Open access peer-reviewed chapter

Genetics of Allergic Asthma and Current Perspectives on Therapeutic Management

Written By

Mina Youssef, Cynthia Kanagaratham, Mohamed I. Saad and Danuta Radzioch

Reviewed: 13 April 2016 Published: 06 July 2016

DOI: 10.5772/63651

From the Edited Volume

Asthma - From Childhood Asthma to ACOS Phenotypes

Edited by Celso Pereira

Chapter metrics overview

3,000 Chapter Downloads

View Full Metrics

Abstract

Globally, more than 300 million people are asthmatics and this number has been estimated to become 400 million by 2025. Asthma is a chronic inflammatory condition, which, although has no cure, is treatable in most patients. The most common structural alterations in asthmatic airways include thickening of the epithelial and sub epithelial layers, increased airway smooth muscle mass, and angiogenesis. Several genetically controlled factors greatly influence the predisposition and severity of allergic asthma. Twin studies have attributed as much as 75% of asthma susceptibility to heredity. Particularly, genome-wide association studies (GWASs) have discovered several asthma and/or atopy susceptibility genes. Current treatment protocols for managing asthma involve the use of corticosteroids and β-agonists. Over the last 40 years, there has been a marked development-targeted therapy for asthma, such as anti-leukotrienes, anti-immunoglobulin (Ig)E, anti-tumor necrosis factor (TNF)-α, and anti-interleukins (ILs) (Th2 cytokines). To identify novel targets and to develop newer drug generations, better understanding of asthma molecular pathophysiology is required. Furthermore, the pharmacogenetic studies, focusing on better understanding of beneficial or/and adverse effects to anti-asthma drugs, will also facilitate the development of more effective and targeted treatments in specific subpopulations of patients suffering from asthma.

Keywords

  • asthma
  • asthma therapies
  • genetics
  • pharmacogenetics

1. Introduction

Asthma is an inflammatory chronic condition that has reached globally epidemic levels. Although no cure exists, symptoms are treatable in most patients [1]. Statistically, the number of asthmatic cases has been on the rise over the past 10 years and affecting up to 10% of adults and 20% of children worldwide [2]. Globally, more than 300 million people are asthmatics, and this estimate is predicted to become 400 million by 2025 [3]. The worldwide economic burden caused by asthma is predicted to be more than that of both acquired immunod eficiency syndrome (AIDS) and tuberculosis combined together. For example, in the United States of America, the annual asthma care costs exceed US$6 billion [4]. Moreover, these numbers are due to the fact that more than 50% of asthmatic cases are poorly controlled by medication, since the response to treatments varies considerably among patients despite having similar clinical features [3, 5]. Asthma is characterized by altered and distinct clinical changes in the lung airways obstructing the flow of air into the lungs. The most prominent airway remodeling features include epithelial and subepithelial layer thickening, increased airway smooth muscle (ASM) mass, and angiogenesis [6]. Different classes of asthma therapies address one or more of the phenotypes of asthma; however, the heterogeneous nature of the disease prevents homogeneous clinical outcomes in response to the current treatment guidelines [7].

In the past two decades, the field of human genetics has evolved due to the advancements in the human genome project and high-throughput sequencing technologies [8, 9]. Currently, the advances in genetic, pharmacodynamic, and pharmacokinetic studies, analyzing responsiveness of patients to various therapies, may eventually allow to prescribe personalized treatment and to shift asthma therapies from classical standards, using mostly corticosteroids and β-adrenergic agonists, to a highly tailored approach [10]. Future genetic profiles of the population would form the basis of tomorrow’s treatments in order to potentiate the required therapeutic benefits, and to diminish any possible adverse effect risks. Overall, there remains a great need for comprehensive drug research, paralleled with high-throughput genetic profiling, in order to treat asthma in a personalized or stratified manner [11].

Advertisement

2. Genetic control of airway hyperresponsiveness, atopy, and allergic asthma

The heritable nature of asthma has been demonstrated through various types of studies over the past decades. Family and twin studies indicate that 60–70% of asthma cases are due to genetic factors. Moreover, it has been proven that the concordance of asthma is greater among monozygotic twins rather than among dizygotic ones. Adoption studies have shown greater disease prevalence within biological relatives of the affected people compared to the adopted family [12].

Higher prevalence of allergic disease phenotypes is observed among relatives of atopic individuals compared to nonatopic individuals. Overall, the heritability estimates remain in between the range of 30–66% for airway hyperresponsiveness, 35–95% for asthma, and 35–84% for total serum IgE levels [13]. It is clear that both the inter-genetic individual differences and the degree of allergen exposure contribute to these variations in heritability. Heritability of asthma is linked to both disease susceptibility and severity. While the main concern of asthma genetic studies has been on disease susceptibility, increasing evidence shows that many genetic variants are important in asthma progression and severity as well [14]. Lung function tests in asthma showed that genes in the T-helper lymphocyte 1 (Th1) pathway affect asthma severity; meanwhile, T-helper lymphocyte 2 (Th2) pathway genes relate to susceptibility [14]. Based on these hypotheses, genes associated with asthma susceptibility differ from those related to asthma severity; hence, it is important to define both groups distinctly.

By knowing the genetic signature associated with allergic asthma, geneticists can help to better understand the molecular mechanism of this disease, and the shared and distinct pathways among other allergic diseases. Moreover, the genetic signature of asthma-associated genes with altered expression during the peak of asthmatic episodes may help predict the severity and response to therapy. Unfavorable response might be identified and, consequently, more targeted and personalized treatments can be considered for this complex trait. The human genome project and the ongoing advancements in sequencing technologies, both, resulted in more successful gene discovery over the last years, even in diseases as complexed as asthma. Since then, dozens of susceptibility genes were identified through a large variety of methods and rationales. ADAM33 is the first asthma susceptibility gene to be discovered through positional cloning [15]. ADAM33 (also known as Disintegrin and metalloproteinase domain-containing protein 33) is a membrane-bound metalloproteinase enzyme that has been involved in several cellular interactions involving cell-cell and cell-matrix events [16]. Variants in this gene have been correlated to asthma susceptibility and bronchial hyperresponsiveness, but not atopy. Due to its clinical significance, ADAM33 studies were conducted among 33 different asthmatic population samples all over the world. Additionally, numerous studies have suggested that altered ADAM33 DNA methylation patterns could result in diversely unbalanced biological effects in the airways [17]. Studies focused on candidate genes have examined a number of genes involved in asthma and highlighted more than 100 interesting genetic spots; however, the role of those loci in asthma susceptibility remains largely unexplored [18].

Genome-wide association studies (GWASs) extensively investigate the unknown genetic bases of many intricate disorders including asthma [19,20]. In the first reported GWAS study for asthma susceptibility, Moffatt et al. [21] identified the 17q21 locus, containing several genes, for example, ORMDL3 and GSDMB as being associated with childhood asthma. Importance of this region was later on replicated in numerous subsequent studies [2224]. Expression levels of the gene ORMDL3 are differentially regulated by distinct haplotypes in this region. This gene encodes protein acting as an inhibitor of sphingolipid biosynthesis and in general Orm family proteins were shown to be implicated in the control of sphingolipid homeostasis [25]. Dysregulated sphingolipid formation in the respiratory tract instigates airway hyperreactivity [26] although exact molecular steps are still not known. The results of these studies suggested that the mechanisms of asthma development are linked with genetically determined abnormalities in some patients resulting in their inability to control balance between oxidative and anti-oxidative reactions. The mechanisms of asthma development are linked with genetically determined abnormalities in the functioning of antioxidant defense enzymes. These alterations seem to be accompanied by a systemic imbalance between oxidative and anti-oxidative reactions with the shift of the redox state toward increased free radical production, oxidation of proteins and phospholipids, and eventually to their selective degradation.

To increase the power of detection of modest alleles due to the large sample size, the results of individual GWAS need to be gathered into a meta-analysis. The scientific literature recognizes two meta-analyses of asthma GWAS. One was done by the GABRIEL Consortium [27] of the European investigators, and the other was conducted by the EVE Consortium of the US investigators [22]. While the EVE meta-analysis included diverse subjects from different ethnic background, US and Mexico population backgrounds, the GABRIEL meta-analysis included only European subjects. Overall, these two thorough meta-analyses present a comprehensive overview of the genetic associations for asthma. Some associations are shared among different populations; by contrast, others are specific to one race. Grouping GWAS in this way increases the power of genetic detection, contrasts different ethnic groups’ genotypes, and highlights the worldwide populations’ genetic patterns. Overall independent GWASs have identified large number of candidate loci that deserve further testing. Replication studies help to prioritize which genes deserve further study, based on their identification in multiple populations.

Additionally, more loci were identified to be associated with asthma; these include interleukin (IL)-33 (on 9p24), HLA-DR/DQ (on 6p21), IL1RL1/IL18R1 (on 2q12), TSLP (on 5q22), and IL13 (on 5q31) [22,27,28]. Collectively with ORMDL3/GSDMB (on 17q21), these are the most remarkable and consistent loci, which are identified for asthma. Since Moffatt et al. had published the first GWAS results for asthma, identifying ORMDL3 as a candidate gene, numerous other studies have been conducted investigating an array of phenotypes which are observed in allergic diseases. In particular, FCER1A, RAD50, and STAT6 have been associated with total serum IgE levels [29].

Advertisement

3. Environmental factors contributing to asthma

Parallel to genetic factors, environmental factors are also involved in the development and progression of asthma (Figure 1). The exposure to some environmental factors was shown to contribute not only to asthma but also to other related respiratory disorders, for example, emphysema development. By contrast, there are also some other environmental factors that seem to be solely linked to the development of asthma but not to other inflammatory or/and respiratory disorders [30]. Various studies assessed the risk factors of asthma and found evidence that allergen exposure, respiratory tract infections, gastroesophageal reflux disease (GERD), and physical and psychosocial stress might represent individual risk factors. It is important to keep in mind that some other environmental factors are protective, such as maternal diet, breastfeeding, and farming conditions [31].

Allergen exposure is the major factor impacting sensitization and constitutes the most common cause of asthmatic exacerbations in adults and children. A wide variety of inhaled allergens may trigger asthma symptoms, for example, house dust mite [32], pollens [33], cockroaches [34], and animal fur [35]. Respiratory tract infections have been implicated in asthma occurrence and exacerbation as well. Examples include infection with viruses [36,37], Mycoplasma [38], and Chlamydia species [39]. Based on the conclusions from the Japanese study, which included 3085 patients, the change in weather followed by smoking was identified as two leading asthma-exacerbating factors [40]. Although (passive) smoking is a predominant contributing factor for the development of asthma [41], one occupational study [42] has shown that nonsmokers might also develop asthma due to occupational air pollutant exposure.

Figure 1.

Environmental stress, in conjunction with genetic factors, both contribute to the development of asthma exacerbations.

Additionally, a correlation has been observed between the presence of asthma and gastroesophageal reflux-induced disease, with reports showing one-third of asthmatic patients also diagnosed with GERD [43,44]. Although the coexistence of GERD in asthmatic patients did not affect asthma severity, the airway resistance was significantly higher in asthmatic patients with GERD [45]. Some other psychosocial factors such as parental stress during childhood [46] and the socioeconomic status [47] are reported to influence allergic inflammation severity. It is estimated that psychopathology is six times more common among asthmatics, and accordingly it correlates more closely with the asthmatic quality of life, rather than with lung physiological functions [48,49]. In both directions, psychopathology is supposed to precipitate asthma or vice versa; psychopathology may develop as a consequence of asthma [50].

Advertisement

4. Asthma pathophysiology

Scientists tried to uncover alterations related to asthma since a long time ago. One of the oldest publications that discussed asthma pathophysiology was in 1873 [51]. Later on, in 1886, F.H. Bosworth concluded a possible relation between asthma and hay fever [52]. Clearly, it is well known that asthmatic patients suffer from reversible airway obstruction resulting from an allergen exposure, consequently releasing multiple broncoconstricting mediators that stimulate airway muscles to contract. Furthermore, airways narrow results from past and current mucus and edema occlusion [53]. The chronic inflammation and associated repair of lung airways leads to structural changes, referred to as “airway remodeling.” Airway remodeling (Figure 2) usually involves lung epithelial layer injury and includes features such as subepithelial thickening, airway smooth muscle hyperplasia, and angiogenesis [6].

Figure 2.

Schematic representation of the major events underlying asthma pathophysiology.

Asthma and COPD (chronic obstructive pulmonary disease) are now considered to be discrete respiratory disorders. Although both share several similar underlying mechanisms, driving airway obstruction in COPD, and hyperresponsiveness in asthma, core molecular pathology remains to be mostly different for both [54]. Pauwels et al. [55] defined COPD as “a disease state characterized by airflow limitation that is not fully reversible. The airflow limitation is usually progressive and associated with an abnormal inflammatory response of lungs to noxious particles and gases.” One important reason of asthma and COPD overlap is the effect of aging. Asthma-COPD overlap syndrome (ACOS) is a medically recognized coexisting syndrome of both asthma and COPD [56]. Some other health conditions may occur more frequently in asthmatic patients. Rhinosinusitis [57], obstructive sleep apnea [58], or GERD [59] are the most common documented comorbidities. Substantially, they can contribute to the same pathophysiological process, which is already triggered by allergic response or alter asthma phenotype detrimentally. The impact of these diseases on asthma is variable and still not fully clear [60].

Advertisement

5. Structural alterations in asthmatic airway walls

5.1. Epithelial/subepithelial layer thickening

Epithelial changes are not unique to asthma, they are also observed, in more or less of similar manner, in lungs of smokers and cancer patients [61]. Epithelial layer damage in asthma includes loss of ciliated cell layer, shedding of the epithelium, goblet cell hyperplasia and growth factors, cytokine and chemokine upregulation [62].

One important feature of asthma, which has been routinely used as an asthma severity index, is the thickening of the subepithelial airways layer. The epithelial and subepithelial layer thickening is caused by the overdeposition of extracellular matrix (ECM) proteins [63]. Roche et al. observed that intensive layers of collagen sedimentation contribute to the thickened subepithelial basement membrane. Through immunohistochemistry, they have shown that the commonly involved collagen types are collagen I, III and V, and fibronectin [64]. Additionally, the cells that are responsible for ECM protein production are myofibroblasts and fibroblasts, as both are embedded in the sophisticated ECM which they secrete [65]. Meanwhile, some inflammatory cells, for example, T cells, mast cells, and eosinophils also accumulate in the submucosal layer [66]. Moreover, transforming growth factor-β (TGF-β), and some similar growth factors, is usually secreted by the lung epithelial cells echoing any ongoing lung injury, and consequently directly impress the matrix proteins’ production by fibroblasts/myofibroblasts. By increasing the airway rigidity, however, Holgate et al. suggested that the airway thickening due to the ECM proteins precipitation may in fact have a remodeling protective effect via postponing long-term bronchoconstriction events [62]. Collectively, the ECM proteins, the lung structural cells (i.e., epithelial cells and fibroblasts), and the immune system inflammatory cells, all interact together and control the overall airway remodeling and fibrosis [67].

5.2. Hyperproliferation of airway smooth muscle mass

Hyperproliferation of airway smooth muscle mass is a common event in asthma and has been suggested to be implicated in its pathophysiology. Hyperplasia and hypertrophy of the ASM in the bronchial airways of asthmatics can be observed by three-dimensional (3D) morphometric studies [68]. Airways smooth muscle layer is estimated to be increased by 25–55% in nonfatal asthma and up to 50–200% in fatal asthma [69]. Meanwhile, in response to some growth factors like TGF-β, vascular endothelial growth factor (VEGF), and connective tissue growth factor (CTGF), ASM cells actively participate in the remodeling process through the process of ECM synthesis [70]. ASM cells also express cellular adhesion molecules (CAMs), receptors for cytokines (e.g., tumor necrosis factor-α), Toll-like receptors, and chemokines (eotaxin, macrophage inflammatory protein 1α, and interleukin 8) presenting multiple mechanisms for the inflammatory and remodeling process [71]. Additionally, one characteristic event of the airway remodeling is the ASM cells migration toward the epithelium [72]. Since ASM cells are crucial in asthma, Zuyderduyn et al. suggested that these cells should be targeted, rather than targeting inflammation or dealing with the symptoms [73].

5.3. Angiogenesis

Accumulating evidences indicate that there is an abnormal elevation in the size and number of blood vessels, as well as microvessels vascular leakage within the bronchial tissue in remodeled airways [74]. It is assumed that VEGF strongly affects airways remodeling via its angiogenic effects, but the exact molecular mechanism linking the increase in the VEGF expression to remodeling of the airways has not been fully understood [75].

Correlation between angiogenesis and asthma severity has also been documented. Dense vascularity occurs in severe asthmatics, followed by moderate, and then finally mild asthmatics, who experience less angiogenesis events [76]. This pattern was also observed in fatal asthmatics compared with nonfatal asthmatics [77]. While current asthma therapeutics is not directly targeting vascular remodeling, recent trials investigate some anti-angiogenic therapies as a new approach for asthma. Yuksel et al. showed that Bevucizamab, which significantly neutralizes VEGF, results in a reduced thickening of lung epithelium, a reduced ASM, and a reduced basement membrane thickness compared with untreated ovalbumin (OVA)-challenged mice [78].

Advertisement

6. Therapies for asthma

Modern treatments for asthma have been tested and used since the early twentieth century [79]. However, the oldest documented drug for asthma dates back to ancient Egypt. Kyphi, an incense mixture drink, was used inside the temples by the priests as a multipurpose lung medicament. There was more than one recipe for Kyphi; each may include as many as 10 herbs [80]. Following this, about 4000 years ago, Atropa Belladonna alkaloids, also called “deadly nightshade” because of their poisonous properties (“Natural Medicinal Herbs”), were derived from the leaves of thorn-apple plant and smoked by the Indians as cough suppressant [82]. Till today, natural and synthesized entities related to the tropane alkaloids class are still widely used. This includes anticholinergics (e.g., natural atropine, hyoscyamine (the levo-isomer of atropine), acopolamine, and the synthetic Ipratropium Bromide and stimulants (e.g., cocaine and hydroxytropacocaine) [83]. In 1872, one of the first papers published on asthma states that rubbing the chest of asthmatics with chloroform liniment can resolve airway constriction [84]. Adrenergic stimulants were in use for asthma over 100 years ago. In 1901, the adrenaline isolated from sheep and oxen adrenal glands was used to treat asthma [85]. The first documented publication of adrenaline as a bronchodilator therapy for asthma was written in 1903 by James Burnett, a physician in Edinburgh [86]. One year later in 1904, adrenaline was synthesized in the laboratories of Friedrich Stolz and Henry Drysdale Dakin, independently [87].

As suggested by the Global Initiative for Asthma (GINA) [88], a five-level step-down approach is widely recognized among the medical practitioners (Figure 3). The GINA approach assigns two types of drug classes for managing asthma:

  • Relievers (bronchodilators) cause immediate dilatation effects on the airways obstruction, mainly by acting on lung’s smooth muscle.

  • Controllers (preventers) provide long-term control of symptoms, mainly by suppressing the underlying disease process.

Figure 3.

Stepwise approach for controlling asthma symptoms and minimizing future morbidity.

β2-agonists and anticholinergics are considered to be bronchodilator relievers. Asthma controllers include corticosteroids, anti-leukotrienes, and anti-IgE. Theophylline is casually classified as both a bronchodilator and a reliever. The following book section will briefly discuss each therapeutic class.

6.1. Corticosteroids

Nowadays, most popular protocols for managing asthma involve the use of corticosteroids and β-agonists [1]. Anti-inflammatory corticosteroids, which are one of most trusted treatments for asthma, were introduced in mid-twentieth century [79]. The principle mode of action of corticosteroids in asthma is through their direct anti-inflammatory effect in different white blood cells including T cells, mast cells, and eosinophils. Among leukocytes, corticosteroids suppress chemotaxis and adhesion, and prevent inflammatory cytokines recruitment [89]. In vitro, corticosteroids reduce human ASM proliferation directly [90] by stimulating p21 gene expression [91], an important regulator of cell cycle progression. Moreover, corticosteroids improve vast majority of vascular remodeling aspects in asthma, reducing angiogenesis, excess blood flow, and vascular leakage [92]. This is mainly mediated by decreasing VEGF activity within the airway wall cells [93].

Various studies describe contradicting effects of corticosteroids on the lung epithelial abnormalities in asthmatics. Dorscheid et al. [94] reported that Dexamethasone treatment resulted in increased epithelial apoptosis and shedding. Similar results were obtained when treating guinea pigs with Budesonide, which did not improve the tracheal epithelium [95]. By contrast, some in vivo studies showed that inhaled corticosteroid (ICS) treatment resulted in improvement of epithelial damage in severe asthmatics [96,97].

ICS has been used around for the past couple of decades. Its idea dates back to the nineteenth century when the hand-held glass bulb nebulizer was used; however, pressurized metered-dose inhaler (pMDI) came to the clinic in 1956. After seeing his daughter’s suffering while using the hand-held nebulizer, George Maison, a medical consultant at 3M Pharmaceuticals, had advocated the use of pMDI. In 1959, George Maison and Irvine Porush were awarded a patent on the first pMDI [98].

6.2 β-adrenergic agonists

Long-acting β-agonists (LABAs), for example, Formoterol [99] and Salmeterol [100], offer a longer period of bronchodilation compared to the short-acting beta agonists (SABAs), for example, Salbutamol [101] and Terbutaline [102]. LABAs persist in the airway tissues for long periods due to their lipophilic nature and they provide a good umbrella of asthma bronchodilation and control, particularly at night [99,100]. However, until recently, the medical literature lacked supporting studies reporting the positive effect of β2 agonists on the chronic airway remodeling [103]. Addition of a β-agonist to the corticosteroid therapy allows a “steroid-sparing” effect, that is, maintains asthma control using lower doses of corticosteroids [104]. LABAs are not used as monotherapies anymore and they must be used in combination with ICS [105], because there have been cases of severe exacerbations and death when LABAs are administrated solely.

6.3. Antimuscarinic agents

Inhaled antimuscarinic agents, also known as inhaled anticholinergics, are considered another alternative bronchodilator group to β-agonists. The bronchodilation effect is functionally mediated via muscarinic receptor subtypes M1, M2, and M3, although five muscarinic receptors have been revealed in the lungs M1, M2, M3, M4, and M5 [106]. It is widely known that parasympathetic stimulation via the vagus nerve leads to immediate smooth muscle contraction and mucus secretion in the airways [107]. It is also suggested that M receptors interact with β2-adrenergic receptors (ADRB2) on the airways smooth muscle, leading to a reduced bronchodilator response of the β-agonists [108]. For years, in both adults and children, short-acting antimuscarinic agents use, for example, Ipratropium [109], has been limited to acute asthma management, in addition to inhaled SABA [110, 111]. Long-acting antimuscarinic agents, for example, Tiotropium [112], appear to have more benefits in difficult-to-control asthma. Adding Tiotropium to the standard asthma therapy significantly reduces asthma symptoms and highly increases the clinical outcomes [113, 114].

6.4. Targeted therapies

Over the last 40 years, there has been a marked increase in the development of targeted treatments for asthma—anti-leukotrienes, anti-IgE, anti-interleukins, and anti-TNF-α [115]. Obviously, as more of the biological basis of asthma is uncovered, more effective targeted asthma treatments might be developed. The list of most recently published clinical trials covering the period from 1 January 2013 to 1 January 2016, as well as the list of currently ongoing registered clinical trials that has started since 2013 for the new asthma medications are summarized in Tables 1 and 2, respectively.

Clinical Trial Identifier Publication Title Phase  Drugs Outcome Responsible Party Reference
NCT01147744 Efficacy, safety, and tolerability of GSK2190915, a 5-lipoxygenase-activating protein inhibitor, in adults and adolescents with persistent asthma: a randomized dose-ranging study. Phase 2 GSK2190915
(5-lipoxygenase-activating protein inhibitor)
GSK2190915 30-mg efficacy was demonstrated in day-time symptom scores and day-time SABA use, compared with placebo.
No additional improvement on efficacy was gained by administration of greater doses than 30 mg.
GSK2190915 was well tolerated.
GlaxoSmithKline [1]
NCT00411814 A phase 1,
randomized,
placebo-
controlled,
dose-escalation
study of an
anti-IL-13
monoclonal
antibody in
healthy
subjects
and mild
asthmatics.
Phase 1 GSK679586
(anti-IL-13)
GSK679586 showed dose-dependent pharmacological activity in the lungs of mild intermittent asthmatic patients.
GSK679586 could be a potential therapeutic candidate for treatment of asthma.
GlaxoSmithKline [2]
NCT00659659) Effects of benralizumab on airway eosinophils in asthmatic patients with sputum eosinophilia. Phase 1 Benralizumab
(Anti-IL-5)
Single-dose I.V. and multiple-dose S.C. of benralizumab reduced eosinophil counts in airway mucosa/submucosa and sputum and decreases eosinophil counts in bone marrow and peripheral blood in asthmatic patients. MedImmune LLC [3]
NCT01007149 A proof-of-concept, randomized, controlled trial of omalizumab in patients with severe, difficult-to-control, nonatopic asthma. Phase 3 Omalizumab
(anti-IgE)
Omalizumab may have a therapeutic potential for treatment of severe nonatopic asthma. Novartis Pharmaceuticals [4]
NCT00971035 Dose-ranging study of lebrikizumab in asthmatic patients not receiving inhaled steroids. Phase 2 Lebrikizumab
(anti-IL-13)
Blocking IL-13 alone was insufficient to improve lung function in asthmatic patients. Genentech, Inc. [5]
NCT00873860 A phase II placebo-controlled study of tralokinumab in moderate-to-severe asthma. Phase 2 Tralokinumab
(anti-IL-13)
Safety profile of tralokinumab was acceptable with no serious adverse effects.
Although tralokinumab treatment was associated with improved lung function, no improvement in asthma control questionnaire score was observed.
MedImm une LLC [6]
NCT01018186 Safety and tolerability of the novel inhaled corticosteroid fluticasone furoate in combination with the β2-agonist vilanterol administered once daily for 52 weeks in patients ≥12 years old with asthma: a randomized trial. Phase 3 Fluticasone furoate (ICS) +
Vilanterol
(LABA)
Fluticasone furoate/Vilanterol (100/25 μg or 200/25 μg) administered once daily over 52 weeks was well tolerated by asthmatic patients aged ≥12 years.
The overall safety profile of Fluticasone furoate/Vilanterol did not reveal any serious adverse effects.
GlaxoSmithKline [7]
NCT00393952 Efficacy and safety
of fluticasone/
formoterol
combination
therapy in
patients with
moderate-to-
severe
asthma.
Phase 3 Fluticasone propionate
(ICS)
Formoterol fumarate
(LABA)
Fluticasone/formoterol combination therapy was an efficient alternative treatment option for moderate-to-severe asthmatic patients. SkyePharma AG [8]
NCT01691521 Mepolizumab treatment in patients with severe eosinophilic asthma. Phase 3 Mepolizumab
(anti-IL-5)
Administration of mepolizumab (I.V. or S.C.) significantly reduced asthma exacerbations and is associated with improvements in markers of asthma control. GlaxoSmithKline [9]
NCT00500539 Immunogenicity
and safety of
omalizumab
in pre-filled
syringes in
patients with
allergic
(IgE-mediated)
asthma.
Phase 3 Omalizumab
(anti-IgE)
Pre-filled syringe of omalizumab was not associated with immunogenicity. Novartis Pharmaceuticals [10]
NCT00781443 The effects of lebrikizumab in patients with mild asthma following whole lung allergen challenge. Phase 2 Lebrikizumab
(anti-IL-13)
Lebrikizumab reduced
the late asthmatic
response in subjects
with mild asthma.
Genentech, Inc. [11]
NCT01181895 Comparison of vilanterol, a novel long-acting beta-2 agonist, with placebo and a Salmeterol reference arm in asthma uncontrolled by inhaled corticosteroids. Phase 3 Vilanterol
(LABA)
The study failed to
show a therapeutic difference
between vilanterol and
placebo for the primary
end point. The magnitude
of placebo effect may be due
to increased compliance with
anti-inflammatory
therapy regimen
during the treatment period.
GlaxoSmithKline [12]
NCT01233284 Tiotropium Respimat® in asthma: a double-blind, randomized, dose-ranging study in adult patients with moderate asthma. Phase 2 Tiotropium
(LAMA)
Administration of
tiotropium
Respimat®
(Once-daily) add-on
to medium-dose
ICS improves
lung function
in symptomatic
patients with
moderate asthma,
and the largest
improvement was
with a dose of
5 μg.
Boehringer Ingelheim [13]
NCT00983658 OX40L blockade and allergen-induced airway responses in subjects with mild asthma. Phase 2 huMAb
OX40L
(anti-OX40L)
Anti-OX40L MAb decreased serum total IgE and airway eosinophils at 16 weeks post dosing, but there was no effect on allergen-induced airway responses. This may be due to the treatment duration or dose of antibody was insufficient to have an effect on the airway responses. Genentech, Inc. [14]
NCT00768079 A randomized
trial of
benralizumab,
an anti-interleukin
5 receptor α
monoclonal
antibody,
after acute
asthma.
Phase 2 Benralizumab
(Anti-IL-5)
A dose of benralizumab—when added to usual care—reduced the rate and severity of asthma exacerbations experienced over 12 weeks by subjects who presented to the emergency department with acute asthma. MedImmune LLC [15]
NCT01369017 IL-1 receptor antagonist reduces endotoxin-induced airway inflammation in healthy volunteers. Phase 1 Anakinra
(Anti-IL-1)
Anakinra effectively
reduced airway
neutrophilic
inflammation
with no
serious adverse
reactions in
a model of
inhaled
lipopolysaccharide
challenge.
Anakinra is
a potential
therapeutic
candidate for
treatment of
asthma.
University of North Carolina, Chapel Hill [16]

Table 1.

Summary of recent published clinical trials for new drugs used in the treatment of asthma (from 1 January 2013 to 1 January 2016).

Abbreviations: IL: Interleukin; IgE: Immunoglobulin E; TNF-α: Tumor necrosis factor – α; PDE: Phosphodiesterase enzyme; ICS: Inhaled corticosteroids; OCS: Oral corticosteroids; SABA: Short-acting β-agonists; LABA: Long-acting β-agonists; LAMA; Long-acting muscarinic antagonists.


Clinical Trial Identifier Title Phase  Drugs Start Date Purpose Study Type Recruitment Status Responsible Party
NCT019
07763
Phase III
study to
assess the
efficacy
and safety
of SOTB07
in asthma
patients
Phase 3 Placebo
SOTB07
Jan 2013 Assessment
of the
efficacy
and safety
of SOTB07
in asthma
patients.
Interve
ntional
Recr
uiting
SK
Chemicals
Co.,Ltd.
NCT023
88997
Treatment
with
Omalizumab
to improve
the asthmatic
response to
an experimental
infection
with
rhinovirus
Phase 2 Omalizumab
(anti-IgE)
Rhinovirus
(strain 16)
Feb 2013 Determination
of whether
anti-IgE
therapy will
lead to
decline in
inflammatory
biomarkers
prior to
virus
inoculation,
and thus
reduce the
severity
of clinical
manifestations
after an
experimental
human RV
challenge.
Interve
ntional
Recr
uiting
University
of Virginia
NCT019
02290
Study of
efficacy
and safety of
Brodalumab
compared
with placebo in
inadequately
controlled
asthma
subjects with
high
bronchodilator
reversibility
Phase 2 Placebo
Brodalumab
(Anti-IL-17)
May 2013 Determination
of the safety
and efficacy
of Brodalumab
(AMG 827).
Interve
ntional
Recr
uiting
Amgen
NCT018
36471
A study to
assess the
effect of
QAW039 in
nonatopic
asthmatic
patients
Phase 2 Placebo
QAW039
ICS
May 2013 Assessment
of the
clinical
effect of
QAW039 in
nonatopic
asthmatics
taking low-
dose ICS as
background
therapy.
Interve
ntional
Recr
uiting
Novartis
Pharmaceuticals
NCT019
55512
Effect of
Clopidogrel
on allergen
challenge
in asthma
Phase 2 Placebo
Clopidogrel
(platelets
P2Y12
receptor
blocker)
May 2013 Determination
if the drug
Clopidogrel
reduces
inflammation
following
breathing
in house
dust mite in
people with
mild asthma.
Interve
ntional
Recr
uiting
University
of Southampton
NCT017
05964
Intramuscular
epinephrine
as an
adjunctive
treatment
for severe
pediatric
asthma
exacerbation
Phase 4 Epinephrine
(IM)
Jun 2013 Determination
if IM
epinephrine
is an
effective
adjunct to
inhaled
β2-agonists
for children
with severe
asthma
exacerbation.
Interve
ntional
Recr
uiting
University
of Louisville
NCT018
68061
A study of
Lebrikizumab
in patients
with
uncontrolled
asthma on
inhaled
corticosteroids
and a second
controller
medication
Phase 3 Placebo
Lebrikizumab
(anti-IL-13)
Jul 2013 Evaluation
of the efficacy
and safety of
Lebrikizumab
in patients
with
uncontrolled
asthma despite
daily
administration
of ICS therapy
and at least 1-s
controller
medication.
Interve
ntional
Recr
uiting
Hoffmann-La Roche
NCT018
67125
A study of
Lebrikizumab
in patients
with
uncontrolled
asthma who
are on
inhaled
corticosteroids
and a
second
controller
medication
Phase 3 Placebo
Lebrikizumab
(anti-IL-13)
Jul 2013 Evaluation
of the
efficacy
and safety
of Lebriki
zumab in
patients
with
uncontrolled
asthma
despite
daily
treatment
with ICS
therapy
and at
least 1-s
controller
medication.
Interve
ntional
Recr
uiting
Hoffmann-
La Roche
NCT018
41281
L-arginine
in severe
asthma
patients
grouped
by exhaled
nitric
oxide
levels
Phase 2 Placebo
L-Arginine
(Nitric
oxide
precursor)
Aug 2013 Identifi
cation of
the benefit
from
supplemental
L-arginine
therapy
in adult
severe
asthma
cohort.
Interve
ntional
Recr
uiting
University
of
California,
Davis
NCT019
12872
Study to
assess the
efficacy
and safety
of Omalizumab
treatment
on ICS
reduction
for severe
IgE-
mediated
asthma
(MEXIC)
Phase 4 Omalizumab
(anti-IgE)
Budesonide
Formoterol
(LABA)
Nov 2013 Assessment
of the
efficacy
and safety
of
Omalizumab
treatment
during 12
months
to reduce
the use
of ICS in
pediatric
and adult
patients
with
severe
IgE-mediated
asthma
inadequately
controlled
with high
doses of
corticosteroids.
Interve
ntional
Recr
uiting
Novartis
Pharmaceuticals
NCT020
41221
Pharmacology
study of
Sun Pharma
Advanced
Research
Company
Limited’s S0597
Phase 1
Phase 2
Placebo
S0597
Jan 2014 Evaluation
of safety,
tolerability,
pharmacokinetics,
and
pharmacodynamics
of S0597
by oral
inhalation.
Interve
ntional
Not yet
recruiting
Sun Pharma
Advanced
Research
Company
Limited
NCT020
49294
Study of
the prednisone-
sparing
effect
of Xolair
(Omalizumab)
in patients
with Prednisone-
dependent
asthma with
eosinophilic
bronchitis
Phase 2
Phase 3
Omalizumab
(anti-IgE)
Placebo
Normal Saline
Mar 2014 Investigation
whether
addition of
Omalizumab
enables a
reduction
in the dose
of prednisone
in patients
with asthma
and
eosinophilic
bronchitis.
Interve
ntional
Recr
uiting
McMaster
University
NCT019
87492
A study of
Lebrikizumab
In patients
with
severe
asthma who
depend on
oral
corticosteroids
Phase 2 Placebo
Lebrikizumab
(anti-IL-13)
Mar 2014 Evaluation
of the
efficacy of
Lebrikizumab
compared
with
placebo as
measured
by the
ability of
patients
to achieve
lower
daily doses
of OCS in
patients
with severe
corticosteroid-
dependent
asthma.
Interve
ntional
Recr
uiting
Hoffmann-
La Roche
NCT020
75008
Long-term
safety
study of
QGE031
in patients
with allergic
asthma who
completed
study
CQGE031
B2201
Phase 2 QGE031 Mar 2014 Assessment
of long-term
safety of
QGE031 during
12 months of
treatment in
asthma
patients who
completed
study
CQGE031B2201.
Interve
ntional
Recr
uiting
Novartis
Pharmaceuticals
NCT020
75255
Efficacy
and safety
study of
Benralizumab
to reduce
OCS use in
patients
with
uncontrolled
asthma
on high-dose
inhaled
corticosteroid
plus LABA
and
chronic
OCS therapy
Phase 3 Placebo
Benralizumab
(anti-IL-5)
Apr 2014 This trial
is to
confirm if
Benralizumab
can reduce
OCS dependence
(after dose
optimization)
in patients
who are
uncontrolled
on high-dose
ICS-LABA,
and
chronically
dependent
on OCS as
part of their
regular
asthma
controller
regimen.
Interve
ntional
Recr
uiting
AstraZeneca
NCT021
35692
A Phase 3a,
repeat dose,
open-label,
long-term
safety study
of Mepolizumab
in asthmatic
subjects
Phase 3 Mepolizumab
(anti-IL-5)
Standard
of Care
May 2014 Collection
of clinical
data for
long-term
use and
further
assessment
of efficacy
in patients
with loss
of asthma
control.
Interve
ntional
Recr
uiting
GlaxoSmithKline
NCT021
26865
Multiple
rising
oral doses
of BI
1060469
in healthy
subjects
and mild
asthma
patients
Phase 1 Placebo
BI 1060469
May 2014 Investigation
of the safety
and
tolerability
of repeated
rising
doses of BI
1060469
in healthy
male and
female subjects
and in
asthmatic male
and female
patients.
Interve
ntional
Recr
uiting
Boehringer
Ingelheim
NCT021
61757
A Phase 3
study to
evaluate the
efficacy and
safety of
Tralokinumab
in adults and
adolescents
with
uncontrolled
asthma
(STRATOS1)
Phase 3 Placebo
Tralokinumab
(Anti-IL-13)
Jun 2014 Evaluation
of the
efficacy
and safety of
Tralokinumab
in adults
and adolescents
with
asthma
inadequately
controlled
on ICS plus
long-acting
β2-agonist.
Interve
ntional
Recr
uiting
AstraZeneca
NCT021
04674
A study
evaluating
the efficacy
and safety of
Lebrikizumab
in adult
patients with
mild to
moderate
asthma
Phase 3 Placebo
Lebrikizumab
(anti-IL-13)
Montelukast
Jun 2014 Assessment
of the efficacy
and safety
of Lebrikizumab
in adult
patients with
mild to
moderate
asthma
treated
with SABA
therapy
alone.
Interve
ntional
Recr
uiting
Hoffmann-
La Roche
NCT020
66298
Steroids In
eosinophil-
negative
asthma
(SIENA)
Phase 3 Placebo
Mometasone
Tiotropium
(LAMA)
Jul 2014 Determination
if patients
who are
persistently
non-
eosinophilic
differ in
their
benefit
from inhaled
corticosteroid
treatment
compared to
patients who
are not
persistently
non-
eosinophilic.
Interve
ntional
Recr
uiting
Milton S.
Hershey
Medical
Center
NCT020
99656
A study
evaluating
the effects of
Lebrikizumab
on airway
eosinophilic
inflammation
in patients
with
uncontrolled
asthma
Phase 2 Placebo
Lebrikizumab
(anti-IL-13)
Nov 2014 Evaluation
of the
effects of
Lebrikizumab
on airway
eosinophilic
inflammation
in patients
with
uncontrolled
asthma on
inhaled
corticosteroids
and a second
controller
medication.
Interve
ntional
Recr
uiting
Hoffmann-
La Roche
NCT022
58542
A safety
extension
study to
evaluate
the safety
and
tolerability
of
Benralizumab
(MEDI-563)
in asthmatic
adults and
adolescents
on inhaled
corticosteroid
plus LABA
(BORA)
Phase 3 Benralizumab
(anti-IL-5)
Nov 2014 Characterization
of safety
profile of
Benralizumab
administration
in asthma
patients
who have
completed
one of the
three
predecessor
studies:
D3250C00017,
D3250C00018,
or
D3250C00020.
Interve
ntional
Recr
uiting
AstraZeneca
NCT022
96411
Efficacy
of LAMA
added to
ICS in
treatment
of asthma
(ELITRA)
Phase 2 Placebo
CHF 5259
Glycopyrrolate
bromide
(LAMA)
Nov 2014 Evaluation
of the
safety and
superiority
of the
glycopyrrolate
bromide
(CHF 5259
pMDI) versus
placebo on
top of
QVAR® pMDI,
in terms of
lung
functions
parameters.
Interve
ntional
Recr
uiting
Chiesi
Farmaceutici
S.p.A.
NCT022
93265
Cross-
sectional
study for
identification
and
description
of severe
asthma
patients
Phase 3 Mepolizumab
(anti-IL-5)
Omalizumab (anti-IgE)
Reslizumab
(anti-IL-5)
Dec 2014 The
potential
overlap of
patients
eligible
for
treatment
with
Mepolizumab
,
Omalizumab
and/or
Reslizumab
will be
estimated.
Interve
ntional
Recr
uiting
GlaxoSmithKline
NCT022
81318
Efficacy
and safety
study of
Mepolizumab
adjunctive
therapy in
participants
with severe
eosinophilic
asthma on
markers of
asthma
control
Phase 3 Placebo
Mepolizumab
(anti-IL-5)
Standard
of Care
Dec 2014 Evaluation
of the
efficacy
and safety
of Mepolizumab
adjunctive
therapy in
participants
with severe
eosinophilic
asthma on
markers of
asthma
control.
Interve
ntional
Recr
uiting
GlaxoSmithKline
NCT023
22775
Study
to evaluate
the efficacy
and safety of
Benralizumab
in adult
patients with
mild to
moderate
persistent
asthma
Phase 3 Placebo
Benralizumab
(anti-IL-5)
Feb 2015 Confirmation
of the safety
and clinical
benefit of
Benralizumab
administration
in asthma
patients
with mild
to moderate
persistent
asthma.
Interve
ntional
Recr
uiting
AstraZeneca
NCT023
82510
Multiple
ascending
dose study
of TRN-157
in stable
mild and
moderate
asthmatics
Phase 2 Placebo
TRN-157
Tiotropium
(LAMA)
Feb 2015 Determination
of the
safety and
bronchodilator
activity
of TRN-157 in
approximately
54 mild and
moderate
asthmatics.
Interve
ntional
Recr
uiting
Theron
Pharmaceuticals,
Inc.
NCT023
15131
Study in
healthy
volunteers
and COPD
patients
to evaluate
the efficacy
and safety
of inhaled
TV46017
Phase 1 Placebo
TV46017
Mar 2015 Assessment
of the
safety
profile
and
duration of
bronchodilation
of a single
dose of
inhaled
TV46017.
Interve
ntional
Not yet
recruiting
Teva
Branded
Pharmaceutical
Products
, R&D Inc.
NCT021
24226
Low-dose
Methotrexate
for reduction
GINA 5
medications
in chronic
severe
asthma
Phase 3 Placebo
Methotrexate
Apr 2015 Investigation
of the role
of an add-on
immunological
modifier in
patients
with chronic
severe
asthma.
Interve
ntional
Not yet
recruiting
Universita
degli
Studi
di Catania
NCT023
77427
Pharmaco
kinetics
and
pharmaco
dynamics
of Mepolizumab
administered
subcutaneously
in children
Phase 2 Mepolizumab
(anti-IL-5)
Apr 2015 Assessment
of the
pharmacokinetics
and
pharmacodynamics
of Mepolizumab
in children
aged 6–11
years with
severe
eosinophilic
asthma.
Interve
ntional
Not yet
recruiting
GlaxoSmithKline
NCT023
36425
Efficacy
and safety
of QGE031
compared
with
placebo
in patients
aged 18–75
years
with asthma
Phase 2 Placebo
QGE031
Apr 2015 The study
will assess
the safety
and efficacy
of different
dose levels
of QGE031 in
asthma
patients.
Interve
ntional
Not yet
recruiting
Novartis
Pharmaceuticals
NCT024
27165
Comparison
of RPL554
With
placebo and
Salbutamol
in asthmatic
patients
Phase 2 Placebo
RPL554
(PDE-3/4
inhibitor)
Salbutamol
Apr 2015 Assessment
of the
effects
of RPL554
compared
with
Salbutamol
and placebo
in patients
with
chronic
asthma.
Interve
ntional
Not yet
recruiting
Verona
Pharma plc
NCT024
22121
Effect
of RNS60
on the
late-phase
asthmatic
response
to allergen
challenge
Phase 2 Placebo
RNS60
Budesonide
May 2015 Evaluation
of the
effects of
multiple
doses
of inhaled
RNS60 and
Budesonide
on the
late-phase asthmatic
response
to allergen
challenge
in patients
with mild
asthma.
Interve
ntional
Not yet
recruiting
Revalesio
Corporation
NCT025
71660
Efficacy
of vitamin
D on the
clinical
management
of pediatric
patients
with
asthma
Phase 3 Vitamin
D (Low-
and high-
supplemen
tation
doses)
Oct 2015 Evaluation
of vitamin D
supplemen
tation
on exacerbation
and clinical
control
of asthma.
Interven
tional
Not yet
recruiting
Hospital
General
Naval de
Alta
Especialidad -
Escuela
Medico
Naval

Table 2.

Summary of recent ongoing clinical trials for new drugs used in the treatment of asthma (started in the past 3 years).

Abbreviations denote: IL: Interleukin; IgE: Immunoglobulin E; PDE: Phosphodiesterase enzyme; ICS: Inhaled corticosteroids; OCS: Oral corticosteroids; SABA: Short-acting β-agonists; LABA: Long-acting β-agonists; LAMA; Long-acting muscarinic antagonists.


6.4.1. Anti-leukotrienes

Leukotrienes are lipid eicosanoids with a wide range of biological activities. They are derived from arachidonic acid through the enzymatic action of 5-lipooxygenase, and play a crucial role in asthma inflammatory pathogenesis, and in other allergic diseases such as allergic rhinitis, rhinosinusitis, atopic dermatitis, and urticaria [116]. Leukotrienes class includes three main types: cysteinyl leukotrienes (CysLTs), LTB4, and LTG4. LTG4 is the metabolite of LTE4 in which the cysteinyl moiety has been oxidized to an α-keto-acid [117]. Since, very little is known about the LTG4-putative leukotriene, most clinical research studies focus on CysLTs and LTB4. CysLTs are strong bronchoconstrictors that powerfully affect airway remodeling, whereas LTB4 is a strong chemoattractant for most leukocyte subsets [118]. Over the last 20 years, since leukotriene antagonists were introduced to the clinic for asthma management, Montelukast [119, 120] and Zafirlukast [121] are the most frequently used drugs in this class.

6.4.2. Anti-IgE

At the moment, Omalizumab, which is the only approved targeted monoclonal antibody against IgE, is used to treat allergic asthma in clinical practice. It can significantly decrease serum IgE levels (up to 99%) within 2 h following subcutaneous administration, and diminish serum, sputum, and tissue eosinophilia [122]. Recently, Omalizumab has also been reported to have steroid-sparing effect, reducing the rate of asthma exacerbations up to 50%, and hence improving the quality of life [123]. However, nearly 45% of patients treated with Omalizumab had adverse reaction at the local injection site, which is considered the most commonly observed adverse event for Omalizumab. Some other minor upper respiratory tract infections and sinusitis have also been reported as well. Patients treated with Omalizumab display a very low (0.09%) frequency of anaphylaxis reaction. Importantly, there are no data reporting any correlation between cancer and Omalizumab treatment [124].

6.4.3. Anti-ILs

Three interleukin pathways are of physiological importance for asthma: IL-5, IL-9, and IL-4/IL-13 pathways. IL-5 is pivotal for both eosinophil differentiation and maturation in the bone marrow. Subsequently, it controls eosinophil mobilization, activation, and survival [125]. Hence, antagonizing IL-5 has been proposed to be beneficial for asthma therapy, particularly for predominantly eosinophilic asthma. A number of anti-IL-5 and anti-IL-5 receptor monoclonal antibodies are in the process of development for allergic diseases: Reslizumab [126], Mepolizumab [127], and Benralizumab [128]. IL-9 is one of the T-helper 2 (Th2) pro-inflammatory cytokines that promote mast cell proliferation and T-cell growth [129]. In mouse models, IL-9 causes several common features of chronic asthma: excessive mucus production, eosinophilic airway inflammation, smooth-muscle cell hyperplasia, and aryl hydrocarbon receptor (AHR) [130]. Currently, a phase IIb clinical trial evaluates the efficacy and safety of subcutaneous Medi-528, a humanized IgG1 anti-IL-9 mAb, in adults with uncontrolled asthma (NCT00968669). Activated mast cells, eosinophils, basophils, and dendritic cells secrete IL-4 and IL-13. IL-4 and IL-13 both play an important role in asthma mainly by enhancing IgE production. They also control mast cells’ growth and development, eosinophil recruitment, and AHR [131]. The first trial aimed at antagonizing the IL-4 used a soluble recombinant human IL-4 receptor antagonist (IL-4RA), altrakincept, which blocked the binding of IL-4 to its cellular receptors [132]. Several humanized IL-13-neutralizing antibodies have entered asthma phase I/II clinical trials—anrukinzumab [133], QAX576 [134], and CAT354 [135].

6.4.4. Anti-TNF-α

TNF-α, a cytokine produced by Th1 cells and macrophages, has diverse biological functions. TNF-α shows crucial, and previously extensively documented, role in Crohn’s disease, rheumatoid arthritis, and psoriasis pathogenesis. The association between TNF-α increase and these disease progressions had inspired studies aiming to extend anti-TNF-α therapies also for the treatment of severe asthma and COPD [136]. Infliximab and Golimumab, two anti-TNF-α mAbs, and Etanercept, a decoy soluble TNF-α receptor, are both able to biologically neutralize TNF-α cytokine, and blunt the immune response, thereby abolishing TNF-α effects in asthma [137].

Advertisement

7. Pharmacogenetics of asthma

The US Food and Drug Administration definition for pharmacogenomics is “the study of variations in DNA and RNA characteristics as related to drug response” [138]. “Pharmacogenomics” differs from “Pharmacogenetics” in that the former is concerned with the whole genome, its components, and regulators, while the latter is focused only on the DNA sequences of individual gene. Thus, in sense, pharmacogenetics is thought to be a subset of pharmacogenomics [139].

Because it is a complex trait, the drug response to asthma is diversely heterogeneous even among patients with apparently similar clinical profiles [7]. It is estimated that up to 50% difference in therapeutic response has been attributed to genetic variations between individuals [140]. Although several possible mechanisms have been postulated, genetic variants affect the pharmacogenetic response to drugs in two different ways:

  1. Pharmacodynamic genetic variations are variations in which the receptor binding the drug ligand or another member of the drug target pathway is altered resulting in different drug effect. Most of the current pharmacogenetic research fall into this mechanistic category. Populations are stratified into responders and nonresponders, and then analyzed for DNA polymorphisms, which distinguish these two groups apart.

  2. Pharmacokinetic genetic variations are related to altered uptake, distribution, and/or metabolism of the administered drug. Fewer examples fall in this category; however, the most common research subfield here is the area of investigating drug-catabolizing or -excreting enzymes. An important example here is the cytochrome P450 (CYP450) family, a widely recognized metabolizing enzyme with several variable pharmacogenetic patterns.

Single nucleotide polymorphism, SNP, denoted by a reference sequence (rs) number, represents a class of polymorphism that is derived from a one-base point mutation in which a single nucleotide is substituted with another one. SNPs may be located in the gene regulatory or coding regions, and so it may affect the gene expression in more than one way; however, in majority of cases, most discovered SNPs do not change the gene function in a significant manner [141]. Consequently, it is essential to investigate whether the DNA sequence variances would actually cause significant functional impacts (i.e., resulting in an altered observed biology), or is a linkage disequilibrium marker of another DNA variant, which is the real cause of the response variability, or is generally nonsignificant. Because of its strong importance, since 15 years, catalogs of SNPs have started to outline the most common genetic polymorphisms among different population groups [141, 142], and this process has attracted more attention during the last couple of years [143].

Figure 4.

Pharmacogenetically significant genes with relevance to corticosteroids, β-adrenergic, and leukotriene biological pathways. Left side: Candidate gene approach studies, Right side: GWAS (Genome Wide association studies).

All genes contain huge number of SNPs and copy-number variations (CNVs). CNVs are another form of structural variations, which account for 13% of the human genome bulkiness, and manifest as kilo-to-mega bases of deletions or duplications [144]. Conjointly, it is challenging to outline which polymorphism is influencing the treatment response and which are not relevant. Two major approaches declaiming this challenge have been practiced so far: candidate gene approach and GWAS. As it combines transcriptomic, proteomic and metabolomic profiling traits, a third approach, the integrative system biology approach, had led to a more comprehensive pharmacogenetic view [3]. To differentiate, candidate gene approach is based on a prior evidence according to the knowledge of the drug pharmacodynamics or/and pharmacokinetics, by contrast, GWAS methodology identifies new associations with the null hypothesis being that no associations exist. GWAS picks the variations which are associated with observable phenotypes by scanning SNP markers that tag, via linkage disequilibrium, the complete human genome. GWAS and integrative system biology approaches are modern tools contributing to the recent advancements of genotyping and statistical technologies.

Current pharmacogenetic studies of the corticosteroids, β-adrenergic, and leukotriene pathways are mostly candidate gene studies, with some GWAS, however, altogether have identified several genetic loci in strong association with therapeutic responsiveness to asthma. Figure 4 summarizes the pharmacogenetically significant genes with relevance to the corticosteroids, β-adrenergic, and leukotriene biological pathways.

7.1. Corticosteroid pathway pharmacogenetics

In cytosol, the glucocorticoids bind to their corresponding glucocorticoid receptor, forming a hetero-complex that is activated by ligand binding, and translocate into the nucleus. In the nucleus, this complex binds to the glucocorticoid response elements in some target genes’ promoter region resulting in their expression regulation. The core role of glucocorticoids is mediated via activating the transcription of anti-inflammatory genes, and suppressing the transcription of pro-inflammatory genes [145, 146]. The glucocorticoid pharmacogenetic studies formerly focused on candidate gene approach. Those candidate genes covered functions related to the corticosteroid biosynthetic pathway, the hetero-complex receptor formation, and the related chaperone proteins.

Corticotrophin-releasing hormone (CRHR1), stress-inducible protein 1 (STIP1), TBX21, CYP3A4, GLCCI1, T gene, and FBXL7 are the most up-to-date potential pharmacogenetic biomarker targets for predicting patients’ response to ICS [147]. Studies of the corticotrophin-releasing hormone gene are considered to be as one of the oldest and remarkable footsteps in asthma pharmacogenetics. CRHR1 protein, also known as CRF1, is the primary receptor controlling the adrenocorticotropic hormone release; hence, it plays a pleiotropic and vital role in steroid actions. A candidate gene study of CRHR1 in 1117 asthmatics administrating ICS therapy, from three clinical cohorts, revealed two SNPs (rs242941 and rs1876828) associated with different response in lung functions [148]. Tantisira et al. [148] found that CRHR1 gene variation was frequently related to augmented therapy response in each of the three studied cohorts. Since 2004, CRHR1 gene studies opened the doors for all other corticosteroid pharmacogenetics and the possible future therapeutic outcomes.

STIP1 or HOP (abbreviated for Hsp70-Hsp90-Organizing Protein) gene mainly functions to reversibly link Hsp70 and Hsp90 together as a co-chaperone [149]. STIP1 pharmacogenetic studies in one adult cohort revealed three SNPs (rs2236647, rs6591838, and rs1011219) within this heat shock-organizing protein and related to improved lung response during ICS therapy [150]. STIP1 rs2236647 variant analysis in healthy and asthmatic children showed that this SNP could serve as an asthma marker for choosing the population who receives corticosteroid therapy[151]; however, further replication studies should be held to confirm those results.

One significant aspect of pharmacogenomics is that it investigates the interactions with genes of other pathways. TBX21 gene is one good example for observing the ICS response outside the glucocorticoid pathway. TBX21 is one of the conserved genes of a family sharing a common DNA-binding domain; the T-box encodes T-box transcription factor TBx21 protein. Tbx21 protein is a Th1 (T-helper1) transcription factor, which regulates one of the Th1 cytokine expression, interferon-gamma (IFNG). In 2004, a nonsynonymous SNP rs2240017 (His33Glu) in the TBX21 gene was linked to improvements in bronchial hyperresponsiveness or “broncho-protection” in response to ICS in individuals participating in the Child Asthma Management Program (CAMP) cohort [152]. This finding was also observed in an independent Korean cohort in 2009 [153]. Thus, TBx21 may be an important determinant pharmacogenetic candidate gene for predicting asthmatics’ response to inhaled corticosteroid therapies.

In 2005, another example demonstrated the glucocorticoid pathway interactions with one other pathway. ADCY9, adenylyl cyclase type 9, gene encodes a membrane-bound enzyme in the β2-adrenergic receptor pathway, which catalyzes the production of cyclic adenosine monophosphate (AMP) from adenosine triphosphate (ATP). This candidate gene contains a pharmacogenetic nonsynonymous SNP, Met772Ile, which was correlated to enhanced Salbutamol (SABA) bronchodilator effects only in patients treated with ICS [154]. An independent Korean cohort replicated the trial, using Formoterol (LABA) treatment in combination with ICS, and confirmed those results [155].

Cytochromes P450s belong to a heme cofactor-containing superfamily of metabolizing enzyme proteins that potentially control the metabolism of drug (i.e., pharmacokinetics), and consequently treatment response in many diseases. For asthma, CYP3A4, CYP3A5, and CYP3A7 candidate genes have been studied among a retrospective analysis of 413 asthmatic children treated with the ICS Fluticasone propionate [156]. The three candidate CYPs of all subjects were genotyped for nine SNPs. Results showed that asthmatics with the CYP3A4*22 allele demonstrated a significant symptom control compared with those lacking that allele. This study included a small number of participants (n = 20), so further large-scale replication is required.

Tantisira et al. [157] conducted the first pharmacogenetic GWAS for ICS treatment in asthma and identified an SNP (rs37972) in the promoter of the glucocorticoid-induced transcript-1 gene (GLCCI1), which significantly associates with lung functions. Replicated in four independent populations (935 persons in total), this candidate SNP was linked to substantial decrements in the response to the ICS in asthmatics. The wild-type allele homozygotes (CC) showed greater forced expiratory volume in 1 s (FEV1) in response to the ICS compared with those identified with the homozygote variant allele (TT). Another functionally correlated SNP (rs37973) in the promoter of the same gene was further validated within in vitro studies [157]. Results showed declined luciferase reporter activity in cells with the minor allele. GLCCI1 GWAS outlines that drug response to asthma treatment is subjected to wide inter-individual variation, and GWAS would uncover more novel pharmacogenetic associations in the future. Tantisira et al. conducted a second GWAS among 418 asthmatics randomized to ICS treatment from the Childhood Asthma Research and Education (CARE), Asthma Clinical Research Network (ACRN), and CAMP trial cohorts. The T-gene (encoding the Brachyury transcription factor protein) compromised two SNPs (rs3127412 and rs6456042) that were associated, out of the successfully genotyped 47 SNPs, with altered lung function response to ICS [158].

7.2. β-adrenergic receptor pathway pharmacogenetics

β2-adrenergic receptor gene remains to be the most studied pharmacogenetic loci among the beta-agonist pathways. ADRB2 gene has several polymorphic variants that were discovered in multi-ethnic genetic asthma cohorts [159, 160]. ADRB2 protein is a cell membrane-spanning receptor that binds epinephrine, but not norepinephrine, unlike the other adrenergic receptors, and consequently mediates both smooth muscle relaxation and bronchodilation [161, 162]. Early ADRB2 studies showed that Gly16Arg, a prevalent coding variant of the amino acid at position 16 of ADRB2, is associated with altered bronchodilator response to SABAs [163].

The BARGE (Beta-Agonist Response by Genotype) study [164], held by the National Heart, Lung and Blood Institute Asthma Clinical Network, was one of the first genotype “stratified” pharmacogenetic studies for asthma. In this study, only Gly16Arg homozygotes for ADRB2 were included (i.e., Arg/Arg and Gly/Gly). Participants were randomly receiving either intermittent or regular albuterol, and then crossed over to receive the alternative treatment dose. For statistical stratification, this study ensured that the Arg16 homozygotes, who are less frequent, were appropriately randomly distributed to both SABA intermittent and regular protocols. Compared to Gly16 homozygotes, the BARGE study showed that the Arg16 homozygotes were good responders only to acute intermittent SABAs rather than to long-term regular treatments, a finding that does not coincide with the current clinical asthma treatment guidelines [165] which recommend SABA as for on-demand intermittent usage. Since the 16th amino acid of ADRB2 controls regular response to albuterol, bronchodilator medications other than SABAs would be more appropriate for Arg/Arg asthmatics.

Collectively, the BARGE study [164], along with some other pharmacogenetic studies [163, 166168] of Gly16Arg and SABAs’ exposure, provided insights for further studies [169171] on LABAs. In contrast to SABAs, a large cohort [169] of 2250 asthmatics, randomly assigned to formoterol plus budesonide, demonstrated no pharmacogenetic action due to ADRB2 variation on therapeutic response. Furthermore, a multicenter trial [170] showed that asthmatics with both Arg/Arg and Gly/Gly genetic signatures had improved airway functions, if they received combination treatment with Salmeterol and ICS, when compared with ICS therapy alone. Similarly, the results of another prospective trial cohort [171] of 544 subjects, also randomized by genotyping, demonstrated no evidence of any pharmacogenetic action due to ADRB2 variation in response to Salmeterol. Together, these findings, confirmed among several asthma populations, suggest that in contrast to SABAs, asthmatics can still be treated with LABAs plus ICS irrespective of their genotyping status.

Genetic variants’ occurrences among different ethnic groups are quantified by their percentage of allele frequencies. Usually, frequent and common variants have only little or modest impacts on disease susceptibility and, subsequently, therapeutic response. On the other hand, as the variant is characterized to be rare or more “private,” its effect size on disease progression and therapeutic response dramatically increases [172]. Early in vitro studies had investigated a rare polymorphism of ADRB2 within the fourth transmembrane domain, the Thr164Ile variant. For the Ile164 genotype, results showed significant lowering in Gs-protein signaling and different SABA- and LABA-binding affinities [173, 174]. While the Thr164Ile polymorphism is pointed out to be a rare coding variant (i.e., <5%), population studies showed that this variant is more common in non-Hispanic white populations [159, 160], a finding that requires further pharmacogenetic investigation in different and larger populations. To replicate results, a study of two large Copenhagen population cohorts [175], with more than 55,000 participants, was held to investigate the relation of Thr164Ile variation and lung responses. Among the general population, the Copenhagen study reported that the Thr164 genotype was associated with decreased FEV1, diminished lung function, and increased the overall COPD risk.

In addition to ADRB2 Gly16Arg and Thr164Ile variants, the (-376 In-Del) polymorphism was extensively studied as another significant pharmacogenetic ADRB2 variant. Presented primarily among African Americans and Puerto Ricans [159, 160], the 24-bp promoter insertion at −376, related to the start codon, is associated with asthma-related hospitalization in asthmatics treated with LABA [160]. Altogether, these variants, being unique to different populations, highlight the increasing need of personalized-based treatments.

Adenylyl cyclase type 9, encoded in humans by ADCY9 gene, is a membrane-bound enzyme that catalyzes the formation of cyclic AMP from ATP. ADCY9 is a widely abundant adenylyl cyclase, and it is stimulated via beta-adrenergic receptor activation [176]. Ile772Met is a coding variant of ADCY9 gene that has been associated with both acute FEV1 bronchodilation in response to SABAs [154] and long-term FEV1 response for LABAs [155]. CRHR2 (which is more commonly known as CRF2) is a type-2 G protein-coupled protein receptor for the corticotropin-releasing hormone [177]. Out of the 28 studied SNPs in CRHR2, five SNPs were significantly correlated with acute bronchodilator response in one, or frequently more than one, cohort. Among those, variant rs7793837 was associated with altered SABA response in all three cohorts of the CRHR2 study containing 607, 427, and 152 participants, respectively [178].

Different variants of ARG1 (Arginase 1) and ARG2 (Arginase 2) show altered acute response to SABAs, while the endothelial nitric oxide synthase (NOS3) shows altered acute response to LABAs. NO (nitric oxide), an endogenous vasorelaxing bronchodilator, is generated by the action of NOS3 on L-arginine. Since ARG1 and ARG2 are metabolizing L-arginine, so it is expected that the entire three genes, ARG1, ARG2, and NOS3, might be implicated in asthma pharmacogenetics. Combined association evidence, surviving Bonferroni correction for multiple testing from the CAMP four asthma cohorts [179], points to SNP rs2781659 in ARG1. C-allele homozygotes for SNP rs2781667 in arginase 1 showed significantly less response to the inhaled corticosteroid treatments [180]. Arginase-2 variants rs17249437 and rs3742879 correlated with increased airway obstruction and airway hyperresponsiveness, and lower reversibility of airway constriction following treatment with beta-2 agonists [180]. A small candidate gene study [181] of NOS3 had revealed one possible variant (Asp298Glu) correlated with lung function response to ICS/LABA combined treatment; however, this result still needs to be replicated in larger cohorts. THRB [182], SLC24A4 [183], SLC22A15 [183], SPATS2L [184], and SNPs (rs892940, rs77441273, rs1281748/rs1281743, and rs295137, respectively) show promising loci for further pharmacogenetic investigations.

7.3. Leukotriene pathway pharmacogenetics

Relative to the corticosteroid and β-adrenergic pathways, the cysteinyl leukotriene pathway pharmacogenetic studies are generally fewer and have smaller sample sizes. The oldest of these studies [185], held in 1999, had investigated the tandem repeat polymorphism in ALOX5 promoter. Among 114 asthmatics, it has been shown that the ALOX5 promoter repeat is associated with altered lung functions in response to a 5-LO inhibitor [185]. It has been shown in children that those who had more or less than five repeats (3, 4, and 6) of the ALOX5 promoter-binding motif experienced increased urinary leukotriene E4 (the terminal cysteinyl leukotriene metabolite) concentrations and reduced FEV1 baseline than the wild-type genotype with five repeats [186]. Further pharmacogenetic studies revealed that the ALOX5 promoter polymorphism, along with the ALOX5 SNPs rs892690, rs2029253, and rs2115819, influences leukotriene pathway antagonist therapy [187190]. Moreover, variants of LTC4S, encoding Leukotriene C4 synthase, and MRP1 (or ABCC1), encoding multidrug resistance-associated protein 1, have been linked to lung function response while treatment with Zileuton and Montelukast [189, 190].

Arg312Gln, rs12422149, which is a coding variant in SLCO2B1 (solute carrier organic anion transporter family member 2B1 gene), has been related to symptom control during Montelukast therapy. This fact was due to the interindividual variability of carrier-mediated Montelukast transport in the intestines, and consequently its plasma levels [191]. By contrast, two other studies, probably due to their small sample sizes, were unable to replicate similar SLCO2B1 pharmacokinetic effects [192, 193]. Overall, larger replicate cohorts, for the leukotriene pathway identified loci, are still needed.

Advertisement

8. Current and future challenges facing asthma pharmacogenetics

As demonstrated above, there has been fundamental progress in the field of asthma pharmacogenetics; however, these efforts have not yet been introduced into clinical practice to guide physician. There are several reasons that account for this gap. Most important is the limited number of asthma pharmacogenetics-focused GWAS, which would compare common candidate gene methodology that would allow combining all patients from small cohorts studied. Small sample sizes prevent any expansion of the pharmacogenetic research of asthma, which needs a large number of subjects for statistical significance. Along with limited cohort size, study defects due to poor ancestry structuring and stratification substantially result in replication inconsistencies. Furthermore, genes interact together in networks; therefore, simply attributing phenotypic variation to individual genes is not appropriate. Epigenetics studies investigate the changes in gene activities, which are heritable to the subsequent generations, but are independent of any DNA sequence alterations [194, 195]. Epigenetic tuning of the genes associated with asthma has a significant impact on determining the drug response. Several mechanisms, related to epigenetics, are currently being investigated for both biomarker tagging and therapeutic innovation intervention [196]. Moreover, epigenetic changes have the ability to override the genetic effects of time, environment, tissue specificity, and other conditions such as age and gender of a patient, nutrition and hygiene, and intestinal microflora, which all highly influence the drug response in addition to the genetic factors. The collective impact of all combination of these factors requires the application of complicated algorithm that could take into consideration each of these factors and their interplay. The prospective genetic profile of an asthmatic should compromise a set of common and rare variants, on ancestral basis, which will be predictive of the pattern of his/her therapeutic responsiveness to different treatment options. The current human variant catalog continues to grow in an exponential manner because of the lower costs associated with whole genomic sequencing. Despite the steep decline in sequencing costs, the technology of sequencing, in terms of speed and quality, enormously increases. The future pharmacogenetic profile would also predict any possible adverse response associated with the chosen line of treatment. Genetic biomarkers are needed to warn the physician about any potential adverse side effects which can be life threatening. It is very important for typical genetic profiling to also consider gene-gene and gene-environment interactions. Gene-gene interactions are predominately crucial in the framework of combination therapies, for example, ICS and β-adrenergic agonists. Interactions between the surrounding environment and the patients’ genes are assumed to be an additional element, because environmental stress, apart from the genetic makeup, contributes to the development of asthma exacerbations. Future pharmacogenetic directions need to cover also the pharmacokinetic side of the patient profile. Altered drug absorption, metabolism, distribution, or excretion extensively influence drug dosing and even drug selection. All in all, the complete asthma pharmacogenetic catalog has many aspects to cover, before being introduced into the clinical practice.

Advertisement

9. Conclusion

Asthma is a complex respiratory and immune disease. Inadequate (or exaggerated) ability of genetically predisposed individuals to control inflammation, induced by innate and environmental factors, results in asthma. Further, studies using allergic asthma and atopy models enable to better understand several interacting gene products and variable responsiveness of asthmatic subjects to current therapies. Eventually, thorough investigation of the complexity of asthma might lead to successful designing of personalized therapies for patients suffering from allergic asthma.

References

  1. 1. Nanzer AM, Menzies-Gow A. Defining severe asthma – an approach to find new therapies. Eur Clin Respir J. 2014;1(5):1–9.
  2. 2. Nakamura Y. Developmental current and future therapy for severe asthma. Inflamm Allergy-Drug Targets. 2013;12(1):54–60.
  3. 3. Park H-W, Tantisira KG, Weiss ST. Pharmacogenomics in asthma therapy: where are we and where do we go? Annu Rev Pharmacol Toxicol. 2015;55(1):129–47.
  4. 4. World Health Organ. (WHO). Bronchial asthma. WHO Fact Sheet 206, World Health Organ. G. No Title [Internet]. Available from: http://www.who.int/mediacentre/factsheets/fs206/en/. Accessed January 2016.
  5. 5. Sossai P, Travaglione AM, Amenta F. Asthma: opinion or evidence based medicine? Clinical Medicine and Diagnositics. 2014;4(2):17-22.
  6. 6. Bergeron C, Boulet L-P. Structural changes in airway diseases. Chest J. 2006;129(4):1068–87.
  7. 7. Szefler SJ, Martin RJ, King TS, Boushey HA, Cherniack RM, Chinchilli VM, et al. Significant variability in response to inhaled corticosteroids for persistent asthma. J Allergy Clin Immunol. 2002;109(3):410–8.
  8. 8. International HapMap Consortium. The International HapMap Project. Nature. 2003 Dec 18;426(6968):789–96.
  9. 9. International HapMap 3 Consortium, Altshuler DM, Gibbs RA, Peltonen L, Altshuler DM, Gibbs RA, et al. Integrating common and rare genetic variation in diverse human populations. Nature. 2010;467(7311):52–8.
  10. 10. Bleecker E, Ortega V, Meyers D. Asthma pharmacogenetics and the development of genetic profiles for personalized medicine. Pharmgenomics Pers Med. 2015;8(16):9–22.
  11. 11. Meyers DA, Bleecker ER, Holloway JW, Holgate ST. Asthma genetics and personalised medicine. Lancet Respir Med. 2014;2(5):405–15.
  12. 12. Duffy DL, Martin NG, Battistutta D, Hopper JL, Mathews JD. Genetics of asthma and hay fever in Australian twins. Am Rev Respir Dis. 1990;142(6 Pt 1):1351–8.
  13. 13. Ober C, Yao TC. The genetics of asthma and allergic disease: a 21st century perspective. Immunol Rev. 2011;242(1):10–30.
  14. 14. Li X, Ampleford EJ, Howard TD, Moore WC, Torgerson DG, Li H, et al. Genome-wide association studies of asthma indicate opposite immunopathogenesis direction from autoimmune diseases. J Allergy Clin Immunol. 2012;130(4):861–8.e7.
  15. 15. Van Eerdewegh P, Little RD, Dupuis J, Del Mastro RG, Falls K, Simon J, et al. Association of the ADAM33 gene with asthma and bronchial hyperresponsiveness. Nature. 2002;418(6896):426–30.
  16. 16. Yoshinaka T, Nishii K, Yamada K, Sawada H, Nishiwaki E, Smith K, et al. Identification and characterization of novel mouse and human ADAM33s with potential metalloprotease activity. Gene. 2002;282(1–2):227–36.
  17. 17. Tripathi P, Awasthi S, Gao P. ADAM metallopeptidase domain 33 (ADAM33): a promising target for asthma. Mediat Inflamm. 2014;2014:5720–5.
  18. 18. March ME, Sleiman PM, Hakonarson H. Genetic polymorphisms and associated susceptibility to asthma. Int J Gen Med. 2013;6(17):253–65.
  19. 19. Altshuler D, Daly MJ, Lander ES. Genetic mapping in human disease. Science. 2008;322(5903):881–8.
  20. 20. McCarthy MI, Abecasis GR, Cardon LR, Goldstein DB, Little J, Ioannidis JPA, et al. Genome-wide association studies for complex traits: consensus, uncertainty and challenges. Nat Rev Genet. 2008;9(5):356–69.
  21. 21. Moffatt MF, Kabesch M, Liang L, Dixon AL, Strachan D, Heath S, et al. Genetic variants regulating ORMDL3 expression contribute to the risk of childhood asthma. Nature. 2007;448(7152):470–3.
  22. 22. Torgerson DG, Ampleford EJ, Chiu GY, Gauderman WJ, Gignoux CR, Graves PE, et al. Meta-analysis of genome-wide association studies of asthma in ethnically diverse North American populations. Nat Genet. 2011;43(9):887–92.
  23. 23. Karunas AS, Iunusbaev BB, Fedorova II, Gimalova GF, Ramazanova NN, Gur'eva LL, et al. Genome-wide association study of bronchial asthma in the Volga-Ural region of Russia. Mol Biol (Mosk). 2011;45(6):992–1003.
  24. 24. Wan YI, Shrine NRG, Soler Artigas M, Wain L V., Blakey JD, Moffatt MF, et al. Genome-wide association study to identify genetic determinants of severe asthma. Thorax. 2012;67(9):762–8.
  25. 25. Breslow DK, Collins SR, Bodenmiller B, Aebersold R, Simons K, Shevchenko A, et al. Orm family proteins mediate sphingolipid homeostasis. Nature. 2010;463(7284):1048–53.
  26. 26. Worgall TS, Veerappan A, Sung B, Kim BI, Weiner E, Bholah R, et al. Impaired sphingolipid synthesis in the respiratory tract induces airway hyperreactivity. Sci Transl Med. 2013;5(186):186ra67.
  27. 27. Moffatt MF, Gut IG, Demenais F, Strachan DP, Bouzigon E, Heath S, et al. A large-scale, consortium-based genomewide association study of asthma. N Engl J Med. 2010;363(13):1211–21.
  28. 28. Gudbjartsson DF, Bjornsdottir US, Halapi E, Helgadottir A, Sulem P, Jonsdottir GM, et al. Sequence variants affecting eosinophil numbers associate with asthma and myocardial infarction. Nat Genet. 2009;41(3):342–7.
  29. 29. Weidinger S, Gieger C, Rodriguez E, Baurecht H, Mempel M, Klopp N, et al. Genome-wide scan on total serum IgE levels identifies FCER1A as novel susceptibility locus. PLoS Genet. 2008;4(8):e1000166.
  30. 30. Postma DS, Reddel HK, ten Hacken NHT, van den Berge M. Asthma and chronic obstructive pulmonary disease: similarities and differences. Clin Chest Med. Elsevier Inc; 2014;35(1):143–56.
  31. 31. Postma DS, Kerkhof M, Boezen HM, Koppelman GH. Asthma and chronic obstructive pulmonary disease: common genes, common environments? Am J Respir Crit Care Med. 2011;183(12):1588–94.
  32. 32. Celedón JC, Milton DK, Ramsey CD, Litonjua AA, Ryan L, Platts-Mills TAE, et al. Exposure to dust mite allergen and endotoxin in early life and asthma and atopy in childhood. J Allergy Clin Immunol. 2007;120(1):144–9.
  33. 33. Balaban J, Bijelic R, Milicevic S. Hypersensitivity to aeroallergens in patients with nasobronchial allergy. Med Arch. 2014;68(2):86.
  34. 34. Gruchalla RS, Pongracic J, Plaut M, Evans R, Visness CM, Walter M, et al. Inner city asthma study: relationships among sensitivity, allergen exposure, and asthma morbidity. J Allergy Clin Immunol. 2005;115(3):478–85.
  35. 35. Arbes SJ, Gergen PJ, Vaughn B, Zeldin DC. Asthma cases attributable to atopy: results from the third national health and nutrition examination survey. J Allergy Clin Immunol. 2007;120(5):1139–45.
  36. 36. Kelly JT, Busse WW. Host immune responses to rhinovirus: mechanisms in asthma. J Allergy Clin Immunol. 2008;122(4):671–82.
  37. 37. Wu P, Dupont WD, Griffin MR, Carroll KN, Mitchel EF, Gebretsadik T, et al. Evidence of a causal role of winter virus infection during infancy in early childhood asthma. Am J Respir Crit Care Med. 2008;178(11):1123–9.
  38. 38. Darveaux JI, Lemanske RFJ. Infection-related asthma. J Allergy Clin Immunol Pract. 2014;2(6):658–63.
  39. 39. Newcomb DC, Peebles RS. Bugs and asthma: a different disease? Proc Am Thorac Soc. 2009;6(3):266–71.
  40. 40. Koyanagi K, Koya T, Sasagawa M, Hasegawa T, Suzuki E, Arakawa M, et al. An analysis of factors that exacerbate asthma, based on a Japanese questionnaire. Allergol Int. 2009;58(4):519–27.
  41. 41. Beasley R, Semprini A, Mitchell EA. Asthma 1 risk factors for asthma : is prevention possible ? Lancet. 2015;386(9998):1075–85.
  42. 42. Greer JR, Abbey DE, Burchette RJ. Asthma related to occupational and ambient air pollutants in nonsmokers. J Occup Med. 1993;35(9):909–15.
  43. 43. Havemann BD, Henderson CA, El-Serag HB. The association between gastro-oesophageal reflux disease and asthma: a systematic review. Gut. 2007;56(12):1654–64.
  44. 44. Jaspersen D, Kulig M, Labenz J, Leodolter A, Lind T, Meyer-Sabellek W, et al. Prevalence of extra-oesophageal manifestations in gastro-oesophageal reflux disease: an analysis based on the ProGERD Study. Aliment Pharmacol Ther. 2003;17(12):1515–20.
  45. 45. Sharifi A, Ansarin K. Effect of gastroesophageal reflux disease on disease severity and characteristics of lung functional changes in patients with asthma. J Cardiovasc Thorac Res. 2014;6(4):223–8.
  46. 46. Wright RJ, Cohen S, Carey V, Weiss ST, Gold DR. Parental stress as a predictor of wheezing in infancy: a prospective birth-cohort study. Am J Respir Crit Care Med. 2002;165(3):358–65.
  47. 47. Chen E, Hanson MD, Paterson LQ, Griffin MJ, Walker HA, Miller GE. Socioeconomic status and inflammatory processes in childhood asthma: the role of psychological stress. J Allergy Clin Immunol. 2006;117(5):1014–20.
  48. 48. Moy ML, Lantin ML, Harver A, Schwartzstein RM. Language of dyspnea in assessment of patients with acute asthma treated with nebulized albuterol. Am J Respir Crit Care Med. 1998;158(3):749–53.
  49. 49. Virchow JC, Crompton GK, Dal Negro R, Pedersen S, Magnan A, Seidenberg J, et al. Importance of inhaler devices in the management of airway disease. Respir Med. 2008;102(1):10–9.
  50. 50. Prins LC, van Son MJ, van Keimpema AR, van Ranst D, Pommer A. Psychopathology in difficult asthma. J Asthma. 2015;(19):1–6.
  51. 51. Thorowgood JC. On bronchial asthma. BMJ. 1873;2(673):600–600.
  52. 52. Bosworth FH. Hay fever, asthma, and allied affections. Trans Am Climatol Assoc Meet. 1886;2(1):151–70.
  53. 53. Gerthoffer WT, Solway J, Camoretti-Mercado B. Emerging targets for novel therapy of asthma. Curr Opin Pharmacol. Elsevier Ltd; 2013;13(3):324–30.
  54. 54. Yayan J, Rasche K. Asthma and COPD: Similarities and Differences in the Pathophysiology, Diagnosis and Therapy. In Respiratory Medicine and Science 2015 (pp. 31–38). Springer International Publishing.
  55. 55. Pauwels RA, Buist AS, Calverley PM, Jenkins CR, Hurd SS. Global strategy for the diagnosis, management, and prevention of chronic obstructive pulmonary disease. NHLBI/WHO Global Initiative for Chronic Obstructive Lung Disease (GOLD) Workshop summary. Am J Respir Crit Care Med. 2001;163(5):1256–76.
  56. 56. Chiba S, Tsuchiya K, Nukui Y, Sema M, Tamaoka M, Sumi Y, et al. Interstitial changes in Asthma-COPD Overlap Syndrome (ACOS). Clin Respir J. 2016; (Epub ahead of print). ((Still Epub. Not assigned to issue yet))
  57. 57. Stanley PJ, Wilson R, Greenstone MA, Mackay IS, Cole PJ. Abnormal nasal mucociliary clearance in patients with rhinitis and its relationship to concomitant chest disease. Br J Dis Chest. 1985;79(1):77–82.
  58. 58. Flenley DC. Breathing during sleep. Ann Acad Med Singapore. 1985;14(3):479–84.
  59. 59. Overholt RH, Voorhees RJ. Esophageal reflux as a trigger in asthma. Dis Chest. 1966;49(5):464–6.
  60. 60. Boulet LP. Influence of comorbid conditions on asthma. Eur Respir J. 2009;33(4):897–906.
  61. 61. Auerbach O, Stout AP, Hammond EC, Garfinkel L. Changes in bronchial epithelium in relation to cigarette smoking and in relation to lung cancer. N Engl J Med. 1961;265(1):253–67.
  62. 62. Holgate ST. Pathogenesis of asthma. Clin Exp Allergy. 2008;38(6):872–97.
  63. 63. Flood-Page P, Menzies-Gow A, Phipps S, Ying S, Wangoo A, Ludwig MS, et al. Anti-IL-5 treatment reduces deposition of ECM proteins in the bronchial subepithelial basement membrane of mild atopic asthmatics. J Clin Invest. 2003;112(7):1029–36.
  64. 64. Roche W, Williams J, Beasley R, Holgate S. Subepithelial fibrosis in the bronchi of asthmatics. Lancet. 1989;1(8637):520–4.
  65. 65. Desmouliere A, Darby IA, Laverdet B, Bonté F. Fibroblasts and myofibroblasts in wound healing. Clin Cosmet Invest Dermatol. 2014;6(7):301–11.
  66. 66. Hamid Q, Song Y, Kotsimbos TC, Minshall E, Bai TR, Hegele RG, et al. Inflammation of small airways in asthma. J Allergy Clin Immunol. 1997;100(1):44–51.
  67. 67. Hostettler KE, Roth M, Burgess JK, Gencay MM, Gambazzi F, Black JL, et al. Airway epithelium-derived transforming growth factor-beta is a regulator of fibroblast proliferation in both fibrotic and normal subjects. Clin Exp Allergy. 2008;38(8):1309–17.
  68. 68. Ebina M, Takahashi T, Chiba T, Motomiya M. Cellular hypertrophy and hyperplasia of airway smooth muscles underlying bronchial asthma. A 3-D morphometric study. Am Rev Respir Dis. 1993;148(3):720–6.
  69. 69. Niimi A, Matsumoto H, Takemura M, Ueda T, Chin K, Mishima M. Relationship of airway wall thickness to airway sensitivity and airway reactivity in asthma. Am J Respir Crit Care Med. CRC Press; 2003;168(8):983–8.
  70. 70. Kazi AS, Lotfi S, Goncharova EA, Tliba O, Amrani Y, Krymskaya VP, et al. Vascular endothelial growth factor-induced secretion of fibronectin is ERK dependent. 2004;(3):L539–45.
  71. 71. Joubert P, Hamid Q. Role of airway smooth muscle in airway remodeling. J Allergy Clin Immunol. Mosby; 2005;116(3):713–6.
  72. 72. Al-Muhsen S, Johnson JR, Hamid Q. Remodeling in asthma. J Allergy Clin Immunol. Elsevier Ltd; 2011;128(3):451–62.
  73. 73. Zuyderduyn S, Sukkar MB, Fust A, Dhaliwal S, Burgess JK. Treating asthma means treating airway smooth muscle cells. Eur Respir J. 2008;32(2):265–74.
  74. 74. Harkness LM, Ashton AW, Burgess JK. Asthma is not only an airway disease, but also a vascular disease. Pharmacol Ther. 2015;148(1):17–33.
  75. 75. Smith R. Is VEGF a potential therapeutic target in asthma? Pneumologia. 2013;63(4):194–7.
  76. 76. Salvato G. Quantitative and morphological analysis of the vascular bed in bronchial biopsy specimens from asthmatic and non-asthmatic subjects. Thorax. 2001;56(12):902–6.
  77. 77. Carroll NG, Cooke C, James AL. Bronchial blood vessel dimensions in asthma. Am J Respir Crit Care Med. 1997;155(2):689–95.
  78. 78. Yuksel H, Yilmaz O, Karaman M, Bagriyanik HA, Firinci F, Kiray M, et al. Role of vascular endothelial growth factor antagonism on airway remodeling in asthma. Ann Allergy Asthma Immunol. 2013;110(3):150–5.
  79. 79. Chu EK, Drazen JM. Asthma one hundred years of treatment and onward. Am J Respir Crit Care Med. 2005;171(11):1202–8.
  80. 80. Manniche L. Sacred luxuries: fragrance, aromatherapy, and cosmetics in ancient Egypt. Cornell University Press, Ithaca, USA; 1999. p. 49
  81. 81. http://www.naturalmedicinalherbs.net/ [Internet]. Available from: http://www.naturalmedicinalherbs.net/ Accessed January 2016
  82. 82. Lane DJ SA, editor. Bronchodilators. In: Asthma: the facts. 2nd ed. New York, NY: Oxford University Press; 1987. pp. 126–40.
  83. 83. O'Neil MJ. The Merck index. 15th ed. O'Neil MJ, editor. Merck Sharp & Dohme Corp. Whitehouse Station, N.J., U.S.A.; 2013.
  84. 84. Gaskoin G. On the treatment of asthma. Br Med J. 1872;1:339.
  85. 85. Takamine J. The blood-pressure raising principle of the suprarenal gland. J Am Med Assoc. American Medical Association; 1902;38(3):153–5.
  86. 86. Burnett J. Adrenalin: a short account of its therapeutic applications. Med Times Hosp Gaz. 1903;23(1):385–7.
  87. 87. Bennett MR. One hundred years of adrenaline: the discovery of autoreceptors. Clin Auton Res. 1999;9(3):145–59.
  88. 88. Global Initiative for Asthma (GINA) [Internet]. From the Global Strategy for Asthma Management and Prevention. 2015. Available from: http://www.ginasthma.org/ Accessed January 2016
  89. 89. Heitzer MD, Wolf IM, Sanchez ER, Witchel SF, DeFranco DB. Glucocorticoid receptor physiology. Rev Endocr Metab Disord. 2007;8(4):321–30.
  90. 90. Stewart AG, Fernandes D, Tomlinson PR. The effect of glucocorticoids on proliferation of human cultured airway smooth muscle. Br J Pharmacol. 1995;116(8):3219–26.
  91. 91. Cha HH, Cram EJ, Wang EC, Huang AJ, Kasler HG, Firestone GL. Glucocorticoids stimulate p21 gene expression by targeting multiple transcriptional elements within a steroid responsive region of the p21(waf1/cip1) promoter in rat hepatoma cells. J Biol Chem. 1998;273(4):1998–2007.
  92. 92. Chetta A, Olivieri D. Role of inhaled steroids in vascular airway remodelling in asthma and COPD. Int J Endocrinol. 2012;10(2):397693.
  93. 93. Feltis BN, Wignarajah D, Reid DW, Ward C, Harding R, Walters EH. Effects of inhaled fluticasone on angiogenesis and vascular endothelial growth factor in asthma. Thorax. 2007;62(4):314–9.
  94. 94. Dorscheid DR, Low E, Conforti A, Shifrin S, Sperling AI, White SR. Corticosteroid-induced apoptosis in mouse airway epithelium: effect in normal airways and after allergen-induced airway inflammation. J Allergy Clin Immunol. 2003;111(2):360–6.
  95. 95. Erjefält JS, Erjefält I, Sundler F, Persson CG. Effects of topical budesonide on epithelial restitution in vivo in guinea pig trachea. Thorax. 1995;50(7):785–92.
  96. 96. Lundgren R, Soderberg M, Horstedt P, Stenling R. Morphological studies of bronchial mucosal biopsies from asthmatics before and after ten years of treatment with inhaled steroids. Eur Respir J. 1988;1(10):883–9.
  97. 97. Laitinen LA, Laitinen A, Haahtela T. A comparative study of the effects of an inhaled corticosteroid, budesonide, and a beta 2-agonist, terbutaline, on airway inflammation in newly diagnosed asthma: a randomized, double-blind, parallel-group controlled trial. J Allergy Clin Immunol. 1992;90(1):32–42.
  98. 98. I . Porush GLM. Self-propelling compositions for inhalation therapy containing a salt of isoproterenol or epinephrine. United States of America; US Patent 2,868,691, 1959. p. US Patent 2,868,691.
  99. 99. Arvidsson P, Larsson S, Löfdahl CG, Melander B, Wåhlander L, Svedmyr N. Formoterol, a new long-acting bronchodilator for inhalation. Eur Respir J Off J Eur Soc Clin Respir Physiol. 1989;2(4):325–30.
  100. 100. Johnson M. Salmeterol: a novel drug for the treatment of asthma. Agents Actions Suppl. 1991;34(2):79–95.
  101. 101. Cullum VA, Farmer JB, Jack D, Levy GP. Salbutamol: a new, selective beta-adrenoceptive receptor stimulant. Br J Pharmacol. 1969;35(1):141–51.
  102. 102. Persson H, Olsson T. Some pharmacological properties of terbutaline (INN), 1-(3,5-dihydroxyphenyl)-2-(T-butylamino)-ethanol. A new sympathomimetic beta-receptor-stimulating agent. Acta Med Scand Suppl. 1970;512(7):11–9.
  103. 103. Berair R, Brightling CE. Asthma therapy and its effect on airway remodelling. Drugs. 2014;74(6):1345–69.
  104. 104. Bowler S. Long acting beta agonists. Aust Fam Phys. 1998;27(12):1115–8.
  105. 105. Nelson HS, Weiss ST, Bleecker EK, Yancey SW, Dorinsky PM. The salmeterol multicenter asthma research trial: a comparison of usual pharmacotherapy for asthma or usual pharmacotherapy plus salmeterol. Chest. 2006;129(1):15–26.
  106. 106. Buels KS, Fryer AD. Muscarinic receptor antagonists: effects on pulmonary function. Handb Exp Pharmacol. 2012;208(3):317–41.
  107. 107. Barnes PJ. Muscarinic receptor subtypes in airways. Life Sci. 1993;52(5–6):521–7.
  108. 108. Proskocil BJ, Fryer AD. Beta2-agonist and anticholinergic drugs in the treatment of lung disease. Proc Am Thorac Soc. 2005;2(4):305–10; discussion 311–2.
  109. 109. Engelhardt A, Klupp H. The pharmacology and toxicology of a new tropane alkaloid derivative. Postgrad Med J. 1975;51(7 SUPPL):82–5.
  110. 110. Powell CV, Cranswick NE. The current role of ipratropium bromide in an acute exacerbation of asthma. J Paediatr Child Health. 2015;51(8):751–2.
  111. 111. Ward MJ, Fentem PH, Smith WH, Davies D. Ipratropium bromide in acute asthma. Br Med J (Clin Res Ed). 1981;282(6264):598–600.
  112. 112. Disse B, Reichl R, Speck G, Traunecker W, Ludwig Rominger KL, Hammer R. Ba 679 BR, a novel long-acting anticholinergic bronchodilator. Life Sci. 1993;52(5-6):537–44.
  113. 113. Kerstjens HAM, Engel M, Dahl R, Paggiaro P, Beck E, Vandewalker M, et al. Tiotropium in asthma poorly controlled with standard combination therapy. N Engl J Med. 2012;367(13):1198–207.
  114. 114. Peters SP, Kunselman SJ, Icitovic N, Moore WC, Pascual R, Ameredes BT, et al. Tiotropium bromide step-up therapy for adults with uncontrolled asthma. N Engl J Med. 2010;363(18):1715–26.
  115. 115. Menzella F, Lusuardi M, Galeone C, Zucchi L, Article I, Url A, et al. Tailored therapy for severe asthma. Multidiscip Respir Med. 2015;10(1):1.
  116. 116. Peters-Golden M, Henderson WR Jr. Leukotrienes. N Engl J Med. 2007;357(18):1841–54.
  117. 117. Tomisawa H, Takanohashi Y, Ichihara S, Fukazawa H, Tateishi M. Transamination of LTE4 by cysteine conjugate aminotransferase. Biochem Biophys Res Commun. 1988;155(3):1119–25.
  118. 118. Liu M, Yokomizo T. The role of leukotrienes in allergic diseases. Allergol Int. 2015;64(1):17–26.
  119. 119. Jones TR, Labelle M, Belley M, Champion E, Charette L, Evans J, et al. Pharmacology of montelukast sodium (Singulair), a potent and selective leukotriene D4 receptor antagonist. Can J Physiol Pharmacol. 1995;73(2):191–201.
  120. 120. Reiss TF, Altman LC, Chervinsky P, Bewtra A, Stricker WE, Noonan GP, et al. Effects of montelukast (MK-0476), a new potent cysteinyl leukotriene (LTD4) receptor antagonist, in patients with chronic asthma. J Allergy Clin Immunol. 1996;98(3):528–34.
  121. 121. Spector, Sheldon L. “Management of asthma with zafirlukast.” Drugs. 1996;52(6):36–46.
  122. 122. Ames SA, Gleeson CD, Kirkpatrick P. Omalizumab. Nat Rev Drug Discov. 2004;3(3):199–200.
  123. 123. Rodrigo GJ, Neffen H, Castro-Rodriguez JA. Efficacy and safety of subcutaneous omalizumab vs placebo as add-on therapy to corticosteroids for children and adults with asthma: a systematic review. Chest. 2011;139(1):28–35.
  124. 124. Polosa R, Casale T. Monoclonal antibodies for chronic refractory asthma and pipeline developments. Drug Discov Today. 2012;17(11–12):591–9.
  125. 125. Leckie MJ, Ten Brinke A, Khan J, Diamant Z, O’Connor BJ, Walls CM, et al. Effects of an interleukin-5 blocking monoclonal antibody on eosinophils, airway hyper-responsiveness, and the late asthmatic response. Lancet. 2000;356(9248):2144–8.
  126. 126. Castro M, Zangrilli J, Wechsler ME, Bateman ED, Brusselle GG, Bardin P, et al. Reslizumab for inadequately controlled asthma with elevated blood eosinophil counts: results from two multicentre, parallel, double-blind, randomised, placebo-controlled, phase 3 trials. Lancet Respir Med. 2015;3(5):355–6.
  127. 127. Haldar P, Brightling CE, Singapuri A, Hargadon B, Gupta S, Monteiro W, et al. Outcomes after cessation of mepolizumab therapy in severe eosinophilic asthma: a 12-month follow-up analysis. J Allergy Clin Immunol. 2014;133(3):921–3.
  128. 128. Nowak RM, Parker JM, Silverman RA, Rowe BH, Smithline H, Khan F, et al. A randomized trial of benralizumab, an antiinterleukin 5 receptor α monoclonal antibody, after acute asthma. Am J Emerg Med. 2015;33(1):14–20.
  129. 129. Levitt RC, McLane MP, MacDonald D, Ferrante V, Weiss C, Zhou T, et al. IL-9 pathway in asthma: new therapeutic targets for allergic inflammatory disorders. J Allergy Clin Immunol. 1999;103(5 Pt 2):S485–91.
  130. 130. Oh CK, Raible D, Geba GP, Molfino NA. Biology of the interleukin-9 pathway and its therapeutic potential for the treatment of asthma. Inflamm Allergy Drug Targets. 2011;10(3):180–6.
  131. 131. Oh CK, Geba GP, Molfino N. Investigational therapeutics targeting the IL-4/IL-13/STAT-6 pathway for the treatment of asthma. Eur Respir Rev. 2010;19(115):46–54.
  132. 132. Borish LC, Nelson HS, Lanz MJ, Claussen L, Whitmore JB, Agosti JM, et al. Interleukin-4 receptor in moderate atopic asthma. A phase I/II randomized, placebo-controlled trial. Am J Respir Crit Care Med. 1999;160(6):1816–23.
  133. 133. Reinisch W, Panés J, Khurana S, Toth G, Hua F, Comer GM, et al. Anrukinzumab, an anti-interleukin 13 monoclonal antibody, in active UC: efficacy and safety from a phase IIa randomised multicentre study. Gut. 2015;64(6):894–900.
  134. 134. Rothenberg ME, Wen T, Greenberg A, Alpan O, Enav B, Hirano I, et al. Intravenous anti–IL-13 mAb QAX576 for the treatment of eosinophilic esophagitis. J Allergy Clin Immunol. 2015;135(2):500–7.
  135. 135. Murray LA, Zhang H, Oak SR, Coelho AL, Herath A, Flaherty KR, et al. Targeting interleukin-13 with tralokinumab attenuates lung fibrosis and epithelial damage in a humanized SCID idiopathic pulmonary fibrosis model. Am J Respir Cell Mol Biol. 2014;50(5):985–94.
  136. 136. Russo C, Polosa R. TNF-alpha as a promising therapeutic target in chronic asthma: a lesson from rheumatoid arthritis. Clin Sci (Lond). 2005;109(2):135–42.
  137. 137. Matera MG, Calzetta L, Cazzola M. TNF-alpha inhibitors in asthma and COPD: we must not throw the baby out with the bath water. Pulm Pharmacol Ther. 2010;23(2):121–8.
  138. 138. US Dep. Health Hum. Serv., Food Drug Admin. [Internet]. Guidance for Industry: E15 Definitions for Genomic Biomarkers, Pharmacogenomics, Pharmacogenetics, Genomic Data and Sample Coding Categories. 2008. Available from: http://www.fda.gov/downloads/drugs/guidancecomplianceregulatoryinformation/guidances/ucm073162.pdfucm073162.pdf. Accessed January 2016.
  139. 139. Ma Q, Lu AYH. Pharmacogenetics, Pharmacogenomics, and individualized medicine. Pharmacol Rev. 2011;63(2):437–59.
  140. 140. Palmer LJ, Silverman ES, Weiss ST, Drazen JM. Pharmacogenetics of asthma. Am J Respir Crit Care Med. 2002;165(7):861–6.
  141. 141. Gray IC, Campbell DA, Spurr NK. Single nucleotide polymorphisms as tools in human genetics. Hum Mol Genet. 2000;9(16):2403–8.
  142. 142. Palmer LJ, Cookson WO. Using single nucleotide polymorphisms as a means to understanding the pathophysiology of asthma. Respir Res. 2001;2(2):102–12.
  143. 143. Chaudhary R, Singh B, Kumar M, Gakhar SK, Saini AK, Parmar VS, et al. Role of single nucleotide polymorphisms in pharmacogenomics and their association with human diseases. Drug Metab Rev. 2015;47(3):1–10.
  144. 144. Stankiewicz P, Lupski JR. Structural variation in the human genome and its role in disease. Annu Rev Med. 2010;61(2):437–55.
  145. 145. Revollo JR, Cidlowski JA. Mechanisms generating diversity in glucocorticoid receptor signaling. Ann N Y Acad Sci. 2009;1179(4):167–78.
  146. 146. Newton R, Holden NS. Separating transrepression and transactivation: a distressing divorce for the glucocorticoid receptor? Mol Pharmacol. 2007;72(4):799–809.
  147. 147. Ting F Leung, FAAAAI MFT and GWW. Personalized medicine for severe asthma: how far have we achieved? Pharmacogenomics Pharmacoproteomics. 2015;6(2):1.
  148. 148. Tantisira KG, Lake S, Silverman ES, Palmer LJ, Lazarus R, Silverman EK, et al. Corticosteroid pharmacogenetics: association of sequence variants in CRHR1 with improved lung function in asthmatics treated with inhaled corticosteroids. Hum Mol Genet. 2004;13(13):1353–9.
  149. 149. Odunuga OO, Longshaw VM, Blatch GL. Hop: more than an Hsp70/Hsp90 adaptor protein. Bioessays. 2004;26(10):1058–68.
  150. 150. Hawkins GA, Lazarus R, Smith RS, Tantisira KG, Meyers DA, Peters SP, et al. The glucocorticoid receptor heterocomplex gene STIP1 is associated with improved lung function in asthmatic subjects treated with inhaled corticosteroids. J Allergy Clin Immunol. 2009;123(6):1376–83.e7.
  151. 151. Einisman H, Reyes ML, Angulo J, López-Lastra M, Cerda J, JAC-R. Use of steroid receptor related STIP1 gene analysis as an asthma marker. Eur Respir J. 2012;40(56):1782–5.
  152. 152. Tantisira KG, Hwang ES, Raby BA, Silverman ES, Lake SL, Richter BG, et al. TBX21: a functional variant predicts improvement in asthma with the use of inhaled corticosteroids. Proc Natl Acad Sci U S A. 2004;101(52):18099–104.
  153. 153. Ye Y-M, Lee H-Y, Kim S-H, Jee Y-K, Lee S-K, Lee S-H, et al. Pharmacogenetic study of the effects of NK2R G231E G>A and TBX21 H33Q C>G polymorphisms on asthma control with inhaled corticosteroid treatment. J Clin Pharm Ther. 2009;34(6):693–701.
  154. 154. Tantisira KG, Small KM, Litonjua AA, Weiss ST, Liggett SB. Molecular properties and pharmacogenetics of a polymorphism of adenylyl cyclase type 9 in asthma: interaction between beta-agonist and corticosteroid pathways. Hum Mol Genet. 2005;14(12):1671–7.
  155. 155. Kim SH, Ye YM, Lee HY, Sin HJ, Park HS. Combined pharmacogenetic effect of ADCY9 and ADRB2 gene polymorphisms on the bronchodilator response to inhaled combination therapy. J Clin Pharm Ther. 2011;36(3):399–405.
  156. 156. Stockmann C, Fassl B, Gaedigk R, Nkoy F, Uchida DA, Monson S, et al. Fluticasone propionate pharmacogenetics: CYP3A4*22 polymorphism and pediatric asthma control. J Pediatr. 2013;162(6):1227.e1–2.
  157. 157. Tantisira KG, Lasky-Su J, Harada M, Murphy A, Litonjua AA, Himes BE, et al. Genomewide association between GLCCI1 and response to glucocorticoid therapy in asthma. N Engl J Med. 2011;365(13):1173–83.
  158. 158. Tantisira KG, Damask A, Szefler SJ, Schuemann B, Markezich A, Su J, et al. Genome-wide association identifies the T gene as a novel asthma pharmacogenetic locus. Am J Respir Crit Care Med. 2012;185(12):1286–91.
  159. 159. Hawkins GA, Tantisira K, Meyers DA, Ampleford EJ, Moore WC, Klanderman B, et al. Sequence, haplotype, and association analysis of ADRB2 in a multiethnic asthma case-control study. Am J Respir Crit Care Med. 2006;174(10):1101–9.
  160. 160. Ortega VE, Hawkins GA, Moore WC, Hastie AT, Ampleford EJ, Busse WW, et al. Effect of rare variants in ADRB2 on risk of severe exacerbations and symptom control during longacting B agonist treatment in a multiethnic asthma population: a genetic study. Lancet Respir Med. 2014;2(3):204–13.
  161. 161. Rosenbaum DM, Cherezov V, Hanson MA, Rasmussen SGF, Thian FS, Kobilka TS, et al. GPCR engineering yields high-resolution structural insights into beta2-adrenergic receptor function. Science. 2007;318(5854):1266–73.
  162. 162. Cherezov V, Rosenbaum DM, Hanson M a, Rasmussen SGF, Thian FS, Kobilka TS, et al. High-resolution crystal structure of an engineered human beta2-adrenergic G protein-coupled receptor. Science. 2007;318(5854):1258–65.
  163. 163. Lima JJ, Thomason DB, Mohamed MH, Eberle L V, Self TH, Johnson JA. Impact of genetic polymorphisms of the beta2-adrenergic receptor on albuterol bronchodilator pharmacodynamics. Clin Pharmacol Ther. 1999;65(5):519–25.
  164. 164. Israel E, Chinchilli VM, Ford JG, Boushey HA, Cherniack R, Craig TJ, et al. Use of regularly scheduled albuterol treatment in asthma: genotype-stratified, randomised, placebo-controlled cross-over trial. Lancet. 2004;364(9444):1505–12.
  165. 165. Global Initiative for Asthma (GINA) [Internet]. 2016 Pocket Guide for Asthma Management and Prevention. Available from: http://www.ginasthma.org/. Accessed January 2016
  166. 166. Taylor DR, Drazen JM, Herbison GP, Yandava CN, Hancox RJ, Town GI. Asthma exacerbations during long term beta agonist use: influence of beta(2) adrenoceptor polymorphism. Thorax. 2000;55(9):762–7.
  167. 167. Israel E, Drazen JM, Liggett SB, Boushey HA, Cherniack RM, Chinchilli VM, et al. Effect of polymorphism of the b2-adrenergic receptor on response to regular use of albuterol in asthma. Int Arch Allergy Immunol. 2001;124(1–3):183–6.
  168. 168. Choudhry S, Ung N, Avila PC, Ziv E, Nazario S, Casal J, et al. Pharmacogenetic differences in response to albuterol between Puerto Ricans and Mexicans with asthma. Am J Respir Crit Care Med. 2005;171(6):563–70.
  169. 169. Bleecker ER, Postma DS, Lawrance RM, Meyers DA, Ambrose HJ, Goldman M. Effect of ADRB2 polymorphisms on response to longacting B2-agonist therapy: a pharmacogenetic analysis of two randomised studies. Lancet. 2007;370(9605):2118–25.
  170. 170. Wechsler ME, Kunselman SJ, Chinchilli VM, Bleecker E, Boushey HA, Calhoun WJ, et al. Effect of B2-adrenergic receptor polymorphism on response to longacting B2 agonist in asthma (LARGE trial): a genotype-stratified, randomised, placebo-controlled, crossover trial. Lancet. 2009;374(9703):1754–64.
  171. 171. Bleecker ER, Nelson HS, Kraft M, Corren J, Meyers DA, Yancey SW, et al. Beta2-receptor polymorphisms in patients receiving salmeterol with or without fluticasone propionate. Am J Respir Crit Care Med. 2010;181(7):676–87.
  172. 172. Tsuji S. Genetics of neurodegenerative diseases: insights from high-throughput resequencing. Hum Mol Genet. 2010;19(R1):R65–70.
  173. 173. Green SA, Cole G, Jacinto M, Innis M, Liggett SB. A polymorphism of the human B2-adrenergic receptor within the fourth transmembrane domain alters ligand binding and functional properties of the receptor. J Biol Chem. 1993;268(31):23116–21.
  174. 174. Green SA, Rathz DA, Schuster AJ, Liggett SB. The Ile164 B2-adrenoceptor polymorphism alters salmeterol exosite binding and conventional agonist coupling to Gs. Eur J Pharmacol. 2001;421(3):141–7.
  175. 175. Thomsen M, Nordestgaard BG, Sethi a a, Tybjærg-Hansen A, Dahl M. β2-adrenergic receptor polymorphisms, asthma and COPD: two large population-based studies. Eur Respir J. 2012;39(3):558–66.
  176. 176. Hacker BM, Tomlinson JE, Wayman G a, Sultana R, Chan G, Villacres E, et al. Cloning, chromosomal mapping, and regulatory properties of the human type 9 adenylyl cyclase (ADCY9). Genomics. 1998;50(1):97–104.
  177. 177. Pal K, Swaminathan K, Xu HE, Pioszak A a. Structural basis for hormone recognition by the Human CRFR2{alpha} G protein-coupled receptor. J Biol Chem. 2010;285(51):40351–61.
  178. 178. Poon AH, Tantisira KG, Litonjua AA, Lazarus R, Xu J, Lasky-Su J, et al. Association of corticotropin-releasing hormone receptor-2 genetic variants with acute bronchodilator response in asthma. Pharmacogenet Genomics. 2008;18(5):373–82.
  179. 179. Litonjua AA, Lasky-Su J, Schneiter K, Tantisira KG, Lazarus R, Klanderman B, et al. ARG1 is a novel bronchodilator response gene: screening and replication in four asthma cohorts. Am J Respir Crit Care Med. 2008;178(7):688–94.
  180. 180. Vonk JM, Postma DS, Maarsingh H, Bruinenberg M, Koppelman GH, Meurs H. Arginase 1 and arginase 2 variations associate with asthma, asthma severity and beta2 agonist and steroid response. Pharmacogenet Genomics. 2010;20:179–86.
  181. 181. Iordanidou M, Paraskakis E, Tavridou A, Paschou P, Chatzimichael A, Manolopoulos VG. G894T polymorphism of eNOS gene is a predictor of response to combination of inhaled corticosteroids with long-lasting β2-agonists in asthmatic children. Pharmacogenomics. 2012;13(12):1363–72.
  182. 182. Duan QL, Du R, Lasky-Su J, Klanderman BJ, Partch a B, Peters SP, et al. A polymorphism in the thyroid hormone receptor gene is associated with bronchodilator response in asthmatics. Pharmacogenomics J. 2013;13(2):130–6.
  183. 183. Drake KA, Torgerson DG, Gignoux CR, Galanter JM, Roth LA, Huntsman S, et al. A genome-wide association study of bronchodilator response in Latinos implicates rare variants. J Allergy Clin Immunol. 2014;133(2):370–8.
  184. 184. Himes BE, Jiang X, Hu R, Wu AC, Lasky-Su JA, Klanderman BJ, et al. Genome-wide association analysis in asthma subjects identifies SPATS2L as a novel bronchodilator response gene. PLoS Genet. 2012;8(7):e1002824.
  185. 185. Drazen JM, Yandava CN, Dubé L, Szczerback N, Hippensteel R, Pillari A, et al. Pharmacogenetic association between ALOX5 promoter genotype and the response to anti-asthma treatment. Nat Genet. 1999;22(2):168–70.
  186. 186. Mougey E, Lang JE, Allayee H, Teague WG, Dozor AJ, Wise RA, et al. ALOX5 polymorphism associates with increased leukotriene production and reduced lung function and asthma control in children with poorly controlled asthma. Clin Exp Allergy. 2013;43(5):512–20.
  187. 187. Telleria JJ, Blanco-Quiros A, Varillas D, Armentia A, Fernandez-Carvajal I, Jesus Alonso M, et al. ALOX5 promoter genotype and response to montelukast in moderate persistent asthma. Respir Med. 2008;102(6):857–61.
  188. 188. Klotsman M, York TP, Pillai SG, Vargas-Irwin C, Sharma SS, van den Oord EJCG, et al. Pharmacogenetics of the 5-lipoxygenase biosynthetic pathway and variable clinical response to montelukast. Pharmacogenet Genomics. 2007;17(3):189–96.
  189. 189. Lima JJ, Zhang S, Grant A, Shao L, Tantisira KG, Allayee H, et al. Influence of leukotriene pathway polymorphisms on response to montelukast in asthma. Am J Respir Crit Care Med. 2006;173(4):379–85.
  190. 190. Tantisira KG, Lima J, Sylvia J, Klanderman B, Weiss ST. 5-lipoxygenase pharmacogenetics in asthma: overlap with Cys-leukotriene receptor antagonist loci. Pharmacogenet Genomics. 2009;19(3):244–7.
  191. 191. E. Mougey, H.Feng, M.Castro, C. Irvin JL. Absorption of montelukast is transporter mediated: a common variant of OATP2B1 is associated with reduced plasma concentrations and poor response. Pharmacogenet Genomics. 2009;19(2):129–38.
  192. 192. Kim KA, Lee HM, Joo HJ, Park IB, Park JY. Effects of polymorphisms of the SLCO2B1 transporter gene on the pharmacokinetics of montelukast in humans. J Clin Pharmacol. 2013;53(11):1186–93.
  193. 193. Tapaninen T, Karonen T, Backman JT, Neuvonen PJ, Niemi M. SLCO2B1 c.935G>A single nucleotide polymorphism has no effect on the pharmacokinetics of montelukast and aliskiren. Pharmacogenet Genomics. 2013;23(1):19–24.
  194. 194. Jirtle RL, Skinner MK. Environmental epigenomics and disease susceptibility. Nat Rev Genet. 2007;8(4):253–62.
  195. 195. Jaenisch R, Bird A. Epigenetic regulation of gene expression: how the genome integrates intrinsic and environmental signals. Nat Genet. 2003;33 Suppl:245–54.
  196. 196. Comer BS, Ba M, Singer CA, Gerthoffer WT. Epigenetic targets for novel therapies of lung diseases. Pharmacol Ther. 2015;147:91–110.
  197. 197. Follows RM, Snowise NG, Ho S-Y, Ambery CL, Smart K, McQuade BA. Efficacy, safety and tolerability of GSK2190915, a 5-lipoxygenase activating protein inhibitor, in adults and adolescents with persistent asthma: a randomised dose-ranging study. Respir Res. 2013;14(1):54.
  198. 198. Hodsman P, Ashman C, Cahn A, De Boever E, Locantore N, Serone A, et al. A phase 1, randomized, placebo-controlled, dose-escalation study of an anti-IL-13 monoclonal antibody in healthy subjects and mild asthmatics. Br J Clin Pharmacol. 2013;75(1):118–28.
  199. 199. Laviolette M, Gossage DL, Gauvreau G, Leigh R, Olivenstein R, Katial R, et al. Effects of benralizumab on airway eosinophils in asthmatic patients with sputum eosinophilia. J Allergy Clin Immunol. 2013;132(5):1086–96.e5.
  200. 200. Garcia G, Magnan A, Chiron R, Contin-Bordes C, Berger P, Taillé C, et al. A proof-of-concept, randomized, controlled trial of omalizumab in patients with severe, difficult-to-control, nonatopic asthma. Chest. 2013;144(2):411–9.
  201. 201. Noonan M, Korenblat P, Mosesova S, Scheerens H, Arron JR, Zheng Y, et al. Dose-ranging study of lebrikizumab in asthmatic patients not receiving inhaled steroids. J Allergy Clin Immunol. 2013;132(3):567–74.e12.
  202. 202. Piper E, Brightling C, Niven R, Oh C, Faggioni R, Poon K, et al. A phase II placebo-controlled study of tralokinumab in moderate-to-severe asthma. Eur Respir J. 2013;41(2):330–8.
  203. 203. Busse WW, O’Byrne PM, Bleecker ER, Lötvall J, Woodcock A, Andersen L, et al. Safety and tolerability of the novel inhaled corticosteroid fluticasone furoate in combination with the β2 agonist vilanterol administered once daily for 52 weeks in patients >=12 years old with asthma: a randomised trial. Thorax. 2013;68(6):513–20.
  204. 204. Corren J, Mansfield LE, Pertseva T, Blahzko V, Kaiser K. Efficacy and safety of fluticasone/formoterol combination therapy in patients with moderate-to-severe asthma. Respir Med. 2013;107(2):180–95.
  205. 205. Ortega HG, Liu MC, Pavord ID, Brusselle GG, FitzGerald JM, Chetta A, et al. Mepolizumab Treatment in Patients with Severe Eosinophilic Asthma. N Engl J Med. 2015 Sep 8;372(18):17.
  206. 206. Somerville L, Bardelas J, Viegas A, D’Andrea P, Blogg M, Peachey G. Immunogenicity and safety of omalizumab in pre-filled syringes in patients with allergic (IgE-mediated) asthma. Curr Med Res Opin. 2014;30(1):59–66.
  207. 207. Scheerens H, Arron JR, Zheng Y, Putnam WS, Erickson RW, Choy DF, et al. The effects of lebrikizumab in patients with mild asthma following whole lung allergen challenge. Clin Exp Allergy. 2014;44(1):38–46.
  208. 208. Lötvall J, Bateman ED, Busse WW, O’Byrne PM, Woodcock A, Toler WT, et al. Comparison of vilanterol, a novel long-acting beta2 agonist, with placebo and a salmeterol reference arm in asthma uncontrolled by inhaled corticosteroids. J Negat Results Biomed. 2014;13(1):9.
  209. 209. Beeh K-M, Moroni-Zentgraf P, Ablinger O, Hollaenderova Z, Unseld A, Engel M, et al. Tiotropium Respimat® in asthma: a double-blind, randomised, dose-ranging study in adult patients with moderate asthma. Respir Res. 2014;15(1):61.
  210. 210. Gauvreau GM, Boulet L-P, Cockcroft DW, FitzGerald JM, Mayers I, Carlsten C, et al. OX40L blockade and allergen-induced airway responses in subjects with mild asthma. Clin Exp Allergy. 2014;44(1):29–37.
  211. 211. Nowak RM, Parker JM, Silverman RA, Rowe BH, Smithline H, Khan F, et al. A randomized trial of benralizumab, an antiinterleukin 5 receptor α monoclonal antibody, after acute asthma. Am J Emerg Med. 2015;33(1):14–20.
  212. 212. Hernandez ML, Mills K, Almond M, Todoric K, Aleman MM, Zhang H, et al. IL-1 receptor antagonist reduces endotoxin-induced airway inflammation in healthy volunteers. J Allergy Clin Immunol. 2015;135(2):379–85.

Written By

Mina Youssef, Cynthia Kanagaratham, Mohamed I. Saad and Danuta Radzioch

Reviewed: 13 April 2016 Published: 06 July 2016