 2000, 161 Pt 1, 769–774. [Google Scholar] [CrossRef]
  129. Lopez-Castejon, G.; Brough, D. Understanding the mechanism of IL-1β secretion. Cytokine Growth Factor Rev. 2011, 22, 189–195. [Google Scholar] [CrossRef]
  130. Thornberry, N.A.; Bull, H.G.; Calaycay, J.R.; Chapman, K.T.; Howard, A.D.; Kostura, M.J.; Miller, D.K.; Molineaux, S.M.; Weidner, J.R.; Aunins, J.; et al. A novel heterodimeric cysteine protease is required for interleukin-1 beta processing in monocytes. Nature 1992, 356, 768–774. [Google Scholar] [CrossRef]
  131. Kim, R.Y.; Pinkerton, J.W.; Essilfie, A.T.; Robertson, A.A.B.; Baines, K.J.; Brown, A.C.; Mayall, J.R.; Ali, M.K.; Starkey, M.R.; Hansbro, N.G.; et al. Role for NLRP3 Inflammasome-mediated, IL-1β-Dependent Responses in Severe, Steroid-Resistant Asthma. Am. J. Respir. Crit. Care Med. 2017, 196, 283–297. [Google Scholar] [CrossRef] [PubMed]
  132. Baines, K.J.; Simpson, J.L.; Wood, L.G.; Scott, R.J.; Gibson, P.G. Transcriptional phenotypes of asthma defined by gene expression profiling of induced sputum samples. J. Allergy Clin. Immunol. 2011, 127, 153–160.e9. [Google Scholar] [CrossRef] [PubMed]
  133. Saikumar Jayalatha, A.K.; Hesse, L.; Ketelaar, M.E.; Koppelman, G.H.; Nawijn, M.C. The central role of IL-33/IL-1RL1 pathway in asthma: From pathogenesis to intervention. Pharmacol. Ther. 2021, 225, 107847. [Google Scholar] [CrossRef] [PubMed]
  134. Calderon, A.A.; Dimond, C.; Choy, D.F.; Pappu, R.; Grimbaldeston, M.A.; Mohan, D.; Chung, K.F. Targeting interleukin-33 and thymic stromal lymphopoietin pathways for novel pulmonary therapeutics in asthma and COPD. Eur. Respir. Rev. 2023, 32, 220144. [Google Scholar] [CrossRef]
  135. Griesenauer, B.; Paczesny, S. The ST2/IL-33 Axis in Immune Cells during Inflammatory Diseases. Front. Immunol. 2017, 8, 475. [Google Scholar] [CrossRef]
  136. Quoc, Q.L.; Cao, T.B.T.; Jang, J.H.; Shin, Y.S.; Choi, Y.; Park, H.S. ST2-Mediated Neutrophilic Airway Inflammation: A Therapeutic Target for Patients with Uncontrolled Asthma. Allergy Asthma Immunol. Res. 2024, 16, 22–41. [Google Scholar] [CrossRef]
  137. Lefrançais, E.; Roga, S.; Gautier, V.; Gonzalez-de-Peredo, A.; Monsarrat, B.; Girard, J.P.; Cayrol, C. IL-33 is processed into mature bioactive forms by neutrophil elastase and cathepsin G. Proc. Natl. Acad. Sci. USA 2012, 109, 1673–1678. [Google Scholar] [CrossRef]
  138. Van Nevel, S.; van Ovost, J.; Holtappels, G.; DeRuyck, N.; Zhang, N.; Braun, H.; Maes, T.; Bachert, C.; Krysko, O. Neutrophils affect IL-33 processing in response to the respiratory allergen Alternaria. Front. Immunol. 2021, 12, 677848. [Google Scholar] [CrossRef]
  139. Curren, B.; Ahmed, T.; Howard, D.R.; Ashik Ullah, M.; Sebina, I.; Rashid, R.B.; Al Amin Sikder, M.; Namubiru, P.; Bissell, A.; Ngo, S.; et al. IL-33-induced neutrophilic inflammation and NETosis underlie rhinovirus-triggered exacerbations of asthma. Mucosal Immunol. 2023, 16, 671–684. [Google Scholar] [CrossRef]
  140. Syabbalo, N. Role of IL-17 and IL-23 in the Pathogenesis of Neutrophilic Asthma. Int. J. Immunol. Immunother. 2020, 7, 049. [Google Scholar] [CrossRef]
  141. Xiong, D.J.P.; Martin, J.G.; Lauzon, A.M. Airway smooth muscle function in asthma. Front. Physiol. 2022, 13, 993406. [Google Scholar] [CrossRef] [PubMed]
  142. Cundall, M.; Sun, Y.; Miranda, C.; Trudeau, J.B.; Barnes, S.; Wenzel, S.E. Neutrophil-derived matrix metalloproteinase-9 is increased in severe asthma and poorly inhibited by glucocorticoids. J. Allergy Clin. Immunol. 2003, 112, 1064–1071. [Google Scholar] [CrossRef] [PubMed]
  143. Nadel, J.A. Role of enzymes from inflammatory cells on airway submucosal gland secretion. Respiration 1991, 58 (Suppl. S1), 3–5. [Google Scholar] [CrossRef]
  144. Vignola, A.M.; Bonanno, A.; Mirabella, A.; Riccobono, L.; Mirabella, F.; Profita, M.; Bellia, V.; Bousquet, J.; Bonsignore, G. Increased levels of elastase and alpha1-antitrypsin in sputum of asthmatic patients. Am. J. Respir. Crit. Care Med. 1998, 157, 505–511. [Google Scholar] [CrossRef]
  145. Chang, Y.; Al-Alwan, L.; Risse, P.A.; Roussel, L.; Rousseau, S.; Halayko, A.J.; Martin, J.G.; Hamid, Q.; Eidelman, D.H. TH17 cytokines induce human airway smooth muscle cell migration. J. Allergy Clin. Immunol. 2011, 127, 1046–1053.e2. [Google Scholar] [CrossRef]
  146. Gaffen, S.L.; Jain, R.; Garg, A.V.; Cua, D.J. The IL-23-IL-17 immune axis: From mechanisms to therapeutic testing. Nat. Rev. Immunol. 2014, 14, 585–600. [Google Scholar] [CrossRef]
  147. Halwani, R.; Al-Muhsen, S.; Al-Jahdali, H.; Hamid, Q. Role of transforming growth factor-β in airway remodeling in asthma. Am. J. Respir. Cell Mol. Biol. 2011, 44, 127–133. [Google Scholar] [CrossRef]
  148. Chu, H.W.; Trudeau, J.B.; Balzar, S.; Wenzel, S.E. Peripheral blood and airway tissue expression of transforming growth factor beta by neutrophils in asthmatic subjects and normal control subjects. J. Allergy Clin. Immunol. 2000, 106, 1115–1123. [Google Scholar] [CrossRef]
  149. Klironomou, A.; Papaiakovou, G.; Bakakos, A.; Anagnostopoulos, N.; Koukaki, E.; Theofani, E.; Semitekolou, M.; Rovina, N. Tezepelumab: A promising therapy for severe uncontrolled asthma. Explor. Asthma Allergy 2024, 2, 485–501. [Google Scholar] [CrossRef]
  150. Busse, W.W.; Holgate, S.; Kerwin, E.; Chon, Y.; Feng, J.; Lin, J.; Lin, S.L. Randomized, double-blind, placebo-controlled study of brodalumab, a human anti-IL-17 receptor monoclonal antibody, in moderate to severe asthma. Am. J. Respir. Crit. Care Med. 2013, 188, 1294–1302. [Google Scholar] [CrossRef] [PubMed]
  151. Amgen. A Randomized, Double-Blind, Placebo-Controlled Study to Evaluate the Safety and Efficacy of Brodalumab in Subjects with Inadequately Controlled Asthma and High Bronchodilator Reversibility; Report No.: NCT01902290; ClinicalTrials.gov: Bethesda, MD, USA, 2022. Available online: https://clinicaltrials.gov/study/NCT01902290 (accessed on 21 September 2022).
  152. Pujol, J.L.; Cosso, B.; Daurès, J.P.; Clot, J.; Michel, F.B.; Godard, P. Interleukin-1 release by alveolar macrophages in asthmatic patients and healthy subjects. Int. Arch. Allergy Appl. Immunol. 1990, 91, 207–210. [Google Scholar] [CrossRef] [PubMed]
  153. Hernandez, M.L.; Mills, K.; Almond, M.; Todoric, K.; Aleman, M.M.; Zhang, H.; Zhou, H.; Peden, D.B. IL-1 receptor antagonist reduces endotoxin-induced airway inflammation in healthy volunteers. J. Allergy Clin. Immunol. 2015, 135, 379–385. [Google Scholar] [CrossRef] [PubMed]
  154. Ulich, T.R.; Yin, S.M.; Guo, K.Z.; del Castillo, J.; Eisenberg, S.P.; Thompson, R.C. The intratracheal administration of endotoxin and cytokines. III. The interleukin-1 (IL-1) receptor antagonist inhibits endotoxin- and IL-1-induced acute inflammation. Am. J. Pathol. 1991, 138, 521–524. [Google Scholar]
  155. Sabroe, I.; Lloyd, C.M.; Whyte, M.K.; Dower, S.K.; Williams, T.J.; Pease, J.E. Chemokines, innate and adaptive immunity, and respiratory disease. Eur. Respir. J. 2002, 19, 350–355. [Google Scholar] [CrossRef]
  156. Leaker, B.R.; Barnes, P.J.; O’Connor, B. Inhibition of LPS-induced airway neutrophilic inflammation in healthy volunteers with an oral CXCR2 antagonist. Respir. Res. 2013, 14, 137. [Google Scholar] [CrossRef]
  157. Nair, P.; Gaga, M.; Zervas, E.; Alagha, K.; Hargreave, F.E.; O’Byrne, P.M.; Stryszak, P.; Gann, L.; Sadeh, J.; Chanez, P.; et al. Safety and efficacy of a CXCR2 antagonist in patients with severe asthma and sputum neutrophils: A randomized, placebo-controlled clinical trial. Clin. Exp. Allergy 2012, 42, 1097–1103. [Google Scholar] [CrossRef]
  158. O’Byrne, P.M.; Metev, H.; Puu, M.; Richter, K.; Keen, C.; Uddin, M.; Larsson, B.; Cullberg, M.; Nair, P. Efficacy and safety of a CXCR2 antagonist, AZD5069, in patients with uncontrolled persistent asthma: A randomised, double-blind, placebo-controlled trial. Lancet Respir. Med. 2016, 4, 797–806. [Google Scholar] [CrossRef]
  159. Chu, D.K.; Al-Garawi, A.; Llop-Guevara, A.; Pillai, R.A.; Radford, K.; Shen, P.; Walker, T.D.; Goncharova, S.; Calhoun, W.J.; Nair, P.; et al. Therapeutic potential of anti-IL-6 therapies for granulocytic airway inflammation in asthma. Allergy Asthma Clin. Immunol. 2015, 11, 14. [Google Scholar] [CrossRef] [PubMed]
  160. Revez, J.A.; Bain, L.M.; Watson, R.M.; Towers, M.; Collins, T.; Killian, K.J.; O’Byrne, P.M.; Gauvreau, G.M.; Upham, J.W.; Ferreira, M.A. Effects of interleukin-6 receptor blockade on allergen-induced airway responses in mild asthmatics. Clin. Transl. Immunol. 2019, 8, e1044. [Google Scholar] [CrossRef]
  161. University of North Carolina. PrecISE (Precision Interventions for Severe and/or Exacerbation-Prone Asthma) Network Study; ClinicalTrials.gov: Bethesda, MD, USA, 2025. Available online: https://www.clinicaltrials.gov/study/NCT04129931 (accessed on 19 June 2025).
  162. Kelsen, S.G.; Agache, I.O.; Soong, W.; Israel, E.; Chupp, G.L.; Cheung, D.S.; Theess, W.; Yang, X.; Staton, T.L.; Choy, D.F.; et al. Astegolimab (anti-ST2) efficacy and safety in adults with severe asthma: A randomized clinical trial. J. Allergy Clin. Immunol. 2021, 148, 790–798. [Google Scholar] [CrossRef] [PubMed]
  163. Wechsler, M.E.; Ruddy, M.K.; Pavord, I.D.; Israel, E.; Rabe, K.F.; Ford, L.B.; Maspero, J.F.; Abdulai, R.M.; Hu, C.C.; Martincova, R.; et al. Efficacy and Safety of Itepekimab in Patients with Moderate-to-Severe Asthma. N. Engl. J. Med. 2021, 385, 1656–1668. [Google Scholar] [CrossRef]
  164. Rabe, K.F.; Martinez, F.J.; Bhatt, S.P.; Kawayama, T.; Cosio, B.G.; Mroz, R.M.; Boomsma, M.M.; Goulaouic, H.; Nivens, M.C.; Djandji, M.; et al. AERIFY-1/2: Two phase 3, randomised, controlled trials of itepekimab in former smokers with moderate-to-severe COPD. ERJ Open Res. 2024, 10, 00718–02023. [Google Scholar] [CrossRef] [PubMed]
  165. Kosloski, M.P.; Guttman-Yassky, E.; Cork, M.J.; Worm, M.; Nahm, D.H.; Zhu, X.; Ruddy, M.K.; Harel, S.; Kamal, M.A.; Goulaouic, H.; et al. Pharmacokinetics and pharmacodynamics of itepekimab in adults with moderate-to-severe atopic dermatitis: Results from two terminated phase II trials. Clin. Transl. Sci. 2024, 17, e13874. [Google Scholar] [CrossRef]
  166. Corren, J.; Reid, F.; Moate, R.; Jimenez, E.; Sadiq, M.; Williams, A.; Rytelewski, M.; Muthas, D.; Brooks, D.; Lindqist, E.; et al. Frontier-3: A randomized, phase 2a study to evaluate the efficacy and safety of torozakimab (an interleukin-33 monoclonal antibody) in early-onset asthma. Thorax 2024, 79 (Suppl. S2), A65. [Google Scholar]
  167. Kere, M.; Klevebro, S.; Hernandez-Pacheco, N.; Ödling, M.; Ekström, S.; Mogensen, I.; Janson, C.; Palmberg, L.; van Hage, M.; Georgelis, A.; et al. Exploring proteomic plasma biomarkers in eosinophilic and neutrophilic asthma. Clin. Exp. Allergy 2023, 53, 186–197. [Google Scholar] [CrossRef]
  168. Quoc, Q.L.; Choi, Y.; Thi Bich, T.C.; Yang, E.M.; Shin, Y.S.; Park, H.S. S100A9 in adult asthmatic patients: A biomarker for neutrophilic asthma. Exp. Mol. Med. 2021, 53, 1170–1179. [Google Scholar] [CrossRef]
  169. Bich, T.; Quoc, Q.; Choi, Y.; Yang, E.; Trinh, H.; Shin, Y.; Park, H.S. Serum amyloid a1: A biomarker for neutrophilic airway inflammation in adult asthmatic patients. Allergy Asthma Immunol. Res. 2022, 14, 40–58. [Google Scholar] [CrossRef] [PubMed]
  170. Lin, Q.; Ni, H.; Zhong, J.; Zheng, Z.; Nie, H. Identification of hub genes and potential biomarkers of neutrophilic asthma: Evidence from a bioinformatics analysis. J. Asthma 2022, 60, 348–359. [Google Scholar] [CrossRef]
  171. Specjalski, K.; Romantowski, J.; Niedoszytko, M. YKL-40 as a possible marker of neutrophilic asthma. Front. Med. 2023, 10, 1115938. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Neutrophils-associated enzymes in asthma.
Figure 1. Neutrophils-associated enzymes in asthma.
Jcm 14 07137 g001
Figure 2. Neutrophils-associated cytokines in asthma.
Figure 2. Neutrophils-associated cytokines in asthma.
Jcm 14 07137 g002
Table 1. Cut-offs used by several authors to define the population with neutrophilic asthma.
Table 1. Cut-offs used by several authors to define the population with neutrophilic asthma.
StudyPopulation IncludedMaterial AssessedNeutrophilic Asthma Diagnostic Criteria
Green et al. [4]259 patients with asthma
34 healthy controls
induced sputumneutrophilia > 65%
Moore et al. [6]423 severe asthma patientsinduced sputumneutrophilia > 40%
Simpson et al. [5]93 patients with asthma
42 healthy controls
induced sputumneutrophilia > 61%
Bullone et al. [11]70 patients with mild-to-severe asthmabronchial biopsies47.17 neutrophils/mm2
Belda et al. [12]118 healthy non-smokersinduced sputumrepeated neutrophilia > 65% or ≥5 × 109/L (at least twice)
Demarche
et al. [7]
833 patients with asthma
94 healthy subjects
induced sputumneutrophil count ≥ 76%
Grunwell
et al. [9]
68 children with severe asthmaBALneutrophil count ≥ 5%
Brooks et al. [13] 194 patients with asthma
243 non-asthmatics
induced sputumneutrophilia 62–76%
depending on age
Schleich et al. [14]508 patients with asthmainduced sputumneutrophil count ≥ 76%
Gibson et al.
[15]
56 non-smoking patients with asthmainduced sputumneutrophilia > 64%, 283 × 106/mL
Shi et al. [16]232 patients with asthmainduced sputum
blood
neutrophilia > 61%
blood neutrophil rate > 69%
Table 2. Comparison of major clinical characteristics of eosinophilic and neutrophilic asthma.
Table 2. Comparison of major clinical characteristics of eosinophilic and neutrophilic asthma.
Neutrophilic AsthmaEosinophilic Asthma
Asthma onsetIn adults, often after the age of 40Mostly before the age of 40, very often in children and young adults
Asthma triggersInfections, air pollution, smokingAllergens
Immunological mechanismT2-lowT2-high, often IgE-mediated
Response to ICSPoorVery good
Clinical courseQuite often severe,
exacerbation-prone
In most patients easy to control with standard therapy
Biomarkers used in practiceNoneBlood eosinophilia
FeNO
tIgE
Concomitant
Diseases
Obesity
Smoking
GERD
Allergic rhinitis
Atopic dermatitis
Chronic sinusitis
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Iwaszko, I.; Specjalski, K.; Chełmińska, M.; Niedoszytko, M. Neutrophilic Asthma—From Mechanisms to New Perspectives of Therapy. J. Clin. Med. 2025, 14, 7137. https://doi.org/10.3390/jcm14207137

AMA Style

Iwaszko I, Specjalski K, Chełmińska M, Niedoszytko M. Neutrophilic Asthma—From Mechanisms to New Perspectives of Therapy. Journal of Clinical Medicine. 2025; 14(20):7137. https://doi.org/10.3390/jcm14207137

Chicago/Turabian Style

Iwaszko, Ilona, Krzysztof Specjalski, Marta Chełmińska, and Marek Niedoszytko. 2025. "Neutrophilic Asthma—From Mechanisms to New Perspectives of Therapy" Journal of Clinical Medicine 14, no. 20: 7137. https://doi.org/10.3390/jcm14207137

APA Style

Iwaszko, I., Specjalski, K., Chełmińska, M., & Niedoszytko, M. (2025). Neutrophilic Asthma—From Mechanisms to New Perspectives of Therapy. Journal of Clinical Medicine, 14(20), 7137. https://doi.org/10.3390/jcm14207137

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop