Journal of Pulmonary Medicine & Respiratory Research Category: Medical Type: Research Article

Clinical Features and Management of Neutrophilic Asthma

Nightingale Syabbalo1*
1 Professor of medicine and physiology, Copperbelt University, M.C. Sata School of Medicine, Kitwe, Zambia

*Corresponding Author(s):
Nightingale Syabbalo
Professor Of Medicine And Physiology, Copperbelt University, M.C. Sata School Of Medicine, Kitwe, Zambia
Tel:+260 966486117,
Email:nightsyab@gmail.com

Received Date: Aug 06, 2020
Accepted Date: Aug 12, 2020
Published Date: Aug 19, 2020

Abstract

Asthma is a chronic inflammatory airway disease with several distinct phenotypes, characterized by different immunopathological pathways, clinical presentation, severity of the disease and response to treatment. The phenotypes of asthma include eosinophilic, neutrophilic, mixed granulocytic and paucigranulocytic asthma. Approximately 3.6-10% of patients with asthma have severe refractory disease, which is uncontrolled on high doses of inhaled corticosteroids and long-acting β2-agonists. Some of these individuals with severe disease suffer from neutrophilic phenotype. Neutrophilic asthma is a severe and persistent disease, with frequent exacerbations and hospitalizations. It is characterized by the presence of high levels of neutrophils in the lungs and airways and fixed airflow obstruction. The T Helper 17 lymphocytes (Th17) cytokines, Interleukin-17 (IL-17) and IL-17F play an important role in the pathogenesis of neutrophilic asthma. IL-17 plays a key role in the immunophysiology of neutrophilic asthma by expressing the secretion of chemoattractant cytokines, chemokines, adhesion molecules and growth factors which lead to the recruitment and activation of neutrophils. Activated neutrophils release multiple proteinases, cytokines, chemokines and reactive oxygen species which cause airway epithelial cell injury, inflammation, hyperresponsiveness and airway remodeling. Neutrophilic asthma is unresponsive to high dose inhaled corticosteroids and to novel monoclonal antibody therapies. There is need for targeted precision biologics and other treatment modalities for patients with neutrophilic asthma, such as long-acting phosphodiesterase 4 inhibitors, macrolide antibiotics and bronchial thermoplasty.

Keywords

Airway smooth muscle; Bronchial thermoplasty; Macrolides; Neutrophilic asthma

INTRODUCTION

Asthma is a significant public health problem, affecting more than 358 million individuals globally [1] and its prevalence has been increasing during the last 40 years [1-3]. It is the most common chronic respiratory disease in children in the developed countries [4] and its prevalence is steadily increasing in the developing world [5]. 

Asthma is a chronic inflammatory airway disease with several distinct phenotypes, characterized by different immunopathological pathways, clinical presentation, severity of the disease and response to treatment [6-11]. The phenotypes of asthma include eosinophilic, neutrophilic, mixed granulocytic and paucigranulocytic asthma [7]. 

Patients with eosinophilic asthma have an eosinophil count ≥3% [12-14], whereas patients with neutrophilic asthma have elevated sputum neutrophil count between ≥61% [14] and ≥64% [15], depending on the study. Mixed granulocytic phenotype is characterized by increase in both eosinophils (>3%) and neutrophils (>61% or >64%) [15]. Paucigranuocytic phenotype embraces patients with very few eosinophils (<3%) and neutrophils (<61% or <64%) in induced sputum [15]. Non-eosinophilic asthma is the term used to classify patients with low eosinophil numbers (<3%), which include neutrophilic asthma and paucigranulocytic phenotype [7]. 

Approximately 3.6-10% of patients with asthma have severe refractory disease, which is uncontrolled despite treatment with high-dose Inhaled Corticosteroids (ICS) and Long-Acting β2-Agonists (LABA) [16-19]. Neutrophilic asthma is the most common phenotype in adult patients presenting with acute severe asthma, whereas eosinophilic asthma is the most common phenotype in children with acute severe asthma [20]. However, paucigranulocytic asthma is the most common phenotype in both adults and children in patients with stable asthma [21]. 

The definition, evaluation and treatment of severe refractory asthma has been refined and continues to be updated [19,22-24]. The American Thoracic Society (ATS) guidelines on the definition of severe refractory asthma is based on two major and seven minor criteria for the diagnosis of severe refractory asthma [22]. The criteria for established diagnosis of refractory asthma include fulfilling one, or both major criteria and at least two minor criteria [21]. The ATS criteria for the diagnosis of severe refractory asthma are given in table 1. 

Major criteria

 

Treatment with continuous or near continuous (>50% of the year) oral corticosteroids

Need for treatment with high-dose inhaled corticosteroids.

Minor criteria

 

Need for additional daily treatment with controller medication (long-acting β2-agonist, leukotriene receptor antagonist, theophylline)

Asthma symptoms needing short-acting β2-agonists use on a daily or near daily basis

Persistent airway obstruction (FEV1 <80% predicted, diurnal peak flow variability <20% predicted)

One or more urgent care visit for asthma

Three or more oral steroid bursts per year

Prompt deterioration with >25% reduction in oral or inhaled corticosteroids

Near fatal asthma event in the past.

Diagnosis

One major criterion plus two minor criteria; other diseases should have been excluded, exacerbating factors treated and patient is general adherent.

Table 1: American thoracic society criteria for severe/refractory asthma.

CLINICAL FEATURES OF NEUTROPHILIC ASTHMA

Neutrophilic asthma is an adult-onset disease which usually starts after 12 years. It is the most common phenotype in adult patients presenting with acute severe asthma compared with eosinophils asthma [20]. However, eosinophilic asthma is the most common phenotype in children presenting with acute severe asthma [20], but paucigranulocytic phenotype is the most common phenotype in both adults and children with stable asthma [21]. 

Neutrophilic phenotype is characterized by severe persistent asthma [25-28], with frequent exacerbations, although the exacerbations are not as severe as those encountered in patients with eosinophilic asthma [29,30]. Patients with neutrophilic asthma have frequent urgent visits to emergence rooms, hospitalization and intubation [31]. This phenotype of asthma has been associated with sudden-onset fatal asthma in about 23% of the patients [32]. Furthermore, patients with severe bronchial neutrophilia are more likely to be admitted to hospital for noninfectious status asthmaticus [33]. 

Neutrophilic asthma is worse at night with frequent nocturnal attacks [34]. Martin et al. [34], found a greater than three-fold increase in the number of granulocytes in Bronchoalveolar (BAL) fluid at 04:00 hr compared with 16:00 hr. Patients with neutrophilic asthma may require chronotherapy with intensification of treatment at night or treatment with long-acting anti-asthma agents [35]. Furthermore, neutrophilic asthma is typically associated with a worse quality of life and has a poor prognosis [7,18,19,28,36]. 

Neutrophilic asthma is characterized by a high neutrophil count in induced sputum ranging from 40% to 76% of sputum cells [30,32-34], or a neutrophil count of 500 × 104/ml. Additionally, patients with neutrophilic asthma have less sputum eosinophil count which has been quoted to be between less than 1.9% and 3% by various authors [7,31,37,38]. 

Increased neutrophils in sputum has been associated with severe persistent asthma [7,23,26], fixed airway obstruction [36,39-42], with very low Forced Expired Volume in 1 second (FEV1) [39,40] and post-brochodilator FEV1 [43]. Shaw and collagues [43], have reported that both patients with eosinophilic asthma and neutrophilic asthma had low pre-bronchodilator FEV1, but only patients with neutrophilic asthma had lowest post-bronchodilator FEV1, indicating persistent airflow limitation. Furthermore, patients with neutrophilic asthma are less atopic [42-44] and have less responsiveness to methacholine challenges compared with patients with eosinophilic asthma [41,42,44]. 

Patients with neutrophilic asthma are unresponsive to LABA and high dose ICS [37,45,46] and the newly introduced targeted biologics [16,47]. There are no specific biomarkers for the diagnosis of neutrophilic asthma. Currently, there are specific biomarkers, such as exhaled Nitric Oxide (FeNO), serum periostin and dipetidyl peptidase 4 for phenotyping asthma, which are useful for the diagnosis and targeted treatment of Th2-driven eosinophilic inflammation [8,16,48-50]. The clinical and diagnostic features of neutrophilic asthma are shown in table 2. 

Adult on-set, most cases after 20 years

Less atopic compared with eosinophilic asthma

Less severe exacerbations compared with eosinophilic asthma

Sputum neutrophil count, 40%-64%; eosinophils <2%-3%

Low FeNO <30 ppb

Low serum periostin and dipeptidyl peptidase 4 levels

High hydrogen sulfide levels

Less subepithelial fibrosis compared with eosinophilic asthma

Fixed airflow limitation (low FEV1)

Less responsiveness to methacholine challenge tests

Corticosteroid unresponsiveness

Poor response to biologics

Table 2: Clinical and diagnostic characteristics of neutrophilic asthma.

AIRWAY REMODELING IN NEUTROPHILIC ASTHMA

The pathophysiological feature of neutrophilic asthma is airway hyperresponsiveness and airway remodeling, which is associated with persistent fixed airway obstruction. There is a strong association between neutrophilc airway inflammation and progression of airflow limitation in patients with neurophilic asthma [51]. Airway remodeling and bronchoconstriction in asthma involves structural changes, such as airway smooth muscle hyperplasia and hypertrophy; subepithelial basement membrane thickening and fibrosis; extacellular matrix protein deposition; hypertrophy of the submucous glands, goblet cell hyperplasia; thickening and shedding of the epithelium; and neoangiogenesis (Table 3) [52-60]. Airway smooth muscle hypertrophy, hyperplasia and changes in phenotype of ASM is considered the main factor involved in airway hyper responsiveness [61]. 

Epithelial injury due to allergens, respiratory viral infections, and pollutants

Release of cytokines, chemokines, growth factors, and adhesion molecules

Airway epithelial thickening, shedding, and further release of "alarmin" cytokines

Submucous glands and goblet cell hyperplasia, and mucus hypersecretion

Airway hyperresponsiveness

Subepithelial basement membrane fibrosis, and thickening

Deposition of extracellular matrix proteins

Mast cell infiltration of airway smooth muscle cells

Activation of myofibroblasts, and fibroblasts

Airway smooth muscle hyperplasia, and hypertrophy

Neoangiogenesis, and exaggerated vasodilatation

Airway remodeling

Table 3: Pathophysiological features of airway remodeling in patients with neutrophilic asthma. 

Patients with severe refractory asthma, including the neutrophilic phenotype have airway smooth muscle cell hyperplasia and hypertrophy, especially patients with fixed airflow limitation [56,59-62]. Additionally, ASM hypertrophy and hyperplasia is associated with progressive decline in lung function (low FEV1), persistent asthma and corticosteroid unresponsiveness [63-66]. 

Airway smooth muscle cells are productive and secretory cells, which display plasticity depending on inflammatory milieu. They display pro-inflammatory and immunomodulatory functions by synthesis and expression of a variety of surface molecules, integrins and Toll-like receptors [67,68]. They are capable of secreting a milieu of pro-inflammatory cytokines (IL-1β, IL-8, IL-5, IL-6, IL-8, IL-10 and IL-11); chemokines (eotaxins and Gro-α); growth factors (EGF-1, FGF, PDGF; VEGF and IGF-1); and angiogenic factors (angiogenin and angiopoietin) [58,67,68]. In addition, ASM cells from asthmatic patients have a distinct hyper reactive “primed” phenotype, which is characterized by increased release of pro-inflammatory cytokines, chemokines and growth factors [56,68]. These mediators act in an autocine or paracrine fashion and amplify airway inflammation, AHR and remodeling [56]. 

Another feature of airway remodeling in asthma is deposition of Extracellular Matrix (ECP) protein in the reticular basement region, lamina propria and submucosa, which contributes to airway wall thickening and airflow obstruction. It is also accompanied by increase in the numbers of airway resident cells, such as fibroblasts, myofibroblasts and ASM cells [62,67-69]. There is an increase in ECM deposition around ASM cells and within the muscle bundle in patients with asthma [66,67]. The ECM proteins contain higher amount of collagen I, III, V, fibronectin, tenascin, glycosaminoglycans, proteoglycans and elastic fibers [66,67] and the composition of the proteins is different compared with non-asthmatic subjects [70]. Extracellular matrix proteins and mediators released by mast cells, T lymphocytes and ASM cells may modulate ASM phenotype and functions, including proliferation, migration and contraction [71,72]. 

Severe refractory asthma is histopathologically characterized by thickening and fibrosis of the Subepithelial Reticular Membrane (SRM) in both adults and children with asthma [69,73-77]. Payne et al. [77] have demonstrated that SRM thickening is already present in children with difficult asthma and to a similar extent to that seen in adults with asthma. In addition they found that SRM thickening is not associated with age, symptom duration, lung function, or concurrent eosinophilic airway inflammation [77]. Thus, SRM thickening and fibrosis can occur in different phenotypes of severe refractory asthma. Thickening of the subepithelial reticular basement membrane is due to increased deposition of collagen I and III, tenascin and fibronectin, versican, laminin and lumican [73,74], produced mainly by fibroblasts [75] and to a less extent myofibroblasts [76]. The increase in subepithelial basement membrane thickness has been shown to correlate with ASM hypertrophy [76] and the severity of airflow limitation [78]. The thickening in SRM may contribute to unesponsiveness to corticosteroids [79]. 

The airway epithelium provide a physical protective barrier against inhaled micro-organisms, allergens and pollutants. Severe asthma, including the neutrophilic asthma is characterized by epithelial thickening, shedding, destruction of ciliated cells, goblet cell, exposure of neuronal terminals, secretion of ‘alarmin’ cytokines (Il-25, IL-33, TSLP) and upregulation of growth factors [80-82]. The extent of epithelial injury parallels with increasing AHR and severity of the disease [83,84]. 

There is an increase in numbers and branching of blood vessels in the airways of patients with asthma due to neoangiogenesis [58,85-87], mediated by overexpression of Vascular Endothelial Growth Factor (VEGF), angiopoietin and angiogenin [88,89]. Increased release of pro-inflammatory mediators in the airways may lead to vasodilatation and edema which may perpetuate airway narrowing. Neoascularization has been shown to correlate with airflow limitation and bronchial hyperresponsiveness [89-91], hence contributing to severity of asthma. The pathophysiological features of airway remodeling in neutrophilic asthma are summarized in table 3.

TREATMENT OF SEVERE NEUTROPHILIC ASTHMA

Severe refractory asthma has classically been treated using stepwise guidelines of increasing drugs and dosages depending on the severity of the disease, such as the Global Initiative for Asthma (GINA) guidelines [1]. The GINA strategy recommends intensification of the treatment according to the severity of the disease, based on the treatment required to control and reduce symptoms and exacerbations. The GINA classic guidelines has five levels of treatments constituting increasing treatment according to severity. Steps 1 to 3 are classified as mild-moderate asthma and steps 4 and 5 include patients with moderate-severe disease. The current paradigm recommends use of relief SABA and ICS/LABA as needed at step 1. The step-wise guidelines recommend treatment with ICS at step 2, followed by increasing the dosage of ICS up to 800 μg/day and adding a long-acting β2-agonist to achieve control at step 3. In patients with severe asthma, steps 4 and 5, the dosage of ICS is increased up to 2000 μg/day and therapeutic alternatives, such as Leukotriene Receptor Antagonists (LTRA), slow-release theophyllines, or Long-Acting Muscarinic Antagonist (LAMA) are added to the regimen [1]. Table 4 lists the drugs used to treat asthma. 

Inhaled β2-agonist

Short acting (salbutamol, levalbuterol, terbutaline, pirbuterol)

Long-acting (salmeterol, formeterol)

New long-acting (indacaterol, olodaterol, vilanterol)

Combination of LABA and inhaled corticosteroids

Salmetrol and fluticasone (Advair Diskus)

Formetorol and budesonide (Symbicort)

Vilanterol and fluticasone

Triple combo (Vilanterol, fluticosone and umeclidium)

Cromones

Cromlyn sodium, nedocromil sodium

Inhaled anti-cholinergics

Short-acting (ipratropium bromide)

Long-acting (oxitropium bromide, tiotropium bromide)

New long-acting (umeclidium bromide, glycopyrrolate)

Corticosteroids

Betamethasone dipropionate

Budenoside, fluticasone, flunisolone

Ciclesonide, mometasone

Oral methylxanthines

Rapid release theophyllines

Sustained release theophyllines (Theo-24, Theocron, Uniphyl)

Phosphodiesterase (PDE)-4 inhibitor (roflumilast)

Leukotriene receptor antagonists

Montelukast, pranlukast

Cinalukast, zafirlukast

5-lipoxygenase inhibitors

Zileuton

Novel therapies

Anti-TNF therapy, e.g., infliximab, etanercept

Prostaglandin D2 receptor antagonists, e.g., fevipiprant, setipiprant

Protein kinase c-kit, Lyn, and Fyn inhibitors, e.g., mastinib, imatinib

Table 4: Standard drugs used for the treatment of asthma. 

Despite treatment according to guidelines, monitoring adherence and adequate inhaler technique, a significant proportion of asthma patients do not achieve adequate control of asthma symptoms with the standard care treatment [1,92]. Between 49% and 53% of adults receiving treatment adequately have poorly controlled asthma [93,94] and up to 64% of adolescent patients have asthma that is inadequately controlled by the currently available therapies [95]. Table 4 shows the list of drugs available for the treatment of asthma. 

Neutrophilic asthma is a difficult to treat phenotype, because it is unresponsive to high-dose ICS and LABA [37,45,46] and to the current anti-interleukin antagonists targeted against eosinophilic asthma [96-100]. 

Treatment of neutrophilic asthma requires novel anti-inflammatory agents and therapeutic strategies targeted against airway smooth muscle hypertrophy and airway remodeling, such as phosphodiesterase 4 inhibitors, macrolide antibiotics and bronchial thermoplasty.

BIOLOGICS FOR NEUTROPHILIC ASTHMA

Interleukin-17 and its associated cytokines, IL-17F, IL-23, IL-22 and IL-8 play a key role in the pathogenesis of neutrophilic asthma [101]. However, most clinical trials using biologics targeted against IL-17, IL-8 and IL-23 have not been very encouraging. Busse at al. [102] in a randomized, placebo-controlled phase IIa trial of brodalumab, a monoclonal antibody against IL-17 receptor, in patients with moderate-to-severe asthma, reported that, brodalumab did not result in any statistically significant benefit in terms of ACQ scores, FEV1, or use of rescue Short-Acting β-Agonists (SABA). 

Blockade of IL-8 receptor CXCR2 also produced temperate results. Nair et al. [103] in a randomized, double-blind, placebo-controlled trial of SCH52713, a selective CXCR2 antagonist in asthmatic patients, noted significant reduction in sputum and blood eosinophil counts. SCH527123 was associated with significantly less exacerbations, but no improvement in lung function (FEV1). In a larger, multicenter, dose finding trial, O’Byrne et al. [104] investigated AZD5069, a selective CXCR2 antagonist, in patients with uncontrolled asthma receiving medium-to-high dose ICS and LABA. In their clinical trial, they did not find significant benefits in the rates of exacerbations, ACQ-5 and FEV1. Thus, combined blockade of CXCR2 and CXCR1 may be more effective approach in targeting the neutrophil chemoattarctant IL-8 [105]. Risankizumab a humanised IgG monoclonal antibody that target the p19 subunit of IL-23 is in phase 2 clinical trials [106]. Brodalumab (Siliq), secukinumab (Cosenyx) and risankizumab (Skyrizi) have been approved in several countries and are excellent drugs for the treatment of plaque psoriasis. Table 5 list the interleukin antagonists for the treatment of eosinophil and neutrophilic asthma. 

Agent

Target

Indication

Stage of Development

Omalizumab

IgE

EA

Marketed 2003

Mepolizumab

IL-5

EA

Marketed 2015

Reslizumab

IL-5

EA

Marketed 2016

Benralizumab

IL-5R

EA

Marketed 2017

Dupilumab

 IL-4?/IL-13

EA

Marketed 2018

Tezepelumab

TSLP

EA

Marketed 2018

Pitrakinra

IL-4? /IL-13

EA

II

Lebrikizumab

IL-13

EA

III

Tralokinumab

IL-13

EA

III

Fezakinumab

IL-22

EA

II

Brodalumab

IL-17RA

NA

II

Secukinumab

IL-17A

NA

II

Risankizumab

IL-23

NA

II

Table 5: Monoclonal antibodies, and interleukin antagonists and their target.

Abbreviations: EA: Eosinophilic Asthma, NA: Neutrophilic Asthma; IL: Interleukin; TSLP: Thymic Stromal Lymphopoietin. Brodalumab, secukinumab and risankizumab are approved for the treatment of plaque psoriasis. 

LONG-ACTING PHOSPHODIESTERASE 4 INHIBITORS

Non-selective Phosphodiesterase (PDE) inhibitors, such as theophylline have been used for the treatment of asthma and COPD for several decades. PDE enzymes metabolize the second intracellular messengers, including Adenosine Monophosphate (cAMP) and cyclic Guanosine Phosphate (cGMP), which play important roles in intracellular signaling in the regulation of multiple cellular metabolisms [107,108]. The PDE superfamily of enzymes contains 11 gene families (PDE1 to PDE11), most of which contain several PDE genes [107]. The PDE4 isoenzymes are encoded by a family of four different genes (PDE4A-4D), which specifically hydrolyze the 3’5’ phosphodiester bond of cAMP to yield 5’Adenosine Monophosphate (5’-AMP) [10,109]. PDE4B isoenzyme is associated with bronchodilatation and anti-inflammatory effects, while PDE4D is associated with gastrointestinal side effects, such as nausea and vomiting, due to its high presence in the vomiting center in the brain [110,111]. 

Inhibition of PDE-4 leads to increase in the intracellular levels of cAMP and subsequent modulation of inflammatory responses and maintenance of the immune balance [112]. Therefore, PDE4 inhibitors are effective therapeutic strategy for the treatment of inflammatory respiratory diseases, characterized by airway remodeling and bronchoconstriction. Increase in the levels of cAMP can activate downstream phosphorylation pathways [113], which leads to relaxation of airway smooth muscle cells, bronchodilatation; and suppression of airway inflammation. 

PDE4 is the most studied subfamily and pharmacological research has wielded several pharmacological agents for the treatment of many chronic inflammatory diseases, such as asthma, Chronic Obstructive Pulmonary Disease (COPD), [113-115] psoriasis [116], atopic dermatitis [117], inflammatory bowel disease [118] and neuropsychiatric disorders [119]. 

Patient with inflammatory diseases have higher expression of PDE4 than healthy individuals [120]. Inhibition of PDE4 results in increase in intracellular cAMP and subsequent activation of PKA, cyclic nucleotide-gated ion channels and Epac1/2. These signaling pathways are involved in the regulation of pro-inflammatory and anti-inflammatory cytokine synthesis [121]. In vitro inhibition of PDE4 has been shown to decrease expression of cell surface markers in many inflammatory cells, such as T cells and decreased release of cytokines, such as TNF-α, IL-1β and IL-10 in many types of cells [122,123]. In vivo inhibition of PDE4 also leads to a broad spectrum of effects, such as inhibition of cell trafficking and cytokine and chemokine release from inflammatory cells, such as neutrophils, eosinophils, macrophages and T cells [124]. In addition, PDE inhibitors promote apoptosis of these cells [122,125]. Animal studies have shown that roflumilast reduced accumulation of neutrophils in Bronchoalveolar Lavage (BAL) fluid following exposure of cigarette smoke in guinea pig and mice [126,127]. Cortijo, et al. [128], have also reported that roflumilast prevented bleomycin-induced infiltration of neutrophils and macrophages in mice lungs. 

In vitro study has shown that roflumilast and its active metabolite roflumilast N-oxide inhibited neutrophil secretion of IL-8, Leukotriene B4 (LTB4), Matrix Metalloproteinase-9 (MMP-9) and neutrophil elastase [122,129]. PDE4 inhibitors have also been reported to inhibit Interleukin-4 (IL-4) and IL-13 generation by human basophils [130]. Furthermore, roflumilast and roflumilast N-oxide reduced liposaccharide-induced release of chemokines (CCL2, CCL3, CCL4 and CXL10) and TNF-α from human lung macrophages in a dose-dependent fashion [131]. 

Roflumilast has been reported to suppress secretion of TNF-α from epithelial cells and exerts antinflammatory and immunomodulatory effects [132]. PDE4 inhibitors, such as cilomilast and roflumilast have been shown to decrease MUC5AC expression induced by Epidermal Growth Factor (EGF) [133] and roflumilast has been reported to improve ciliary function and mucociliary clearance [134]. In vitro study have shown that roflumilast antagonized profibrotic activity of fibroblasts stimulated by TGF-β [135,136]. Hence, PDE4 inhibitors have the potential to prevent progressive subepithelial basement membrane fibrosis and pulmonary fibrosis [135]. 

Phosphodiesterase 4 inhibitors are appropriate as add-on therapy for patients with neutrophilic asthma, because they suppress immune cell trafficking, activation and degranulation. They also suppress the release of cytokines, chemokines and growth factors which promote subepithelial membrane fibrosis, ASM cell proliferation, ASM hypertrophy and airway remodeling [122]. Long-acting selective PDE4 inhibitors, such as roflumilast have been shown to significantly reduce airway hyper responsiveness [137], which is a key feature of neutrophic asthma. Oral roflumilast 500 μg morning or evening has been shown be beneficial as add-on treatment for fixed airflow limitation in patients with increased ASM mass, AHR and airway remodeling [138]. Similarly, clinical trials have documented that roflumilast improves symptom control, exacerbations, lung function and quality of life [139-141]. 

Roflumilast helps improve efficacy of other anti-inflammatory agents and bronchodilators, such as corticosteroids, LABA and LTRA. Roflumilast and its active derivative roflumilast N-oxide have been shown to enhance activity of the glucocorticoid receptor activity and glucocorticoid-dependent gene transcription in peripheral blood mononuclear cell of asthmatic patients compared with control [139]. The combination of roflumilast and fluticasone significantly reduced AHR compared with roflumilast dosage alone [138]. 

Roflumilast can be used as an add-on treatment to ICS and LABA and/or LTRA therapies [141] and is beneficial in reducing gradual decline in lung function associated with increase in ASM hypertrophy and airway remodeling. The GINA guidelines [1], recommends addition of slow-release theophyllines, including the long-acting PDE-4 inhibitor roflumilast for the treatment of asthma at step 3. Roflumilast (Daliresp) is the only approved long-acting selective PGE4-inhibitor for the treatment COPD and asthma and was approved for these indications by the European Union (EU) in 2010 and in the USA in 2011. It has better selectivity and tolerance.

MACROLIDE ANTIBIOTICS

Patients with neutrophilic asthma are unresponsive to corticosteroids [142], anti-IgE and anti-interleukin Monoclonal Antibody (mAb) therapies targeting eosinophilic asthma. Thus, there is need for alternative anti-inflammatory therapies for patients suffering from neutrophilic asthma [143]. 

Macrolides have a macrocytic lactone ring, whose size and features have been modified from the 14 carbon structure of erythromycin in order to develop newer agents such as clarithromycin and roxithromycin, or the 15 carbon structure to produce azithromycin [144]. They are derived from the product of the microbial order Actinomycetales (Saccharopolyspora erythrae, formerly Streptomyces erythrae). 

Macrolide antibiotics, such as Erythromycin (ERM), Azithromycin (AZM), Clarithromycin (CAM) and Roxithromycin (RXM) and new ketolide antibiotic telithromycin have antibacterial, antiviral and anti-inflammatory effects. They are mostly used to treat infections caused by Chlamydophila pneumonia and Mycoplasma pneumonia, especially respiratory and genitourinary infections. They now constitute part of guideline-recommended therapy in community acquired pneumonia. Azithromycin and clarithromycin are also used to treat legionellosis. 

Chlamydophila pnuemoniae and Mycoplasma pneumoniae may play an important role in the pathogenesis of neutrophilic asthma [145]. Moreover, atopic subjects have increase frequencies of detection of C pneumonia in nasal aspirate sampling, independent of symptoms compared with healthy volunteers [146]. Similarly, asthmatic patients are more likely to harbor M pneumonia in their airways [147]. There is cumulative evidence suggesting infections with Chamydophila pneumoniae and or Mycoplasma pneumoniae might play a role in the pathogenesis of different phenotypes of asthma, including neutrophilic asthma. Treatment of these atypical bacteria may be gratifying in preventing severe exacerbations associated with these microbes. 

Macrolides and ketolides have been shown to have both in vitro and in vivo anti-inflammatory activity, including suppression of neutrophil inflammation, which makes them relevant to respiratory conditions associated with airway neutrophilia, such as neutrophilic asthma and atypical bacterial infection. 

Treatment with macrolides, such as erythromycin and azithromycin has been shown to reduce neutrophil counts and Bronchoalveolar Lavage (BAL) fluid Interleukin-8 (IL-8) levels in patients with panbronchiolitis [146], bronchiolitis obliterans syndrome (BOS [148,149] and asthma [150]. The clinical improvement in these conditions occur in the absence of active bacterial colonization, thus confirming the imunomodulatory effects of macrolides. 

Several studies have reported that treatment with AZM, CAM and RXM decrease eosinophil and neutrophil numbers, inhibit neutrophil migration and oxidative burst activity and mediator release. Consequently, there is a decrease in the concentrations of neutrophil elastase, metalloproteinase-9, IL-8, IL-6, IL-1β, TNF-α and Eosinophilic Cationic Protein (ECP) [143,150-155].

Simpson et al. [143], have shown that clarithromycin in patients severe refractory asthma reduced neutrophil count and sputum IL-8 levels, although they did not observe any change in lung function or asthma control [143]. The Azithromycin for prevention of exacerbations in severe asthma (AZISAST) randomized, placebo-controlled trial in patients with severe asthma with history of severe exacerbations, despite receiving high-dose ICS and LABA, studied the effect of AZM on asthma exacerbations [156]. Azithromycin (250 mg daily three times per week) as add-on treatment in patients with nonesonophilic asthma, defined by normal blood eosinophil counts and normal FeNO, resulted in significantly fewer severe exacerbations during 26-week period compared with controls [156]. Azithromycin significantly reduced severe exacerbations and lower respiratory tract infection in non-eosinophilic asthma phenotype by approximately 67% compared to 38% in placebo group. AZM was ineffective in reducing exacerbations in patients with esosinophilic asthma, who tended to have more exacerbations on AZM treatment [156]. 

The second randomized double-blinded, placebo-controlled trial (AMAZES) compared add-on azithromycin (500 mg three times per week) with placebo for 48 weeks in patients with symptomatic asthma despite medium-to-high dose ICS and LABA [157]. Treatment with add-on azithromycin significantly reduced the incidence of medium and severe exacerbation by 1.07 versus 1.86 per person-year, for AZM and placebo, respectively. AZM treatment was also associated with an improvement in Asthma Quality of Life Questionnaire (AQLQ) scores in both groups of patients with eosinophilic and noneosinophilic asthma phenotypes [157]. 

The Telithromycin, Chlamydophila and Asthma (TELICAST) multicenter, randomized, double-blind, placebo-controlled study in 278 patients with moderate-to-severe asthma reported significantly greater improvement in symptoms and lung function in patients receiving telithromycin, 800 mg once daily, for 10 days compared with placebo [158]. Patients receiving telithromycin had improvement in symptoms from exacerbation at the end of treatment by 51% versus 29% in the placebo treated patients. The FEV1 improved by 0.63 L in telithromycin-treated patients versus 0.29 L in placebo-treated [158]. 

The Azithromycin Against Placebo in Exacerbations of Asthma (AZALEA) study investigated the effectiveness of azithromycin treatment as add-on to standard therapy for adult patients with with exacerbation [159]. In the AZALEA clinical trial, addition of azithromycin 500 mg daily for 3 days to the standard treatment resulted in no statistically significant clinical improvement, including symptoms and quality of life scores and FEV1 [159]. 

This large trial in the UK had challenges in the recruitment of subjects, because there was widespread use of antibiotics in the 31 centers enrolled for the study. The study was therefore underpowered because a large number of patients (2044) were excluded because they were already taking antibiotics for their exacerbation [159]. 

It is possible that the population randomized was not representative of the larger population, because more than 2000 other patients were excluded from the study for other reasons [159]. 

From the above studies, different macrolides including the dosages of the specific drugs may influence the immunomodulatory and immunosuppressive response to macrolide antibiotics. Selection and exclusion of asthmatic patients, including phenotypes may also influence outcome of the effects of different macrocytic lactone ring macrolides, including the 16-membered (spiramycin, jasamycin and midecamycin) [144]. 

Macrolides and ketolides have proven anti-inflammatory and immunomodulatory effects via activation or inhibition of immunopathological pathways. Macrolides have the ability to alter intracellular signaling, particularly through inhibition of TNF-kB activation and expression of activator protein-1 [160,161]. 

Azithomycin has been reported to significantly reduced Nuclear Factor-kB (NF-kB) expression, Tumor Necrosis Factor Alpha (TNF-α) RNA levels and TNF-α secretion in a CF-derived airway epithelial cell line [162]. Treatment of Bronchopulmonary Dysplasia (BPD) with AZM has been shown to suppress TNF-α-stimulated NF-kB activation in tracheal aspirate cells from premature infants with the reduction in Interleukin-6 (IL-6) and IL-8 secretion compared to control levels [163]. 

Macrolides have several immunomodulatory effects in numerous cells via modulation of intracellular signaling of multiple pathways, including intracellular Ca2+ regulation, Mitogen-Activated Protein Kinase (MAPK) signaling pathways and modulation of transcription factor function [164]. The anti-inflammatory effects of macrolides include reduction of cytokine expression, reduction of adhesion molecule expression on inflammatory cells, reduction of chemical mediator release and Reactive Oxygen Species (ROS) and increased apoptosis and efferocytosis [164]. Shinkai, et al. [165] have suggested that macrolides might suppress IL-8 and extracellular signal-regulated kinase related to neutrophil chemotaxis and migration. 

Neutrophilic asthma is nonresponsive to corticosteroids. Corticosteroid-resistance is associated with airway hyperresposniveness and decreased Histone Deactylase 2 (HDAC2) activity and expression [166]. HDAC2 has been shown to inhibit inflammatory protein coding genes such as granulocyte macrophage colony stimulating factor or cyclooxygenase 2, promoted by IL-1β, TNF-α and NFkB kinase [166]. Macrolides reverse corticosteroid insensitivity by restoring the HDAC activity, via inhibiting Phosphoinositol 3 Kinase (PI3K) pathway [167] and by attenuating TNF-α and IL-17 immune responses [168]. 

Mucus hypersecretion and plugging is one of the characteristic features of COPD and severe refractory asthma, including the neutrophilic phenotype. Macrolides have also been shown to reduce mucus secretion and ion transport in the airways and increase mucociliary function of epithelial cells [169]. 

Macrolides such as AZM and CAM and telithromycin are suitable as add-on treatment to ICS and LABA in patients with neutrophilic asthma. Clarithromycin has been shown as an effective add-on therapy in prednisone-dependent patients with asthma [170]. However, the use of long-term macrolides particularly in patients on high-dose ICS carries its own risks, such as adverse reactions, including diarrhoea, cholestasis, tinnitus and torsades de pointes ventricular tachycardia. It may also a promote antibiotic resistance against macrolides, ketolides, tetracyclines and other related antibiotics [157,171-173].

BRONCHIAL THERMOPLASTY

Corticosteroids are the mainstay of asthma treatment; however, they do not suppress ASM hyperplasia and hypertrophy which is one of the histopathological features of severe asthma responsible for airway hyperresponsiveness [174]. One of the strategies for the treatment of severe asthma is to target airway smooth muscle hypertrophy, using bronchial thermoplasty, a non-pharmacological procedure aimed at reducing airway smooth muscle mass [175,176]. 

Bronchial Thermoplasty (BT) is a bronchoscopic treatment for subjects aged 18 and above with severe persistent asthma not responding to high-dose ICS and LABA. Selection and preparation of patients for BT is very important and the procedure should be performed by experienced pulmonologists or bronchoscopists [176-179]. 

Patients for BT should be in an optimal stable condition, without any asthma exacerbation or respiratory infection for at least 2 weeks. In addition to their standard medical treatment for severe asthma they should be pre-treated with prednisolone 50 mg/day for 3 days before BT, on the day of BT and the day after bronchial thermoplasty [179]. Patients with low FEV1 <80% should have the procedure postponed until their lung function improves. Before the procedure, all patients should be pre-treated with nebulized salbutamol and/or ipratropium bromide [179]. 

Bronchial thermoplasty is performed under moderate-to-deep sedation or general anesthesia [176-179]. At bronchoscopy a special catheter with a basket design is inserted through the instrument channel which delivers radiofrequency energy generated by a Radiofrequency (RF) generator (AlairTM Bronchial Thermoplasty System) to the airway wall [180,181]. 

The AlairTM catheter electrode delivers targeted radiofrequency energy to bronchial airway wall and results in reduction of the hypertrophied airway smooth muscle mass which is responsible for bronchoconstriction [178-186]. The procedure also decreases subepithelial fibrosis, extracellular matrix, submucous glands, airway nerve endings, epithelial cells and neuroendocrine cells [176,178,179,185,187]. Bronchial thermoplasty may lead to functional denervation, thus, reducing sensory neural axonal reflex bronchoconstriction [187]. 

Bronchial thermoplasty is given over three bronchoscopy sessions at approximately 3-week intervals, one for each lower lobe and one for both upper lobes [178,179,182]. Radiofrequency electrical energy delivered by a Radiofrequency (RF) generator (AlairTM Bronchial Thermoplasty system) is applied to the airways distal to the mainstem bronchi between 3 and 10 mm in diameter throughout the tracheobronchial tree, except the right middle lobe [179,182,183,186]. In practice, 40-70 RF activations are delivered in the lower lobes and between 50 and 100 activations in the upper lobes combined depending on the patient’s size and airway caliber [179]. Traditionally, the Right Middle Lobe (RML) is typically avoided due to concerns about the right middle lobe syndrome [179]. The right is usually not treated because it has a narrow, more horizontal orifice and RF energy could lead to post-procedure inflammation, obstruction and atelectasis of the right middle lobe (syndrome). Otherwise, the lingula is treated. 

The AlairTM Bronchial Thermoplasty System (Boston Scientific, Natick, MA, USA) uses continuous feedback to tightly control the degree of tissue heating to avoid bronchial perforation, scorching and stenosis [179,180,186]. The AlairTM catheter delivers radiofrequency energy in order to warm the airway wall to a targeted temperature of 65°C, which reduces the ASM mass by approximately 50% after 3-6 weeks after the procedure [179, 182,186]. 

Patients should be screened adequately to confirm a correct diagnosis of asthma (including phenotyping), verify the criteria for severe refractory asthma despite adherence to appropriate pharmacological and non-pharmacological therapies, review and address co-morbidities that could affect asthma control, such as obesity, gastroesophageal refux disease, post-nasal drip and obstructive sleep apnea [176]. 

Bronchial thermoplasty requires proper preparation and management of patients pre- and post-thermoplasty. It also requires identification of the right patients, implementation of proper BT technique and intense post-procedural care and follow-up [176]. The procedure should be performed meticulously to avoid bronchoconstriction, airway edema and bleeding. Minor radiological features occur following BT and a chest X-ray should be performed after the procedure. 

Bronchial thermoplasty is a safe and effective procedure, however, it is associated with a short-term increase in asthma-related symptoms such as cough and sputum production, exacerbations and hospital admissions for asthma during the treatment phase [176,179,188]. Occasionally, bronchiectasis [189], atelectasis [190] and rarely pneumothorax and lung cysts [191], have been observed as complications following the bronchial thermoplasty. One case report describes hemoptysis associated with bronchial nodule which resolved by the third session of bronchial thermoplasty [192]. Table 6 lists the complications of bronchial thermoplasty. 

Worsening of asthma control

Hospital re-admissions

Severe exacerbations

Cough with sputum production

Hemoptysis

Acute CT peribronchial consolidation

Nasopharyngitis

Pulmonary infection

Lung abscess

Central bronchiectasis

Upper lobe atelectasis

Collapse of airway by mucous plugging

Pulmonary cysts and pneumothorax

Table 6: Complications of bronchial thermoplasty 

Several randomized controlled trials and prospective multicenter studies in Australia, Canada, France, Japan, Netherland, Spain, UK and USA in patients with severe persistent asthma have documented improvement in asthma control, fewer exacerbations and hospitalization and better quality of life score which persist up to 5 years following bronchial thermoplasty [178,179,181,191-195]. 

The first randomized unblinded clinical trial (Asthma Intervention Research [AIR] 1) In 112 patients with moderate-to-severe asthma was done by Cox and colleagues [196]. Patients treated with BT showed a significant reduction in mild exacerbations as compared with baseline, whereas there were no changes in the frequency of exacerbations in the control group. Additionally, there was a significant improvement in asthma control assessed by the Asthma Control Questionnaire and quality of life assessed by the Asthma Quality of Life Questionnaire in the BT-treated patients compared with controls [196]. However, there were no differences in FEV1 or airway hyperresponsiveness (defined as a provocative concentration of methacholine required to lower the FEV1 by 20% (PC20) or less than 8 mg/mL [196]. 

The second randomized clinical unblinded trial was the Research in Severe Asthma (RISA) trial [197]. This trial which studied 32 patients with severe asthma for the safety and efficacy of BT, reported a significant improvement in the asthma control, quality of life, rescue medication use and pre-bronchodilator FEV1 in BT-treated subjects compared with control. The beneficial effects of BT persisted even after reduction in the dosages of ICS and OCS [197]. 

The largest randomized double-blind sham-controlled trial was the Asthma Intervention Research 2 (AIR2) trial in 297 patients with severe asthma which compared BT with a sham procedure [198]. The AIR2 trial reported significant improvement in AQLQ scores, reduced frequency of severe exacerbations and decreased emergency department visits and days lost from work or school in the year after bronchial thermoplasty compared with treatment with sham procedure [198]. 

Chupp, et al. [199] compared the outcome of BT after a follow-up of 3 years in 190 PAS2 (Post-FDA Approved Clinical Trial Evaluating Bronchial Thermoplasty in Severe Persistent Asthma) subjects with 190 bronchial thermoplasty-treated subjects in the AIR2 trial at 3 years of follow-up. At year 3 after BT, the percentage of PAS2 subjects with severe exacerbations, emergency department visits and hospitalizations significantly decreased by 45%, 55% and 40% respectively [199], resembling the AIR2 results [198]. The PAS2 study showed similar improvements in asthma control after BT compared with the AIR2 trial despite enrolling subjects who had poorer asthma control [199]. After 3-year follow-up, PAS2 subjects were able to significantly reduce their mean ICS dose to 2070 μg/day, whereas, the AIR2 subjects significantly reduced their mean ICS to 1841 μg/day [199]. Previous observational studies on the effectiveness of bronchial thermoplasty for severe asthma have reported reductions in exacerbations and/or a step-down in treatment in 50-75% of patients undergoing the procedure [192-194,197]. 

A systemic review of the long-term safety of BT in the AIR, RISA and AIR2 trials demonstrated no long-term decline in FEV1, no change in the number of emergency room visits or hospitalization for adverse respiratory events [200]. The reduction in exacerbations seen in the first year after remained stable for up to 5 years [176,201-203] and follow-up CT scans performed on the subgroup of the treated patient cohort demonstrated no evidence of bronchiectasis of bronchial stenosis [203]. Recently, Chaudhuri, et al. [204] have reported that after 10 years post-thermoplasty the AQLQ scores and frequency of severe exacerbation were comparable to those recorded 1 year after bronchial thermoplasty. This suggests that the beneficial effects of bronchial thermoplasty may be sustained for up to 10 years or longer. 

Bronchial thermoplasty has a long-term safety profile and may be considered for patients with predominant chronic airflow obstruction and patients who do not respond to anti-IgE, anti-interleukin biologics, or macrolides [176,205,206]. Patients with neutrophilic phenotype of asthma are suitable candidates for bronchial thermoplasty because they have excessive ASM hypertrophy, hyperplasia and hyper responsiveness. They are also unresponsive to treatment with high-dose ICS, LABA, LTRA and interleukin antagonists targeted against eosinophilic asthma. 

The US Food and Drug Administration (FDA) approved BT in 2010 as a safe procedure indicated for the treatment of severe persist asthma in patients 18 years and older, that is not controlled with high-dose ICS and LABA [180]. It is also approved in several EU countries, Australia, Canada, Japan, UK and USA. 

The GINA guideline recommends bronchial thermoplasty for the treatment of severe corticosteroid-resistant asthma at step 5 [1]. The British guideline on the management of asthma states that bronchial thermoplasty can be considered for the treatment of adult patients (aged 18 and over) with severe asthma who have poorly controlled asthma despite optimal therapy [207].

CONCLUSION

Neutrophilic asthma is a phenotype of asthma that is severe and persistent, with frequent exacerbations and hospitalizations. It is characterized by the presence of high levels of neutrophils in the lungs and airways, fixed airflow obstruction and low FEV1. Histopathologically, it is characterized by ASM hyperplasia and hypertrophy, increase in ECM proteins and subepithelial basement membrane fibrosis, which all lead to fixed airflow limitation. Neutrophilic asthma is unresponsive to high-dose ICS, LABA and LTRA and to interleukin antagonist targeted against eosinophilic asthma. This phenotype of asthma requires specific therapeutic interventions aimed at prevention and reducing airway remodeling, such as novel anti-inflammatory agents, including long-acting PDE 4 inhibitors and macrolide antibiotics; and reduction of ASM mass which is the main cause of severe bronchoconstriction, by bronchial thermoplasty.

CONFLICTS OF INTEREST

The author reports no conflicts of interest in this manuscript.

REFERENCES

  1. Global Initiative for Asthma (2018) Global strategy for asthma management and prevention. Global Initiative for Asthma, Kolding, Denmark.
  2. Global Asthma Network (2014) The global asthma report. Global Asthma Network, Auckland, New Zealand.
  3. Masoli M, Fabian D, Holt S, Beasley R (2004) The global burden of asthma: Executive summary of the GINA Dissemination Committee report. Allergy 59: 469-478.
  4. Asher MI, Montefort S, Bjorksten B (2006) Worldwide time trends in the prevalence of symptoms of asthma, allergic rhinoconjunctivitis and eczema in childhood: ISAAC Phase One and three repeat multi-country cross-sectional surveys. Lancet 368: 733-743.
  5. Behbehani N, Abul A, Syabbalo NC, Azeem A (2000) Prevalence of asthma, allergic rhinitis and eczema in 13-to-14-year-old children in Kuwait: An ISAAC study. Ann Allergy Asthma immunol 85: 58-63.
  6. Wenzel SE, Schwartz LB, Langmack EL, Halliday JL, Trudeau JB, et al. (1999) Evidence that severe asthma can be divided pathologically into two inflammatory subtypes with distinct physiologic and clinical characteristics. Am J Respir Crit Care Med 160: 1001-1008.
  7. Simpson JL, Scott R, Boyle MJ, Gibson PG (2006) Inflammatory subtypes in asthma: Assessment and identification using induced sputum. Respirology 11: 54-61.
  8. Chung KF (2016) Asthma phenotyping: A necessity for improved therapeutic precision and new targeted therapies. J Intern Med 279: 192-204.
  9. Anderson GP (2008) Endotypying asthma: New insights into key pathogenic mechanism in a heterogenous disease. Lancet 372: 1107-1119.
  10. Wenzel SE (2012) Asthma phenotypes: The evolution from clinical to molecular approach. Nat Med 18: 716-725.
  11. Pavlidis S, Takahashi K, Kwang FNK, Xie J, Hoda U, et al. (2019) “T2-high” in severe asthma relate to blood eosinophil, exhaled nitric oxide and serum periostin. Eur Respir J 53: 1800938
  12. Aleman F, Lim HF, Nair P (2016) Eosinophilic endotype of asthma. Immunol Allergy North Am
    36: 559-568.
  13. Taylor SL, Leong LEX, Choo JM, Wesselingh S, Yang IA, et al. (2018) Inflammatory phenotypes in patients with severe asthma are associated with distinct airway microbiology. J Allergy Clin Imnunol 141: 94-103.
  14. Arron JR, Choy DF, Scheerens H, Mathews JG (2013) Noninvasive biomarkers that predict treatment benefit from biologic therapies in asthma. Ann Am Thorac Soc 10: 206-213.
  15. Svenningsen S, Nair P (2017) Asthma endotypes and an overview of targeted therapy for asthma. Front Med (Lausanne) 4:158.
  16. Thomson N (2016) Novel approaches to the management of noneosinophilic asthma. Ther Adv Respir Dis 10: 211-234.
  17. Hekking PP, Wener RR, Amelink M, Zwinderman AH, Bouvy ML, et al. (2015) The prevalence of severe refractory asthma. J Allergy Clin Immunol 135: 896-902.
  18. Moore WC, Meyer DA, Wenzel SE, Teague WG, Li H, et al. (2010) Identification of asthma phenotypes using cluster analysis in the severe asthma research program. Am J Respir Crit Care Med 181: 315-323.
  19. Chung KF, Wenzel SE, Brozek JL, Bush A, Castro M, et al. (2014) International ERS/ATS guidelines on definition, evaluation and treatment of severe asthma. Eur Respir J 43: 343-373.
  20. Wang F, He XY, Baines KJ, Gunawardhan LP, Simpson JL, et al. (2011) Different phenotypes in adults and children with acute asthma. Eur Respir J 38: 567-574.
  21. Ntontsi P, Loukides S, Bakakos P, Kostikas K, Papatheodorou G, et al. (2016) Clinical, functional and inflammatory characteristics in patients with paucigranulocytic asthma. Eur Respir J 48: 4173.
  22. American Thoracic Society (2000) Proceeding of the ATS workshop on refractory asthma: Current understanding, recommendations and unanswered questions. American Thoracic Society. Am J Respir Crit Care Med 162: 2341-2351.
  23. Bel EH, Sousa A, Fleming I, Bush KF, Versnel J, et al. (2011) Diagnosis and definition of severe asthma: An international consensus statement from the Innovative Medicine Initiative (IMI). Thorax 66: 910-917.
  24. Wener RR, Bel EH (2013) Severe refractory asthma: An update. Eur Respir Rev 22: 227-235.
  25. Jatakanon A, Uasuf C, Maziak W, Lim S, Chung KF, et al. (1999) Neutrophilic inflammation in severe persistent asthma. Am J Respir Crit Care Med 160: 1532-1539.
  26. Moore WC, Meyer DA, Wenzel SE, Teague WG, Li H, et al. (2010) Identification of asthma phenotypes using cluster analysis in the severe asthma program. Am J Respir Crit Care Med 181: 315-323.
  27. Esteban-Gorgojo I, Antolin-Amerigo D, Dominguez-Ortega J, Quirce S (2018) Non-eosinophilic asthma: Current perspective. J Asthma Allergy 11: 267-281.
  28. Moore WC, Hastie AT, Li XN, Li H, Busse WW, et al. (2014) Sputum neutrophil counts are associated with more severe asthma phenotype using cluster analysis. J Allergy Clin Immunol 133: 1557-1563.
  29. Wenzel SE, Schartz LB, Langmark EL, Halliday JL, Trudeau JB, et al. (1999) Evidence that severe asthma can be divided pathologically into two inflammatory subtypes with distinct physiologic and clinical characteristics. Am J Respir Crit Care Med 160: 1001-1008.
  30. Haldar P, Pavord ID (2007) Noneosinophilic asthma: A distinct clinical and pathologic phenotype. J Allergy Clin Immunol 119: 1043-1052.
  31. Fahy JV, Kim KW, Liu J, Boushey HA (1995) Prominent neutrophil inflammation in sputum from subjects with asthma exacerbations. J Allergy Clin Immunol 95: 843-852.
  32. Sur S, Crotty TB, Kephart GM, Hyama BA, Colby TV, et al. (1993) Sudden-onset fatal asthma: A distinct entity with few eosinophils and relatively more neutrophils in the airway mucosa? Am Rev Respir Dis 148:713-719.
  33. Lamblin C, Gosset P, Tillie-Leblond I, Saulner F, Marguette CH, et al. (1998) Bronchial neutrophilia in patients with noninfectious status asthmaticus. Am J Respir Crit Care Med 157: 394-402.
  34. Martin RJ, Cicutto LC, Smith HR (1991) Airway inflammation in nocturnal asthma. Am Rev Respir Dis 143: 351-357.
  35. Syabbalo NC (1999) Chronobiology and chronopathophysiology of nocturnal asthma. Br J Clin Pract 51: 455-462.
  36. Taylor SL, Leong LEX, Choo JM, Wesselingh S, Yang IA, et al. (2018) Inflammatory phenotypes in patients with severe asthma are associated with distinct microbiology. J Allergy Clin Immunol 141: 94-103.
  37. Green RH, Brightling CE, Woltmann G, Parker D, Wardlaw AJ, et al. (2002) Analysis of induced sputum in adults with asthma: Identification of subgroup with isolated neutrophilia and poor response to inhaled corticosteroids. Thorax 57: 875-879.
  38. Harder P, Pavord ID, Shaw DE, Barry MA, Thomas M (2008) Cluster analysis and clinical asthma phenotypes. Am Rev Respir Crit Care Med 178: 218-224.
  39. Ray A, Kolls JK (2017) Neutrophilic inflammation in asthma is associated with disease severity. Trends Immunol 38: 942-954.
  40. Little SA, Macleod KJ, Chalmers GW (2002) Association of forced expired volume with duration and sputum neutrophils in chronic asthma. Am J Med 112: 446-452.
  41. Choi JS, Jang AS, Park JS (2012) Role of neutrophils in persistent airway obstruction due to refractory asthma. Respirology 17: 322-329.
  42. Jayaram L, Pizzichini MM, Cook RJ, Boulet LP, Lamiere C, et al. (2006) Determining asthma treatment by monitoring sputum cell counts. Eur Respir J 27: 483-494.
  43. Shaw DE, Berry MA, Hargadon B, Mckenna S, Sherry MJ, et al. (2007) Association
    between neutrophilic airway inflammation and airflow limitation in adults with asthma. Chest 132: 1871-1875.
  44. Woodruff PG, Khashayar R, Lazarus SC, Janson A, Avila P, et al. (2001) Relationship between airway inflammation, hyperresponsiveness and obstruction in asthma. J Allergy Clin Immunol 108: 753-758.
  45. Wenzel SE, Szefler SJ, Leung DY, Sloan SI, Rex MD, et al. (1997) Bronchoscopic evaluation of severe asthma. Persistent inflammation associated with high-dose glucocorticoids. Am J Respir Crit Care Med 156: 737-743.
  46. Pavord ID, Brightling CE, Woltman G, Wardlaw AL (1999) Non-eosinophilic asthma corticosteroid unresponsive asthma. Lancet 353: 2213-2214.
  47. Syabbalo N (2020) Neutrophilic asthma: A complex phenotype of asthma. J Lung Pulm Respir Res 7: 18-24.
  48. Yan XC, Woodruff PG (2016) Biomarkers in severe asthma. Immunol Allergy North Am 36: 547-557.
  49. Schleich F, Sophie, D, Renaud L (2016) Biomarkers in the management of difficult asthma. Curr Top Med Chem 16: 1561-1573.
  50. Kim H, Ellis AK, Fischer D, Noseworthy M, Olivenstein R, et al. (2017) Asthma biomarkers in the age of biologics. Allergy Asthma Clin Immunol 13: 48.
  51. Choi JS, Jang AS, Park JS, Park SW, Paik SH, et al. (2012) Role of neutrophils in persistent airway obstruction due to refractory asthma. Respirology 17: 322-329.
  52. Ebina M, Takahashi T, Chiba T (1993) Cellular hypertrophy and hyperplasia of airway smooth muscles underlying bronchial asthma. A 3-D morphometric study. Am Rev Respir Dis 148: 720-726.
  53. Niimi A, Matsumoto H, Amitani R, Nakano Y, Mishima M, et al. (2000) Airway wall thickness in asthma assessed by computed tomography. Relation to clinical indices. Am J Respir Crit Care Med 162: 1518-1523.
  54. Munakata M (2006) remodeling and airway smooth muscle in asthma. Allergol Int 55: 235-245.
  55. Girodet PO, Ozier A, Trian T, Marthan R, Berger P, et al. (2011) The pivotal role of airway smooth muscle in asthma pathophysiology. J Allergy 742710.
  56. Ozier A, Allard B, Bara I, Girodet P-O, Trian T, et al. (2011) The pivotal role of airway smooth muscle in asthma pathophysiology. J Allergy 742710. 
  1. Coi S, Hoffman EA, Wenzel SE, Castro M, Fain S, (2017) National Heart, Lung and Blood Institute’s Severe Asthma Research Programme. Quantitative computed tomography imaging-based clustering differentiates asthmatic subgroups into distinct clinical phenotypes. J Allergy Clin Immunol 69: 6749-30146.
  2. Keglowich LF, Borger P (2015) the three A’s in asthma: Airway smooth muscle, airway remodeling & angiogenesis. Open Respir Med 9: 70-80.
  3. Lai HY, Rogers DF (2010) Mucus hypersecretion in asthma: Intracellular signalling pathways as target for pharmacotherapy. Curr Opin Allergy Clin Immunol 10: 67-76.
  4. Lambrecht BN, Hammad H (2012) the airway epithelium in asthma. Nat Med 18: 684-692.
  5. Zuyderduyn S, Sukkar MB, Fust A, Dhaliwal S, Burgess JK (2008) Treating asthma means treating airway smooth muscle cells. Eur Respi J 32: 265-274.
  6. Bara I, Ozier A, J-M Tunon de Lara R, Marthan R, Berger P (2010) Pathophysiology of bronchial smooth muscle remodeling in asthma. Eur Respir J 36: 1174-1184.
  7. James AL, Elliot JG, Carroll ML, Mauad T, Bai TR, et al. (2012) Airway smooth muscle hypertrophy and hyperplasia in asthma. Am J Respir Crit Care Med 11: 145.
  8. Pepe C, Foley S, Shannon J, Lemiere C, Olivenstein R, et al. (2005) Differences in airway remodeling between subjects with severe and moderate asthma. J Allergy Clin Immunol 116: 544-559.
  9. Kaminska M, Foley S, Maghini K, Coxson H, Ghezzo H, et al. (2009) Airway remodeling in subjects with or without chronic persistent airflow obstruction. J Allergy Clin Immunol 124: 45-51.
  10. Roth M, Johnson PR, Borger P, Bihl MP, Rudiger JJ, et al. (2004) Dysfunctional interaction of C/EBPα and the glucocorticoid receptor in asthmatic bronchial smooth-muscle cells. N Engl Med 351: 560-674.
  11. Damera G, Tliba O, Panettieri RA (2009) Airway smooth muscle as an immunomodulatory cell. Pulm Pharmacol Ther 22: 353-359.
  12. Hirst SJ (2003) Regulation of airway smooth muscle cell immunomodulatory function: Role in asthma. Respir Physiol Neurobiol 137: 309-326.
  13. Hough KP, Curtiss ML, Trevo J, Deshane JS, Thannickal VJ, et al. (2020) Airway remodeling in asthma. Front Med.
  14. Araujo BB, Dolhnikoff M, Silva LF, Mauad T, James A, et al. (2008) Extracellular matrix components and regulators in the airway smooth muscle in asthma. Eur Respir J 32: 61-69.
  15. Begueret H, Berger P, Vernejoux JM, Dubuisson L, Marthan R, et.al (2007) Inflammation of bronchial smooth muscle in allergic asthma. Thorax 62: 8-15.
  16. Bai TR, Cooper J, Koelmeyer T, pare DP, Weir TD (2000)The effect of age and duration of disease on airway structure in fatal asthma. Am J Respir Crit Care Med 162: 663-669.
  17. Roche WR, Beasley R, Williams JH, Holgate ST (1989) Subepithelial fibrosis in the bronchi of asthmatics. Lancet 1: 520-524.
  18. Reeves SR, Kolstad T, Lien TY, Elliot M, Ziegler SF, et al. (2014) Asthmatic airway epithelial cells differentially regulate fibroblast expression of extracellular matrix components. J Allergy Clin Immunol 134: 663-670.
  19. Brewster CE, Howarth PH, Djukanovic R, Wilson J, Holgate ST, et al. (1990) Myofibroblasts and subepithelial fibrosis in bronchial asthma. AM J Respir Cell Mol Biol 3: 507-511.
  20. James AL, Maxwell PS, Pearce-Pinto G, Elliot JG, Carroll NG (2002) The relationship of reticular basement membrane thickness to airway remodeling in asthma. Am J Respir Crit Care Med 166: 1590-1595.
  21. Payne DNR, Rogers AV, Adelroth E, Bandi V, Guntupalli KK, et al. (2003) Early thickening of the reticular basement membrane in children with difficult asthma. Am J Respir Crit Care Med 167: 78-82.
  22. Bonsignore MR, Profita M, Gagliardo R, Riccobono L, Chiappara G, et al. (2015) Advances in asthma pathophysiology: Stepping forward from Maurizio Vignola experience. Eur Respir Rev 24: 30-39.
  23. Bourdin A, Kleis S, Chakra M, Vachier I, Paganin F, et al. (2012) Limited short-term steroid responsiveness is associated with thickening of bronchial basement membrane in severe asthma. Chest; 14: 1504-1511.
  24. Carroll N, Elliot J, Morton A, James A (1993) The structure of large and small airways in nonfatal and fatal asthma. Am Rev Respir Dis 147: 405-410.
  25. Laitinen LA, Heino M, Laitinen A, Kava T, Haahtela T (1985) Damage of the airway epithelium and bronchial reactivity in patients with asthma. Am Rev Respir Dis 131: 599-606.
  26. Hackett TL, Knight DA (2007) the role of epithelial injury and repair in the origins of asthma. Curr Opin Allergy Clin Imunol 7: 63-68.
  27. Boulet LP, Laviolette M, Turcotte H, cartier A, Dugas M, et al. (1997) Bronchial subepithelial fibrosis correlates with airway responsiveness to methacholine. Chest 112: 45-52.
  28. Jeffery PK, Wardlaw AJ, Nelson FC, Collins JV, Kay AB (1989) Bronchial biopsies in asthma. An ultrastructural, quantitative study correlation with hyperreactivity. Am Rev Respir Dis 140: 1145-1153.
  29. Tanaka H, Yamada G, Saikai T, Hashimoto M, Tanaka S, et al. (2003) Increased airway vascularity in newly diagnosed asthma using a high-magnification bronchovideoscope. Am J Respir Crit Care Med 168: 1495-1499.
  30. Hashimoto M, Tanaka H, Abe S (2005) Quantitative analysis of bronchial wall vascularity in the medium and small airways of patients with asthma and COPD. Chest 127: 965-972.
  31. Vrugt B, Wilson S, Bron A, Holgate ST, Djukanovic R, et al. (2000) Bronchial angiogenesis in severe glucocorticoid-dependent asthma. Eur Respir J 15: 1014-1021.
  32. Hoshino M, Takahashi M, Aoike N (2001) Expression of vascular endothelial growth factor, basic fibroblast growth factor and angiogenin immunoreactivity in asthmatic airways and its relationship to angiogenesis. J Allergy Clin Immunol 107: 295-301.
  33. Simcock DE, Kanabar V, Clarke GW, Mahn K, Karner C, et al. (2008) Induction of angiogenesis by airway smooth muscle from patients with asthma. Am J Respir Crit Care Med Care 178: 460-468.
  34. Orsida BE, Li X, Hickey B, Thien F, Wilson JW, et al. (1999) Vascularity in asthma airways: Relation to inhaled steroid dose. Thorax 54: 289-295.
  35. Kanazawa H, Asai K, Hirata K, Yoshikawa J (2002) Vascular involvement in exercise-induced airway narrowing in patients with bronchial asthma. Chest 122: 166-170.
  36. Demoly P, Paggiaro P, Plaza V, Bolge S, Kannan H, et al. (2009) Prevalence of asthma control among adults in France, German, Italy, Spain and the UK. Eur Respir J 105: 105-112.
  37. Rabe KF, Adachi M, Lai CKW, Soriano JB, Vermeire AP, et al. (2004) Worldwide severity and control of asthma in children and adults: the global Asthma Insight and Reality survey. J Allergy Clin Immunol 114: 40-47.
  38. FitzGerald JM, Boulet LP, McIvor RA, Zimmerman S, Chapman KR (2006) Asthma control in Canada remains suboptimal: The Reality of Asthma Control (TRAC) study. Can Respir J 13: 253-259.
  39. Liu AH, Gilsenan AW, Stanford RH, Lincourt W, Ziemiecki R, et al. (2010) Status of asthma control in pediatric primary care: Results from the pediatric Asthma Control Characteristics and Prevalence Survey Study (ACCESS). J Pediatr 157: 276-28
  40. Pavord ID, Korn S, Howarth P, Bleecker ER, Buhl R, Keene ON, et al. (2012) Mepolizumab for severe asthma (DREAM): A multicenter, parallel, double-blind, randomsed, placebo-controlled, phase 3 trial. Lancet 380: 651-659.
  41. Ortega HG, Yancey SW, Mayer B, Gunsoy NB, Keene ON, Bleecker ER, et al. (2016) Severe eosinophilic asthma treated with mepolizumab stratified by baseline eosinophil thresholds: A secondary analysis of the DREAM and MENSA studies. Lancet Respir Med 4: 549-556.
  42. Bjemer L, Lemiere C, Maspero J, Weiss S, Zangrilli J, et al. (2016) Reslizumab for inadequately controlled asthma with elevated eosinophil levels: a randomized phase 3 study. Chest 150: 89-98.
  43. Bleecker ER, FitzGerald JM, Chanez P, Papi A, Weinstein SF, et al. (2016) Efficacy and safety of benralizumab for patients with severe asthma uncontrolled with high-dosage inhaled corticosteroids and long-acting β2-agonists (SIROCCO): A randomised, multicentre, placebo-controlled phase 3 trial. Lancet 388: 2115-2127.
  44. Castro M, Corren J, Pavord ID, Maspero J, Wenzel S, et al. (2018) Dupilumab efficacy and safety in moderate-to-severe uncontrolled asthma. N Engl J Med 378: 2486-2496.
  45. Syabbalo N (2020) Mechanisms of interleukin-17 in the pathogenesis of neutrophilic asthma. J Pulm Med Respir Res 6: 032.
  46. Busse WW, Holgate S, Kerwin E, Chon Y, Feng J, et al. (2013)Randomized, double-blind, placebo-controlled study of brodalumab, a humanized anti-IL-17 receptor monoclonal antibody, in moderate to severe asthma. Am J Respir Crit Care Med 188: 1294-1302.
  47. Nair P, Gaga M, Zerves E, Alagha K, Hargreave FE, et al. (2012) Safety and efficacy of a CXCR2 antagonist in patients with severe asthma and sputum neutrophils: A randomized, double-blind, placebo-controlled clinical trial. Clin Exp Allergy 42: 1097-1103.
  48. Byrne PM, Metev H, Puu M, Richter K, Keen C, et al. (2016) Efficacy and safety of a CXCR2 antagonist, AZD5059, in patients with uncontrolled persistent asthma: A randomized, double-blind, placebo-controlled trial. Lancet Respir Med 4: 797-806.
  49. Planaguma A, Domènech T, Pont M, Calama E, Gacia-Gonzále V, et al. (2015) Combined anti CXC receptors 1 and 2 therapy is a promising anti-inflammatory treatment for respiratory disease by reducing neutrophil migration and activation. Pulm Pharmacol Ther 34: 37-45.
  1. Moro L (2018) Risankizumab. Anti-IL-23p19 monclonal antibody, Treatment of psoriasis, Treatment of bowel disease, Treatment of asthma. Drugs of the Future 43: 331.
  2. Baillie GS, Tejeda GS, Kelly MP (2019) Therapeutic targeting of 3’5’-cyclic nucleotide phosphodiesterase: Inhibition and beyond. Nat Rev Drug Discov 18: 770-796.
  3. Kumar N, Goldminz AM, Kim N, Gottlieb AB (2013) Phosphodiesterase 4-targeted treatment for autoimmune diseases. BMC Med 11: 96.
  1. Halpin DM (2008) ABCD of the phosphodiesterase family: Interaction and differential activity in COPD. In J Chron Obstruct Pulmon Dis 3: 543-561.
  2. Muller T, Engels P, Fozard JR (1996) Subtypes of the type 4 cAMP phosphodiesterases: Structure, regulation and selective inhibition. Trends Pharmacol Sci 17: 294-298.
  3. Page CP (2014) Phosphodiesterase inhibitors for the treatment of asthma and chronic obstructive pulmonary disease. Int Arch Allergy Immunol 165: 152-164.
  4. Maurice DK, Ke H, Ahmad F, Wang Y, Chung J, et al. (2014) Advances in targeting cyclic nucleotide phosphodiesterases. Nat Rev Drug Discov 13: 290-314.
  5. Li H, Zuo J, Tang W (2018) Phosphodiesterase-4 inhibitors in the treatment of inflammatory diseases. Front Pharmacol 9: 1048.
  1. Mulhall AM, Droege CA, Ernst NE, Panos RJ, Zafar MA (2015) Phosphodiesterase 4 inhibitors for the treatment of chronic obstructive pulmonary disease: A review of current and developing drugs. Expert Opin Inv Drug 24: 1597-1611.
  2. Cazzola M, Calzetta L, Rogliani P, Matera MG (2016) The discovery of roflumilast for the treatment of chronic obstructive pulmonary disease. Expert Opin Drug Discov 11: 733-744.
  3. Chiricozzi A, Caposiena D, Garofalo V, Cannizzaro MV, Chimenti S, (2016) A new therapeutic for the treatment of moderate-to-severe plaque psoriasis: Apremilast. Expert Rev Clin Clin Immunol 12: 237-249.
  4. Cheape AC, Murrell DF (2017) 2% Crisaborole topic ointment for the treatment of mild-to-moderate dermatitis. Exprt Rev Clin Immunol 13: 415-423.
  5. Salari P, Abdollahi M (2012) Phosphodiesterase inhibitors in inflammatory bowel disease. Expert Opin Investig Drugs 261: 264.
  6. Donnell JM, Zhang HT (2004) Antidepressant effects of inhibitors of cAMP phoaphodiesterase (PDE4). Trends Pharmacol 25: 158-163.
  7. Souness JE, Aldous D, Sargent C (2000) Immunosuppressive and anti-inflammatory effects of cyclic AMP phosphodiesterase (PDE) type 4 inhibitors. Immunopharmacology 47: 127-162.
  8. Schafer PH, Truzzi F, Parton A, Wu L, Kosek J, et al. (2016) Phosphodiesterase 4 in inflammatory diseases: Effects of apremilast in psoriatic blood and in dermal myofibroblasts through the PDE4/CD271 complex. Cell Signal 28: 753-763.
  1. Kawamatawong T (2017) Roles of roflumilast, a selective phosphodiesterase 4 inhibitor, in airway diseases. J Thorac Dis 9: 1144-1154.
  2. Schafer P (2012) Apremilast mechanism of action and application to psoriasis and psoriatic arthritis. Biochem Pharmacol 83: 1583-1590.
  1. Sanz MJ, Cortijo J, Morcillo EJ (2005) PDE inhibitors as new anti-inflammatory drugs: effects on cell trafficking and cell adhesion molecules expression. Pharmacol Ther 106: 269-297.
  2. Sousa LP, Lopes F, Silva DM, Tavares LP, Vieira TA, et al. (2010) PDE4 inhibition drives resolution of neutrophilic inflammation by inducing apoptosis in a PKA-PI3K/Akt-dependent and NF-κB-independent manner. J Leukoc Biol 87: 895-904.
  3. Fitzgerald MF, Spicer D, McAulay AE, Wollin L, Beume R (2006) Roflumilast but not methylprednisolone inhibited cigarette smoke-induced pulmonary inflammation in guinea pigs. Eur Respir J 28: 663.
  4. Martorana PA, Lunghi B, Lucattelli M, De CG, Beume R, et al. (2008) Effect of roflumilast on inflammatory cells in the lungs of cigarette smoke-exposed mice. BMC Pulm Med 8: 17-20.
  5. Cortijo J, Iranzo A, Milara X, Mata M, Cerda-Nicolas M, et al. (2009) Roflumilast, a phsphodiesterase 4inhibitor, alleviates bleomycin-induced lung injury. Br J Pharmacol 156: 534-544.
  6. Jones NA, Boswell-Smith V, Lever R, (2005) the effect of selective phosphodiesterase isoenzyme inhibition on neutrophil function in vitro. Pulm Pharmacol Ther 18: 93-101.
  7. Eskandari N, Wickramasinghe T, Peachell PT (2004) Effects of phosphodiesterase inhibitors on interleukin-4 and interleukin-13 generation from human basophils. R J Pharmacol 142: 1265-1272.
  8. Buenestado A, Grassin-Delyle S, Guitard F, Naline E, Faisy C, et al. (2012) Roflumilast inhibits the release of chemokines and TNF-α from human lung macrophages stimulated with liposaccharide. Br J Pharmacol 165: 177-1890.
  9. Hatzelmann A, Morcillo EJ, Lungarella G, Adnot S, Sanjar S, et al. (2010) The preclinical pharmacology of roflumilast – a selective, phosphodiesterase 4 inhibitor in development for chronic obstructive pulmonary disease. Pulm Pharmacol Ther 23: 235-256.
  10. Mata M, Sarria B, Buenestado A, Cortijo J, Cerda M, et al. (2005) Phosphodiesterase 4 inhibition decreases MUC5AC expression induced by epidermal growth factor inhuman airway epithelial cells. Thorax 60: 144-152.
  11. Milara J, Armengot M, Banuls P, Tenor H, Beume R, et al. (2012) Roflumilast N-oxide, a PDE4inhibitor, improves cilia motility and ciliated human bronchial epithelial cells compromised by cigarette smoke in vitro. Br J Pharmacol 166: 2243-2262.
  12. Togo S, Liu X, Wang X, Sugiura H, Kamio k, et al. (2009) PDE4 inhibitors roflumilast and rolipram augment PGE2 inhibition of TGF-β1-stimuated fibroblasts. Am J Physiol Lung Cell Mol Physiol 296: L959-969.
  13. Kohyama T, Liu X, Wen FQ, Wen FQ, Zhu KY, et al. (2002) PDE4 inhibitors attenuate fibroblast chemotaxis and contraction of native collagen gels. Am J Respir Cell Mol Biol 26: 694-701.
  14. Louw C, Williams Z, Venter L, Leichtl S, Wirlitsch CS, et al. (2007) Roflumilast, a phosphodiesterase 4 inhibitor, reduces airway hyperresponsiveness after allergen challenge. Respiration 74: 411-417.
  15. Bateman ED, Bousequet J, Aubier M, Bredenbröker D, O’Byrne PM (2015) Roflumilast for asthma: efficacy findings in non-placebo-controlled comparator and dosing studies. Pulm Pharmacol Ther 35: 11-19.
  16. Bardin P, Kanniess F, Gauvreau G, Bredenbröker D, Rabe KF (2105) Roflumilast for asthma: Efficacy findings in mechanism of action studies. Pulm Pharmacol Ther 35: 4-10.
  17. Meltzer EO, Chervinsky P, Busse W, Ohta K, Bardin P, et al. (2015) Roflumilast for asthma: Efficacy findings in placebo-controlled studies. Pulm Pharmacol Ther 35: 20-27.
  18. Bateman ED, Goehring U-M, Richard F, Watz H (2016) Roflumilast combined with montelukast versus montelukast alone as add-on treatment in patients with moderate-to-severe asthma. J Allergy Clin Immunol 138: 142-149. 
  1. Berry MA, Morgan A, Shaw DE, Parker D, Green RH, et al. (2007) Pathological features and inhaled corticosteroid response of eosinophilic and non-eosinophilic asthma. Thorax 62: 1043-1049.
  2. Simpson J, Powell H, Boyle MJ, Scott RJ, Gibson PG (2008) Clarithromycin targets neutrophilic airway inflammation in refractory asthma. Am J Respir Crit Care Med 177: 148-155.
  3. Zuckerman JM (2004) Macrolide and ketolides: Azithromycin, clarithromycin, telithromycin. Inf Dis Clin North Am 18: 621-649.
  1. Johnston SL, Martin RJ (2005) Chlamydophila pneumoniae and Mycoplasma pneumonia: A role in asthma pathogenesis? Am J Rspir Crit Care Med 172: 1078-1089.
  2. Biscione GL, Corne AJ, Chauhan AJ, Johnson SL (2004) Increased frequency of detection of Chlamydophila pneumoniae in asthma. Eur Respir J 24: 745-749.
  3. Martin RJ, Kraft M, Chu HW, Berns EA, Cassell GH (2001) A link between chronic asthma and chronic infection J Allergy Clin Immunol 107: 595-601.
  4. Sakito O, Kadota J, Kohno S, Abe K, Shirai R, et al. (1966) Interleukin 1 beta, tumor necrosis factor alpha and interleukin 8 in bronchoalveolar lavage fluid of patients with panbronchiolitis: a potential mechanism of macrolide therapy. Respiration 63: 42-48.
  5. Verleden GM, Vanaudenaerde BM, Dupot LJ (2006) Azithromycin reduces airway neutrophilia and interleukin-8 in patients with bronchiolitis obliterans syndrome Am J Respir Crit Care Med 174: 566-570.
  6. Piacentini GL, Peroni DG, Bodini A, Pigozzi R, Costella S, et al. (2007) Azithromycin reduces bronchial hyperresponsiveness and neutrophilic airway inflammation in asthmatic children: A preliminary report. Allergy Asthma Proc 28: 194-198.
  7. He J, Zhu N, Chen X (2009) Clinical impacts of azithromycin on lung function and cytokines for asthmatic patients. JMS 36: 719-722. 
  1. Fonseca-Aten M, Okada PJ, Bowlware KL, Chavez-Bueno S, Mejias A, et al. (2006) Effect of clarithromycin on cytokines and chemokines in children with an exacerbation of recurrent wheezing: A double-blind, randomized, placebo-controlled trial. Ann Allergy Asthma Immunol 97: 457-463.
  2. Wang Y, Zhang S, Qu Y (2012) Effect of clarithromycin on non-eosinophilic refractory asthma. J Clin Pulm Med 17: 1948-1951.
  3. Kamoi H, Kurihar N, Fujiwara H, Hirata K, Takeda T (1995) The macrolide antibacterial roxithromycin reduces bronchial hyperresponsiveness and superoxide anion production by polymorphonuclear leukocytes in patients with asthma. J Asthma 32: 191-197.
  4. Zimmermann P, Ziesenitz VC, Curtis N, Ritz N (2018) Effects of macrolides - a systemic review of underlying mechanisms. Front Immunol.
  5. Brusselle GG, Vandertichele C, Jorgens P, Deman R, Slabbynck H, et al. (2013) Azithromycin for prevention of exacerbations in severe asthma (AZISAST): A multicentre randomised double-blind placebo-controlled trial. Thorax 68: 322-329.
  6. Gibson PG, Yang IA, Upham JW, Reynolds PN, Hodge S, et al. (2017) Effect of azithromycin on asthma exacerbations and quality of life in adults with persistent uncontrolled asthma (AMAZES): A randomized, double-blind, placebo-controlled trial. Lancet 390: 659-668.
  7. Johnston SL, Blasi F, Black PN, Martin RJ, Farrell DJ, et al. (2006) The effect of telithromycin in acute exacerbations of asthma. N Engl J Med 354: 1589-1600.
  8. Johnston SL, Szigeti M, Cross M, Brightling C, Chaudhuri R, et al. (2016) Azthromycin for acute exacerbaions of asthma: The AZALEA randomized clinical trial. JAMA Int Med 176: 1630-1637.
  9. Bosnar M, Cužic S, Bošnjak B, Nujic K, Ergovic G, et al. (2011) Azithromycin inhibits macrophage interleukin-1beta production through inhibition of activator protein-1 in liposaccharide-induced murine pulmonary neutrophilia. In Immunopharmacol 11: 424-434.
  10. Cheung PS, Hosseini K (2010) Anti-inflammatory activity of azithromycin as measured by its NF-kappaB, inhibitory activity. Ocul Immunol Inflamm 18: 32-37.
  11. Cigana C, Assael BM, Melotti P (2007) Azithromycin selectively reduces necrosis factor alpha levels in cystic fibrosis airway epithelial cells. Antimicrob Agents Chemother 51: 975-981.
  12. Aghai ZH, Kode A, Saslow JG, Nakhla T, Farhath S, et al. (2007) Azithromycin suppresses activation of nuclera factor-kappa B and synthesis of pro-inflammatory cytokines in tracheal aspirate cells from premature infants. Pediatr Res 62: 483-488.
  13. Kanoh S, Rubin BK (2010) Mechanisms of action and clinical application of macrolides as immunomodulatory medications. Clin Microbiol Rev 23: 590-615.
  14. Shinkai M, Henke MO, Rubin BK (2008) Macrolide antibiotics as immunomodulatory medications: Proposed mechanisms of action. Pharmacol Ther 117: 393-405.
  15. Ito K, Herbert C, Siegle JS, Vuppusetty C, Hansbro N, et al. (2008) Steroid-resistant neutrophilic inflammation in a mouse model of an acute exacerbation of asthma. Am J Respir Crit Care Med 39: 543-550.
  16. Essilfie AT, Horvat JC, Kim RY, Mayall JR, Pinkerton JW, et al. (2015) Macrolide therapy suppresses key features of experimental steroid-sensitive and steroid-insensitive asthma. Thorax 70: 458-467.
  17. Kobayashi Y, Wada H, Rossios C, Takagi D, Charron C, et al. (2013) A novel macrolide/fluoroketolide, solithromycin (CEM-101), reverses corticosteroid insensitivity via phosphoinositide 3-kinase pathway inhibition. Br J Pharmacol 169: 1024-1034.
  18. Barnes PJ (2013) Corticosteroid resistance in patients with asthma and chronic obstructive pulmonary disease. J Allergy Clin Immunol 131: 638-645.
  19. Garey KW, Gotfried MH, Khan IJ, Varma S, Danzinger LH (2000) Long-term clarithromycin decreases prednisone requirements in elderly patients with prednisone-dependent asthma. Chest 118: 1826-1827.
  20. Serisier DJ (2013) Risks of population antimicrobial resistance associated with chronic macrolide use for inflammatory airway diseases. Lancet Respir Med 1: 262-274.
  21. Hare KM, Singleton RJ, Grimwood K, Valery PC, Cheng AC, et al. (2013) Longitudinal nasopharyngeal carriage and antibiotic resistance of respiratory bacteria in indigenous Australian and Alaska native children with bronchiectasis. PLoS One 8: 70478.
  22. Idris SF, Chilvers ER, Haworth C, Mckeon D, Condliffe AM (2008) Azithromycin therapy for neutrophilic airways disease: Myth or magic? Thorax.
  23. Girodet PO, Ozier A, Bara I, Tunon de Lara JM, Marthan R, et al. (2011) Airway remodeling in asthma: New mechanisms and potential for pharmacological intervention. Pharmacol Ther 130: 325-337.
  24. Zuyderduyn S, Sukkar MB, Fust A, Dhaliwal S, Burgess JK (2008) Treating asthma means treating airway smooth muscle cells. Eur Respir J32: 265-274.
  25. Tan LD, Yoneda KY, Louie S, Hogarth DK, Castro M (2019) Bronchial thermoplasty: A decade of experience: State of the art. J Allergy Clin Immunol in Pract 7: 71-80.
  26. Laxmanan B, Hogarth DK (2015) Bronchial thermoplasty in asthma: Current perspectives. J Asthma Allergy 8: 39-49.
  27. Thomson NC (2018) Bronchial thermoplasty as a treatment for severe asthma: Controversies, progress and uncertainties. Exp Rev Respir Med 12: 269-282.
  28. Bonta PI, Chanez P, Annema JT, Shah PI, Niven R (2018) Bronchial thermoplasty in severe asthma: Best practice recommendations from an expert panel. Respiration 95: 289-300.
  29. Food and Drug Administration (2010) Alair bronchial thermoplasty system: Alair catheter and Alair RF controller. Food and Drug Administration, USA.
  30. National Library of Medicine (2014) Boston scientific corporation bronchial thermoplasty in severe asthma (PAS2). National Library of Medicine, USA.
  31. Cox PG, Miller J, Mitzner W, Leff AR (2004) Radiofrequency ablation of airway smooth muscle for sustained treatment of asthma: Preliminary investigations. Eur Respir J 24: 659-663.
  32. Thomson NC, Bicknell S, Chaudhuri R (2012) Bronchial thermoplasty for severe asthma. Curr Opin Allergy Clin Immunol 12: 241-248.
  33. Dombret MC, Alagha K, Boulet LP, Brillet PY, Joos G, et al. (2014) Bronchial thermoplasty: A new therapeutic option for the treatment of severe, uncontrolled asthma in adults. Eur Respir Rev 23: 510-518.
  34. Thomson NC (2019) Recent developments in bronchial thermoplasty for severe asthma. J Asthma Allergy 12: 375-387.
  35. Pretolani M, Dombret MC, Thabut G, Knap D, Hamidi F, et al. (2014) Reduction of airway smooth muscle mass by bronchial thermoplasty in patients with severe asthma. Am J Respir Crit Care Med 190: 1452-1454.
  36. Facciolongo N, Stefano AD, Pietrini V, Galeone C, Bellanova F, et al. (2018) Nerve ablation after bronchial thermoplasty and sustained improvement in severe asthma. BMC Pulm Med 18: 29.
  37. Debray MP, Dombret MC, Pretolani M, Thabut G, Alavoine L, et al. (2017) Early computed tomography modifications following bronchial thermoplasty in patients with severe asthma. Eur Respir J 49: 1601565.
  38. Qui M, Lai Z, Wei S, Jiang Q, Xie J, et al. (2018) Bronchiectasis after bronchial thermoplasty. J Thorac Dis 10: 721-726.
  39. Facciolongo N, Menzella F, Lusuardi M, Piro R, Galeone C, et al. (2015) Recurrent lung atelectasis from fibrin plugs as a very early complication of bronchial thermoplasty: a case report. Multidisplin Respir Med 10: 9.
  40. Funatsu A, Kobayashi K, Iikura M, Ishii S, Izumi S, et al. (2018) A case of pulmonary cyst and pneumothorax after bronchial thermoplasty. Respirol Case Rep 6: 00286.
  41. Menzella F, Lusuardi M, Galeone C, Montanari G, Cavazza A, et al. (2018) Heat-induced necrosis after bronchial thermoplasty: A new concern? Allergy Asthma Clin Immunol 14: 25.
  42. Pretolani M, Bergqvist A, Thabut G, Dombret MC, Knapp D, et al. (2017) Effectiveness of bronchial thermoplasty in patients with severe refractory asthma: Clinical and histopathologic correlations. J Allergy Clin Immunol 139: 1176-1185.
  43. Langton D, Sha J, Ing A, Fielding D, Wood E (2017) Bronchial thermoplasty in severe asthma in Australia. Intern Med J 47: 536-541.
  44. Thomson NC, Chanez P (2017) How effective is bronchial thermoplasty for severe asthma in clinical practice? Eur Respir J 50: 1701140.
  45. Cox G, Thomson NC, Rubin AS, Niven RM, Corris PA, et al. (2007) Asthma control during the year after bronchial thermoplasty. N Engl J Med 356: 1327-1337.
  46. Pavord ID, Cox G, Thomson NC, Rubin AS, Corris PA, et al. (2007) Safety and efficacy of bronchial thermoplasty in symptomatic, severe asthma. Am J Respir Crit Care Med 176: 1185-1191.
  47. Castro M, Rubin AS, Laviolette M, Fiterman J, Lima M DA, et al. (2010) Effectiveness and safety of bronchial thermoplasty in the treatment of severe asthma: A multicenter, randomized, double-blind, Sham-controlled clinical trial. Am J Respir Crit Care Med 181: 116-124.
  48. Chupp G, Laviolette M, Cohn L, McEvory C, Bansal S, et al. (2017) Long-term outcomes of bronchial thermoplasty in subjects with severe asthma: A comparison of 3-year follow-up results from two prospective multicentre studies. Eur Respir J 50: 1700017.
  49. Zhou JP, Feng Y, Wang Q, Zhou LN, Wan HY, et al. (2016) Long-term efficacy and safety of bronchial thermoplasty in patients with moderate-to-severe persistent asthma: a systemic review and meta-analysis. J Asthma 53: 94-100.
  50. Pavord ID, Thomson NC, Niven RM, Corris PA, Chung KF, et al. (2013) Safety of bronchial thermoplasty in patients with severe refractory asthma. Ann Allergy Asthma Immunol 111: 402-407.
  51. Thomson NC, Rubin AS, Niven RM, Corris PA, Siersted HC, et al. (2011) Long-term (5 year) safety of bronchial thermoplasty: Asthma Intervention Research (AIR) trial. BMC Pulm Med 11: 8.
  52. Wechsler ME, Laviolette M, Rubin AS, Fiterman J, Silva JR LE, et al. (2013) Bronchial thermoplasty: Long-term safety and effectiveness in patients with severe persistent asthma. J Allergy Clin Immunol 132: 1295-1302.
  53. Chaudhuri R, Rubin A, Fiterman JDC, Sumino K, Silva JRLE, et al. (2019) Ten-year follow-up of subjects who received bronchial thermoplasty (BT) in 3 randomized controlled studies (BT10+). Eur Respir Soc Congr 54: 63.
  54. d’Hooghe JNS, Hacken NHTT, Weersink EJM, Sterk PJ, Annema JT, et al. (2018) Emerging understanding of the mechanism of action of bronchial thermoplasty in asthma. Pharmacol Ther 181: 101-107.
  55. Mainardi AS, Catro M, Chupp G (2019) Bronchial theroplasty. Clin Chest Med 40: 193-207.
  56. Healthcare Improvement Scotland (2019) British guideline on the management of asthma. Healthcare Improvement Scotland, Edinburgh, UK.

Citation: Syabbalo N (2020) Clinical Features and Management of Neutrophilic Asthma. J Pulm Med Respir Res 6: 036.

Copyright: © 2020  Nightingale Syabbalo, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.


Herald Scholarly Open Access is a leading, internationally publishing house in the fields of Sciences. Our mission is to provide an access to knowledge globally.



© 2024, Copyrights Herald Scholarly Open Access. All Rights Reserved!