U.S. flag

An official website of the United States government

The .gov means it’s official. Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

The site is secure. The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

  • Publications
  • Account settings

Preview improvements coming to the PMC website in October 2024. Learn More or Try it out now .

  • Advanced Search
  • Journal List
  • Allergy Asthma Clin Immunol

Logo of aaci

Treatment strategies for asthma: reshaping the concept of asthma management

Alberto papi.

1 Section of Cardiorespiratory and Internal Medicine, Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy

7 Respiratory Unit, Emergency Department, University Hospital S. Anna, Via Aldo Moro 8, 44124 Ferrara, Italy

Francesco Blasi

2 Internal Medicine Department, Respiratory Unit and Adult Cystic Fibrosis Center, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy

3 Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy

Giorgio Walter Canonica

4 Personalized Medicine Asthma & Allergy Clinic, Humanitas University & Istituto Clinico Humanitas, Milan, Italy

Luca Morandi

Luca richeldi.

5 Università Cattolica del Sacro Cuore, Fondazione Policlinico A. Gemelli IRCCS, Rome, Italy

Andrea Rossi

6 Respiratory Section, Department of Medicine, University of Verona, Verona, Italy

Associated Data

Not applicable.

Asthma is a common chronic disease characterized by episodic or persistent respiratory symptoms and airflow limitation. Asthma treatment is based on a stepwise and control-based approach that involves an iterative cycle of assessment, adjustment of the treatment and review of the response aimed to minimize symptom burden and risk of exacerbations. Anti-inflammatory treatment is the mainstay of asthma management. In this review we will discuss the rationale and barriers to the treatment of asthma that may result in poor outcomes. The benefits of currently available treatments and the possible strategies to overcome the barriers that limit the achievement of asthma control in real-life conditions and how these led to the GINA 2019 guidelines for asthma treatment and prevention will also be discussed.

Asthma, a major global health problem affecting as many as 235 million people worldwide [ 1 ], is a common, non-communicable, and variable chronic disease that can result in episodic or persistent respiratory symptoms (e.g. shortness of breath, wheezing, chest tightness, cough) and airflow limitation, the latter being due to bronchoconstriction, airway wall thickening, and increased mucus.

The pathophysiology of the disease is complex and heterogeneous, involving various host-environment interactions occurring at various scales, from genes to organ [ 2 ].

Asthma is a chronic disease requiring ongoing and comprehensive treatment aimed to reduce the symptom burden (i.e. good symptom control while maintaining normal activity levels), and minimize the risk of adverse events such as exacerbations, fixed airflow limitation and treatment side effects [ 3 , 4 ].

Asthma treatment is based on a stepwise approach. The management of the patient is control-based; that is, it involves an iterative cycle of assessment (e.g. symptoms, risk factors, etc.), adjustment of treatment (i.e. pharmacological, non-pharmacological and treatment of modifiable risk factors) and review of the response (e.g. symptoms, side effects, exacerbations, etc.). Patients’ preferences should be taken into account and effective asthma management should be the result of a partnership between the health care provider and the person with asthma, particularly when considering that patients and clinicians might aim for different goals [ 4 ].

This review will discuss the rationale and barriers to the treatment of asthma, that may result in poor patient outcomes. The benefits of currently available treatments and the possible strategies to overcome the barriers that limit the achievement of asthma control in real-life situations will also be discussed.

The treatment of asthma: where are we? Evolution of a concept

Asthma control medications reduce airway inflammation and help to prevent asthma symptoms; among these, inhaled corticosteroids (ICS) are the mainstay in the treatment of asthma, whereas quick-relief (reliever) or rescue medicines quickly ease symptoms that may arise acutely. Among these, short-acting beta-agonists (SABAs) rapidly reduce airway bronchoconstriction (causing relaxation of airway smooth muscles).

National and international guidelines have recommended SABAs as first-line treatment for patients with mild asthma, since the Global Initiative for Asthma guidelines (GINA) were first published in 1995, adopting an approach aimed to control the symptoms rather than the underlying condition; a SABA has been the recommended rescue medication for rapid symptom relief. This approach stems from the dated idea that asthma symptoms are related to bronchial smooth muscle contraction (bronchoconstriction) rather than a condition concomitantly caused by airway inflammation. In 2019, the GINA guidelines review (GINA 2019) [ 4 ] introduced substantial changes overcoming some of the limitations and “weaknesses” of the previously proposed stepwise approach to adjusting asthma treatment for individual patients. The concept of an anti-inflammatory reliever has been adopted at all degrees of severity as a crucial component in the management of the disease, increasing the efficacy of the treatment while lowering SABA risks associated with patients’ tendency to rely or over-rely on the as-needed medication.

Until 2017, the GINA strategy proposed a pharmacological approach based on a controller treatment (an anti-inflammatory, the pillar of asthma treatment), with a SABA as an additional rescue intervention. The reliever, a short-acting bronc hodilator, was merely an addendum , a medication to be used in case the real treatment (the controller) failed to maintain disease control: SABAs effectively induce rapid symptom relief but are ineffective on the underlying inflammatory process. Based on the requirement to achieve control, the intensity of the controller treatment was related to the severity of the disease, varying from low-dose ICS to combination low-dose ICS/long-acting beta-agonist (LABA), medium-dose ICS/LABA, up to high-dose ICS/LABA, as preferred controller choice, with a SABA as the rescue medication. As a result, milder patients were left without any anti-inflammatory treatment and could only rely on SABA rescue treatment.

Poor adherence to therapy is a major limitation of a treatment strategy based on the early introduction of the regular use of controller therapy [ 5 ]. Indeed, a number of surveys have highlighted a common pattern in the use of inhaled medication [ 6 ], in which treatment is administered only when asthma symptoms occur; in the absence of symptoms, treatment is avoided as patients perceive it as unnecessary. When symptoms worsen, patients prefer to use reliever therapies, which may result in the overuse of SABAs [ 7 ]. Indirect evidence suggests that the overuse of beta-agonists alone is associated with increased risk of death from asthma [ 8 ].

In patients with mild persistent disease, low-dose ICS decreases the risk of severe exacerbations leading to hospitalization and improves asthma control [ 9 ]. When low-dose ICS are ineffective in controlling the disease (Step 3 of the stepwise approach), a combination of low-dose ICS with LABA maintenance was the recommended first-choice treatment, plus as-needed SABA [ 3 , 10 ]. Alternatively, the combination low-dose ICS/LABA (formoterol) was to be used as single maintenance and reliever treatment (SMART). The SMART strategy containing the rapid-acting formoterol was recommended throughout GINA Steps 3 to 5 based on solid clinical-data evidence [ 3 ].

The addition of a LABA to ICS treatment reduces both severe and mild asthma exacerbation rates, as shown in the one-year, randomized, double-blind, parallel-group FACET study [ 11 ]. This study focused on patients with persistent asthma symptoms despite receiving ICS and investigated the efficacy of the addition of formoterol to two dose levels of budesonide (100 and 400 µg bid ) in decreasing the incidence of both severe and mild asthma exacerbations. Adding formoterol decreased the incidence of both severe and mild asthma exacerbations, independent of ICS dose. Severe and mild exacerbation rates were reduced by 26% and 40%, respectively, with the addition of formoterol to the lower dose of budesonide; the corresponding reductions were 63% and 62%, respectively, when formoterol was added to budesonide at the higher dose.

The efficacy of the ICS/LABA combination was confirmed in the post hoc analysis of the FACET study, in which patients were exposed to a combination of formoterol and low-dose budesonide [ 12 ]. However, such high levels of asthma control are not achieved in real life [ 5 ]. An explanation for this is that asthma is a variable condition and this variability might include the exposure of patients to factors which may cause a transient steroid insensitivity in the inflammatory process. This, in turn, may lead to an uncontrolled inflammatory response and to exacerbations, despite optimal controller treatment. A typical example of this mechanism is given by viral infections, the most frequent triggers of asthma exacerbations. Rhinoviruses, the most common viruses found in patients with asthma exacerbations, interfere with the mechanism of action of corticosteroids making the anti-inflammatory treatment transiently ineffective. A transient increase in the anti-inflammatory dose would overcome the trigger-induced anti-inflammatory resistance, avoiding uncontrolled inflammation leading to an exacerbation episode [ 13 – 15 ].

Indeed, symptoms are associated with worsening inflammation and not only with bronchoconstriction. Romagnoli et al. showed that inflammation, as evidenced by sputum eosinophilia and eosinophilic markers, is associated with symptomatic asthma [ 16 ]. A transient escalation of the ICS dose would prevent loss of control over inflammation and decrease the risk of progression toward an acute episode. In real life, when experiencing a deterioration of asthma control, patients self-treat by substantially increasing their SABA medication (Fig.  1 ); it is only subsequently that they (modestly) increase the maintenance treatment [ 17 ].

An external file that holds a picture, illustration, etc.
Object name is 13223_2020_472_Fig1_HTML.jpg

Mean use of SABA at different stages of asthma worsening. Patients have been grouped according to maintenance therapy shown in the legend. From [ 17 ], modified

As bronchodilators, SABAs do not control the underlying inflammation associated with increased symptoms. The “as required” use of SABAs is not the most effective therapeutic option in controlling a worsening of inflammation, as signaled by the occurrence of symptoms; instead, an anti-inflammatory therapy included in the rescue medication along with a rapid-acting bronchodilator could provide both rapid symptom relief and control over the underlying inflammation. Thus, there is a need for a paradigm shift, a new therapeutic approach based on the rescue use of an inhaled rapid-acting beta-agonist combined with an ICS: an anti-inflammatory reliever strategy [ 18 ].

The symptoms of an exacerbation episode, as reported by Tattersfield and colleagues in their extension of the FACET study, increase gradually before the peak of the exacerbation (Fig.  2 ); and the best marker of worsening asthma is the increased use of rescue beta-agonist treatment that follows exactly the pattern of worsening symptomatology [ 19 ]. When an ICS is administered with the rescue bronchodilator, the patient would receive anti-inflammatory therapy when it is required; that is, when the inflammation is uncontrolled, thus increasing the efficiency of the anti-inflammatory treatment.

An external file that holds a picture, illustration, etc.
Object name is 13223_2020_472_Fig2_HTML.jpg

Percent variation in symptoms, rescue beta-agonist use and peak expiratory flow (PEF) during an exacerbation. In order to allow comparison over time, data have been standardized (Day-14 = 0%; maximum change = 100%)

(From [ 19 ])

Barriers and paradoxes of asthma management

A number of barriers and controversies in the pharmacological treatment of asthma have prevented the achievement of effective disease management [ 20 ]. O’Byrne and colleagues described several such controversies in a commentary published in 2017, including: (1) the recommendation in Step 1 of earlier guidelines for SABA bronchodilator use alone, despite asthma being a chronic inflammatory condition; and (2) the autonomy given to patients over perception of need and disease control at Step 1, as opposed to the recommendation of a fixed-dose approach with treatment-step increase, regardless of the level of symptoms [ 20 ]. Other controversies outlined were: (3) a difficulty for patients in understanding the recommendation to minimize SABA use at Step 2 and switch to a fixed-dose ICS regimen, when they perceive SABA use as more effective; (4) apparent conflicting safety messages within the guidelines that patient-administered SABA monotherapy is safe, but patient-administered LABA monotherapy is not; and (5) a discrepancy as to patients’ understanding of “controlled asthma” and their symptom frequency, impact and severity [ 20 ].

Controversies (1) and (2) can both establish an early over-dependence on SABAs. Indeed, asthma patients freely use (and possibly overuse) SABAs as rescue medication. UK registry data have recently suggested SABA overuse or overreliance may be linked to asthma-related deaths: among 165 patients on short-acting relievers at the time of death, 56%, 39%, and 4% had been prescribed > 6, > 12, and > 50 SABA inhalers respectively in the previous year [ 21 ]. Registry studies have shown the number of SABA canisters used per year to be directly related to the risk of death in patients with asthma. Conversely, the number of ICS canisters used per year is inversely related to the rate of death from asthma, when compared with non-users of ICS [ 8 , 22 ]. Furthermore, low-dose ICS used regularly are associated with a decreased risk of asthma death, with discontinuation of these agents possibly detrimental [ 22 ].

Other barriers to asthma pharmacotherapy have included the suggestion that prolonged treatment with LABAs may mask airway inflammation or promote tolerance to their effects. Investigating this, Pauwels and colleagues found that in patients with asthma symptoms that were persistent despite taking inhaled glucocorticoids, the addition of regular treatment with formoterol to budesonide for a 12-month period did not decrease asthma control, and improved asthma symptoms and lung function [ 11 ].

Treatment strategies across all levels of asthma severity

Focusing on risk reduction, the 2014 update of the GINA guidelines recommended as-needed SABA for Step 1 of the stepwise treatment approach, with low-dose ICS maintenance therapy as an alternative approach for long-term anti-inflammatory treatment [ 23 ]. Such a strategy was only supported by the evidence from a post hoc efficacy analysis of the START study in patients with recently diagnosed mild asthma [ 24 ]. The authors showed that low-dose budesonide reduced the decline of lung-function over 3 years and consistently reduced severe exacerbations, regardless of symptom frequency at baseline, even in subjects with symptoms below the then-threshold of eligibility for ICS [ 24 ]. However, as for all post hoc analyses, the study by Reddel and colleagues does not provide conclusive evidence and, even so, their results could have questionable clinical significance for the management of patients with early mild asthma. To be effective, this approach would require patients to be compliant to regular twice-daily ICS for 10 years to have the number of exacerbations reduce by one. In real life, it is highly unlikely that patients with mild asthma would adhere to such a regular regimen [ 25 ].

The 2016 update to the GINA guidelines lowered the threshold for the use of low-dose ICS (GINA Step 2) to two episodes of asthma symptoms per month (in the absence of any supportive evidence for the previous cut-off). The objective was to effectively increase the asthma population eligible to receive regular ICS treatment and reduce the population treated with a SABA only, given the lack of robust evidence of the latter’s efficacy and safety and the fact that asthma is a variable condition characterized by acute exacerbations [ 26 ]. Similarly, UK authorities recommended low-dose ICS treatment in mild asthma, even for patients with suspected asthma, rather than treatment with a SABA alone [ 10 ]. However, these patients are unlikely to have good adherence to the regular use of an ICS. It is well known that poor adherence to treatment is a major problem in asthma management, even for patients with severe asthma. In their prospective study of 2004, Krishnan and colleagues evaluated the adherence to ICS and oral corticosteroids (OCS) in a cohort of patients hospitalized for asthma exacerbations [ 27 ]. The trend in the data showed that adherence to ICS and OCS treatment in patients dropped rapidly to reach nearly 50% within 7 days of hospital discharge, with the rate of OCS discontinuation per day nearly double the rate of ICS discontinuation per day (− 5.2% vs. − 2.7%; p < 0.0001 respectively, Fig.  3 ), thus showing that even after a severe event, patients’ adherence to treatment is suboptimal [ 27 ].

An external file that holds a picture, illustration, etc.
Object name is 13223_2020_472_Fig3_HTML.jpg

Use of inhaled (ICS) and oral (OCS) corticosteroids in patients after hospital discharge among high-risk adult patients with asthma. The corticosteroid use was monitored electronically. Error bars represent the standard errors of the measured ICS and OCS use

(From [ 27 ])

Guidelines set criteria with the aim of achieving optimal control of asthma; however, the attitude of patients towards asthma management is suboptimal. Partridge and colleagues were the first in 2006 to evaluate the level of asthma control and the attitude of patients towards asthma management. Patients self-managed their condition using their medication as and when they felt the need, and adjusted their treatment by increasing their intake of SABA, aiming for an immediate relief from symptoms [ 17 ]. The authors concluded that the adoption of a patient-centered approach in asthma management could be advantageous to improve asthma control.

The concomitant administration of an as-needed bronchodilator and ICS would provide rapid relief while administering anti-inflammatory therapy. This concept is not new: in the maintenance and reliever approach, patients are treated with ICS/formoterol (fast-acting, long-acting bronchodilator) combinations for both maintenance and reliever therapy. An effective example of this therapeutic approach is provided in the SMILE study in which symptomatic patients with moderate to severe asthma and treated with budesonide/formoterol as maintenance therapy were exposed to three different as-needed options: SABA (terbutaline), rapid-onset LABA (formoterol) and a combination of LABA and ICS (budesonide/formoterol) [ 28 ]. When compared with formoterol, budesonide/formoterol as reliever therapy significantly reduced the risk of severe exacerbations, indicating the efficacy of ICS as rescue medication and the importance of the as-needed use of the anti-inflammatory reliever.

The combination of an ICS and a LABA (budesonide/formoterol) in one inhaler for both maintenance and reliever therapy is even more effective than higher doses of maintenance ICS and LABA, as evidenced by Kuna and colleagues and Bousquet and colleagues (Fig.  4 ) [ 29 , 30 ].

An external file that holds a picture, illustration, etc.
Object name is 13223_2020_472_Fig4_HTML.jpg

Comparison between the improvements in daily asthma control resulting from the use of budesonide/formoterol maintenance and reliever therapy vs. higher dose of ICS/LABA + SABAZ and steroid load for the two regimens

(Data from [ 29 , 30 ])

The effects of single maintenance and reliever therapy versus ICS with or without LABA (controller therapy) and SABA (reliever therapy) have been recently addressed in the meta-analysis by Sobieraj and colleagues, who analysed 16 randomized clinical trials involving patients with persistent asthma [ 31 ]. The systematic review supported the use of single maintenance and reliever therapy, which reduces the risk of exacerbations requiring systemic corticosteroids and/or hospitalization when compared with various strategies using SABA as rescue medication [ 31 ].

This concept was applied to mild asthma by the BEST study group, who were the first to challenge the regular use of ICS. A pilot study by Papi and colleagues evaluated the efficacy of the symptom-driven use of beclomethasone dipropionate plus albuterol in a single inhaler versus maintenance with inhaled beclomethasone and as-needed albuterol. In this six-month, double-blind, double-dummy, randomized, parallel-group trial, 455 patients with mild asthma were randomized to one of four treatment groups: an as-needed combination therapy of placebo bid plus 250 μg of beclomethasone and 100 μg of albuterol in a single inhaler; an as-needed albuterol combination therapy consisting of placebo bid plus 100 μg of albuterol; regular beclomethasone therapy, comprising beclomethasone 250 μg bid and 100 μg albuterol as needed); and regular combination therapy with beclomethasone 250 μg and albuterol 100 μg in a single inhaler bid plus albuterol 100 μg as needed.

The rescue use of beclomethasone/albuterol in a single inhaler was as efficacious as the regular use of inhaled beclomethasone (250 μg bid ) and it was associated with a lower 6-month cumulative dose of the ICS [ 32 ].

The time to first exacerbation differed significantly among groups ( p  = 0.003), with the shortest in the as-needed albuterol and placebo group (Fig.  5 ). Figure  5 also shows equivalence between the as-needed combination therapy and the regular beclomethasone therapy. However, these results were not conclusive since the study was not powered to evaluate the effect of the treatment on exacerbations. In conclusion, as suggested by the study findings, mild asthma patients may require the use of an as-needed ICS and an inhaled bronchodilator rather than a regular treatment with ICS [ 32 ].

An external file that holds a picture, illustration, etc.
Object name is 13223_2020_472_Fig5_HTML.jpg

Kaplan Meier analysis of the time to first exacerbation (modified intention-to-treat population). First asthma exacerbations are shown as thick marks. As-needed albuterol therapy = placebo bid plus 100 μg of albuterol as needed; regular combination therapy = 250 μg of beclomethasone and 100 μg of albuterol in a single inhaler bid plus 100 μg of albuterol as needed; regular beclomethasone therapy = 250 μg of beclomethasone bid and 100 μg of albuterol as needed; as-needed combination therapy = placebo bid plus 250 μg of beclomethasone and 100 μg of albuterol in a single inhaler as needed

(From [ 32 ])

Moving forward: a new approach to the management of asthma patients

Nearly a decade after the publication of the BEST study in 2007, the use of this alternative therapeutic strategy was addressed in the SYGMA 1 and SYGMA 2 trials. These double-blind, randomized, parallel-group, 52-week phase III trials evaluated the efficacy of as-needed use of combination formoterol (LABA) and the ICS budesonide as an anti-inflammatory reliever in patients requiring GINA Step 2 treatment, with the current reliever therapy (e.g. as-needed SABA) or with low-dose maintenance ICS (inhaled budesonide bid ) plus as-needed SABA, administered as regular controller therapy [ 33 , 34 ].

The SYGMA 1 trial, which enrolled 3849 patients, aimed to demonstrate the superiority of the as-needed use of the combination budesonide/formoterol over as-needed terbutaline, as measured by the electronically-recorded proportion of weeks with well-controlled asthma [ 34 ]. The more pragmatic SYGMA 2 trial enrolled 4215 patients with the aim to demonstrate that the budesonide/formoterol combination is non-inferior to budesonide plus as-needed terbutaline in reducing the relative rate of annual severe asthma exacerbations [ 33 ]. Both trials met their primary efficacy outcomes. In particular, as-needed budesonide/formoterol was superior to as-needed SABA in controlling asthma symptoms (34.4% versus 31.1%) and preventing exacerbations, achieving a 64% reduction in exacerbations. In both trials, budesonide/formoterol as-needed was similar to budesonide maintenance bid at preventing severe exacerbations, with a substantial reduction of the inhaled steroid load over the study period (83% in the SYGMA 1 trial and 75% in the SYGMA 2 trial). The time to first exacerbation did not differ significantly between the two regimens; however, budesonide/formoterol was superior to SABA in prolonging the time to first severe exacerbation [ 33 , 34 ].

The double-blind, placebo-controlled design of the SYGMA trials does not fully address the advantages of anti-inflammatory reliever strategy in patients who often rely on SABAs for symptom relief, so to what extent the study findings could apply to real-life practice settings was unclear.

These limitations were overcome by the results of the Novel START study, an open-label, randomized, parallel-group, controlled trial designed to reflect real-world practice, which demonstrated the effectiveness in mild asthma of budesonide/formoterol as an anti-inflammatory reliever therapy [ 35 ].

In real-world practice, mild asthma patients are treated with an as-needed SABA reliever or with daily low-dose ICS maintenance therapy plus a SABA reliever. In the Novel START study, 668 patients with mild asthma were randomized to receive either as-needed albuterol 100 µg, two inhalations (SABA reliever as a continuation of the Step 1 treatment according to the 2017 GINA guidelines), budesonide 200 µg (ICS maintenance treatment) plus as-needed albuterol (Step 2 therapy of the GINA 2017 guidelines), or 200 µg/6 µg budesonide/formoterol as anti-inflammatory reliever therapy taken as-needed for a 52-week study period.

In this study, the rate of asthma exacerbations for budesonide/formoterol was lower compared with albuterol (51%) and similar to the twice-daily maintenance budesonide plus albuterol, despite a 52% reduction in the mean steroid dose with the single combination inhaler treatment [ 35 ]. In addition, severe exacerbation rate was lower with budesonide/formoterol as compared with as-needed albuterol and regular twice-daily budesonide. These data support the findings of the SYGMA 1 and 2 trials, highlighting the need for a critical re-examination of current clinical practice. Along with the results of the SYGMA trials, they provide convincing evidence of the advantages of the anti-inflammatory reliever strategy, particularly in real-life settings.

The SYGMA 1, SYGMA 2 and the novel START studies complete the picture of the treatment strategies for asthma at any degree of severity, including mild asthma. A growing body of evidence shows that an anti-inflammatory reliever strategy, when compared with all other strategies with SABA reliever, consistently reduces the rate of exacerbations across all levels of asthma severity (Fig.  6 ) [ 28 , 29 , 34 , 36 – 39 ].

An external file that holds a picture, illustration, etc.
Object name is 13223_2020_472_Fig6_HTML.jpg

Risk reduction of severe asthma attack of anti-inflammatory reliever versus SABA across all levels of asthma severity. Bud = budesonide; form = formoterol; TBH = turbohaler. Data from: 1: [ 36 ]; 2: [ 37 ]; 3: [ 38 ]; 4: [ 28 ]; 5: [ 29 ]; 6: [ 30 ]; 7: [ 34 ]

(Data source: [ 39 ])

This evidence set the ground (Fig.  7 ) for the release of the 2019 GINA strategy updates. The document provides a consistent approach towards the management of the disease and aims to avoid the overreliance and overuse of SABAs, even in the early course of the disease. The 2019 GINA has introduced key changes in the treatment of mild asthma: for safety reasons, asthmatic adults and adolescents should receive ICS-containing controller treatment instead of the SABA-only treatment, which is no longer recommended.

An external file that holds a picture, illustration, etc.
Object name is 13223_2020_472_Fig7_HTML.jpg

Timeline of key randomized controlled trials and meta-analyses providing the supporting evidence base leading to the Global Initiative for Asthma (GINA) 2019 guidelines. GINA global initiative for asthma, MART maintenance and reliever therapy, SMART single inhaler maintenance and reliever therapy

In Step 1 of the stepwise approach to adjusting asthma treatment, the preferred controller option for patients with fewer than two symptoms/month and no exacerbation risk factors is low-dose ICS/formoterol as needed. This strategy is indirectly supported by the results of the SYGMA 1 study which evaluated the efficacy and safety of budesonide/formoterol as needed, compared with as-needed terbutaline and budesonide bid plus as-needed terbutaline (see above). In patients with mild asthma, the use of an ICS/LABA (budesonide/formoterol) combination as needed provided superior symptom control to as-needed SABA, resulting in a 64% lower rate of exacerbations (p = 0.07) with a lower steroid dose (17% of the budesonide maintenance dose) [ 34 ]. The changes extend to the other controller options as well. In the 2017 GINA guidelines, the preferred treatment was as-needed SABA with the option to consider adding a regular low-dose ICS to the reliever. In order to overcome the poor adherence with the ICS regimen, and with the aim to reduce the risk of severe exacerbations, the 2019 GINA document recommends taking low-dose ICS whenever SABA is taken, with the daily ICS option no longer listed.

Previous studies including the TREXA study in children and adolescents [ 40 ], the BASALT study [ 41 ] and research conducted by the BEST study group [ 32 ] have already added to the evidence that a low-dose ICS with a bronchodilator is an effective strategy for symptom control in patients with mild asthma. A recently published study in African-American children with mild asthma found that the use of as-needed ICS with SABA provides similar asthma control, exacerbation rates and lung function measures at 1 year, compared with daily ICS controller therapy [ 42 ], adding support to TREXA findings that in children with well controlled, mild asthma, ICS used as rescue medication with SABA may be an efficacious step-down strategy [ 40 ].

In Step 2 of the stepwise approach, there are now two preferred controller options: (a) a daily low-dose ICS plus an as-needed SABA; and (b) as-needed low-dose ICS/formoterol. Recommendation (a) is supported by a large body of evidence from randomized controlled trials and observations showing a substantial reduction of exacerbation, hospitalization, and death with regular low-dose ICS [ 7 – 9 , 24 , 43 ], whereas recommendation (b) stems from evidence on the reduction or non-inferiority for severe exacerbations when as-needed low-dose ICS/formoterol is compared with regular ICS [ 33 , 34 ].

The new GINA document also suggests low-dose ICS is taken whenever SABA is taken, either as separate inhalers or in combination. This recommendation is supported by studies showing reduced exacerbation rates compared with taking a SABA only [ 32 , 40 ], or similar rates compared with regular ICS [ 32 , 40 , 41 ]. Low-dose theophylline, suggested as an alternative controller in the 2017 GINA guidelines, is no longer recommended.

Airway inflammation is present in the majority of patients with asthma, and although patients with mild asthma may have only infrequent symptoms, they face ongoing chronic inflammation of the lower airways and risk acute exacerbations. The GINA 2019 strategy recognizes the importance of reducing the risk of asthma exacerbations, even in patients with mild asthma (Steps 1 and 2) [ 4 ]. In this regard, the new recommendations note that SABA alone for symptomatic treatment is non-protective against severe exacerbation and may actually increase exacerbation risk if used regularly or frequently [ 4 ].

The reluctance by patients to regularly use an ICS controller means they may instead try and manage their asthma symptoms by increasing their SABA reliever use. This can result in SABA overuse and increased prescribing, and increased risk of exacerbations.

As part of the global SABINA (SABA use IN Asthma) observational study programme, a UK study examined primary care records to describe the pattern of SABA and ICS use over a 10-year period in 373,256 patients with mild asthma [ 44 ]. Results showed that year-to-year SABA prescribing was more variable than that of ICS indicating that, in response to fluctuations in asthma symptom control, SABA use was increased in preference to ICS use. Furthermore, more than 33% of patients were prescribed SABA inhalers at a level equivalent to around ≥ 3 puffs per week which, according to GINA, suggests inadequate asthma control.

The problem of SABA overuse is further highlighted by two studies [ 45 , 46 ], also as part of the SABINA programme. These analysed data from 365,324 patients in a Swedish cohort prescribed two medications for obstructive lung disease in any 12-month period (HERA).

The first study identified SABA overuse (defined as ≥ 3 SABA canisters a year) in 30% of patients, irrespective of their ICS use; 21% of patients were collecting 3–5 canisters annually, 7% were collecting 6–10, and 2% more than 11 [ 45 ]. Those patients who were overusing SABA had significantly more asthma exacerbations relative to those using < 3 canisters (20.0 versus 12.5 per 100 patient years; relative risk 1.60, 95% CI 1.57–1.63, p < 0.001). Moreover, patients overusing SABA and whose asthma was more severe (GINA Steps 3 and 4) had greater exacerbation risk compared with overusing patients whose asthma was milder (GINA Steps 1 and 2).

The second study found those patients using three or more SABA reliever canisters a year had an increased all-cause mortality risk relative to patients using fewer SABA canisters: hazard ratios after adjustment were 1.26 (95% CI 1.14–1.39) for 3–5 canisters annually, 1.67 (1.49–1.87) for 6–10 canisters, and 2.35 (2.02–2.72) for > 11 canisters, relative to patients collecting < 3 canisters annually [ 46 ].

The recently published PRACTICAL study lends further support to as-needed low-dose ICS/formoterol as an alternative option to daily low-dose ICS plus as-needed SABA, outlined in Step 2 of the guidelines [ 47 ]. In their one-year, open-label, multicentre, randomized, superiority trial in 890 patients with mild to moderate asthma, Hardy and colleagues found that the rate of severe exacerbations per patient per year (the primary outcome) was lower in patients who received as-needed budesonide/formoterol than in patients who received controller budesonide plus as-needed terbutaline (relative rate 0.69, 95% CI 0.48–1.00; p < 0.05). Indeed, they suggest that of these two treatment options, as-needed low-dose ICS/formoterol may be preferred over controller low-dose ICS plus as-needed SABA for the prevention of severe exacerbations in this patient population.

Step 3 recommendations have been left unchanged from 2017, whereas Step 4 treatment has changed from recommending medium/high-dose ICS/LABA [ 3 ] to medium-dose ICS/LABA; the high-dose recommendation has been escalated to Step 5. Patients who have asthma that remains uncontrolled after Step 4 treatment should be referred for phenotypic assessment with or without add-on therapy.

To summarise, the use of ICS medications is of paramount importance for optimal asthma control. The onset and increase of symptoms are indicative of a worsening inflammation leading to severe exacerbations, the risk of which is reduced by a maintenance plus as-needed ICS/LABA combination therapy. The inhaled ICS/bronchodilator combination is as effective as the regular use of inhaled steroids.

The efficacy of anti-inflammatory reliever therapy (budesonide/formoterol) versus current standard-of-care therapies in mild asthma (e.g. reliever therapy with a SABA as needed and regular maintenance controller therapy plus a SABA as-needed) has been evaluated in two randomized, phase III trials which confirmed that, with respect to as-needed SABA, the anti-inflammatory reliever as needed is superior in controlling asthma and reduces exacerbation rates, exposing the patients to a substantially lower glucocorticoid dose.

Conclusions

A growing body of evidence shows that anti-inflammatory reliever strategy is more effective than other strategies with SABA reliever in controlling asthma and reducing exacerbations across all levels of asthma severity. A budesonide/formoterol therapy exposes asthma patients to a substantially lower glucocorticoid dose while cutting the need for adherence to scheduled therapy.

Acknowledgements

The Authors thank Maurizio Tarzia and Gayle Robins, independent medical writers who provided editorial assistance on behalf of Springer Healthcare Communications. The editorial assistance was funded by AstraZeneca.

Abbreviations

GINAGlobal Initiative for Asthma
ICSInhaled corticosteroids
LABALong-acting beta-agonists
OCSOral corticosteroids
SABAShort-acting beta-agonists
SMARTSingle inhaler maintenance and reliever treatment

Authors’ contributions

AP, FB, GWC, LM, LR and AR contributed to writing. All authors read and approved the final manuscript.

No funding was received for this study. The editorial assistance was funded by AstraZeneca.

Availability of data and materials

Ethics approval and consent to participate, consent for publication, competing interests.

AP reports grants, personal fees, non-financial support and payment for advisory board membership, consultancy, payment for lectures, grants for research, and travel expenses reimbursement from Chiesi, AstraZeneca, GlaxoSmithKline, Boehringer Ingelheim, Mundipharma and Teva, and personal fees and non-financial support from Menarini, Novartis, Zambon and Sanofi.

FB reports having received in the last three years research grants as well as lecture or advisory board fees from: Alk-Abelló, AstraZeneca, Boehringer Ingelheim, Chiesi, Guidotti, Glaxo Smith Kline, Grifols, Menarini, Novartis, Sanofi, Valeas, Zambon.

GWC reports having received in the last 3 years research grants as well as lecture or advisory board fees from: A. Menarini, Alk-Abelló, AstraZeneca-Medimmune, Boehringer Ingelheim, Chiesi Farmaceutici, Genentech, Guidotti-Malesci, Glaxo Smith Kline, Hal Allergy, Merck Sharp & Dohme, Mundipharma, Novartis, Orion, Sanofi-Aventis, Sanofi Genzyme/Regeneron, Stallergenes-Greers, UCB Pharma, Uriach Pharma, Valeas.

LR Receipt of grants/research supports: Roche, Boehringer Ingelheim.

Receipt of honoraria or consultation fees: Boehringer Ingelheim, Roche, Biogen, FibroGen,

Sanofi-Aventis, Anthera, Promedior, ImmuneWorks, Asahi-Kasei, Bayer, Celgene, RespiVant,

Nitto, Bristol Myers Squibb, Prometic, Pliant Therapeutics, Toray, Global Blood Therapeutics,

Zambon, Veracyte, Acceleron, CSL Behring.

LM and AR reports no conflicts of interest in the last 3 years.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • View all journals
  • Explore content
  • About the journal
  • Publish with us
  • Sign up for alerts
  • Published: 10 September 2015
  • Stephen T. Holgate 1 , 2 ,
  • Sally Wenzel 3 ,
  • Dirkje S. Postma 4 ,
  • Scott T. Weiss 5 ,
  • Harald Renz 6 &
  • Peter D. Sly 7  

Nature Reviews Disease Primers volume  1 , Article number:  15025 ( 2015 ) Cite this article

77k Accesses

410 Citations

112 Altmetric

Metrics details

  • Inflammation
  • Respiratory tract diseases

Asthma is the most common inflammatory disease of the lungs. The prevalence of asthma is increasing in many parts of the world that have adopted aspects of the Western lifestyle, and the disease poses a substantial global health and economic burden. Asthma involves both the large-conducting and the small-conducting airways, and is characterized by a combination of inflammation and structural remodelling that might begin in utero . Disease progression occurs in the context of a developmental background in which the postnatal acquisition of asthma is strongly linked with allergic sensitization. Most asthma cases follow a variable course, involving viral-induced wheezing and allergen sensitization, that is associated with various underlying mechanisms (or endotypes) that can differ between individuals. Each set of endotypes, in turn, produces specific asthma characteristics that evolve across the lifecourse of the patient. Strong genetic and environmental drivers of asthma interconnect through novel epigenetic mechanisms that operate prenatally and throughout childhood. Asthma can spontaneously remit or begin de novo in adulthood, and the factors that lead to the emergence and regression of asthma, irrespective of age, are poorly understood. Nonetheless, there is mounting evidence that supports a primary role for structural changes in the airways with asthma acquisition, on which altered innate immune mechanisms and microbiota interactions are superimposed. On the basis of the identification of new causative pathways, the subphenotyping of asthma across the lifecourse of patients is paving the way for more-personalized and precise pathway-specific approaches for the prevention and treatment of asthma, creating the real possibility of total prevention and cure for this chronic inflammatory disease.

Similar content being viewed by others

research articles on asthma pdf

Longitudinal follow-up of the asthma status in a French–Canadian cohort

research articles on asthma pdf

Defining pediatric asthma: phenotypes to endotypes and beyond

research articles on asthma pdf

Genome-wide analysis highlights contribution of immune system pathways to the genetic architecture of asthma

Introduction.

The 2015 Global Strategy for Asthma Management and Prevention by the Global Initiative for Asthma (GINA) defined asthma as a heterogeneous disease characterized by chronic airway inflammation and variable remodelling that results in a range of clinical presentations, treatment responses and natural history across the lifecourse of the patient 1 . Asthma involves a history of respiratory symptoms — including wheeze, shortness of breath, chest tightness and cough — that vary over time and in intensity, variable expiratory airflow limitation and airway hyper-responsiveness to a range of stimuli, such as exercise and inhaled irritants. At the population level, a subset of individuals with asthma exhibit an accelerated decline in lung function over their lifetime 2 , which, in severe chronic disease, manifests as fixed airflow obstruction. This decline is especially prominent in late-onset asthma 3 . The origin and severity of asthma are driven by strong genetic and environmental factors. Although most cases of asthma begin in childhood in association with IgE-dependent sensitization to common environmental allergens 4 , asthma can also emerge later in life. Adult-onset asthma often occurs in the absence of allergy but can be accompanied by intolerance to NSAIDs, rhinosinusitis and nasal polyps 5 . Intolerance to NSAIDs most likely results from reduced production of the anti-bronchoconstrictor prostaglandin E 2 under conditions of inflammation.

Asthma is often accompanied by co-morbidities including multi-organ allergies, such as allergic rhinitis, conjunctivitis, atopic dermatitis and food allergy, as well as non-allergic disorders, such as obesity, gastro-oesophageal reflux and psychiatric conditions 6 . Asthma is subject to periods of rapid deterioration (or exacerbations) that are provoked by viral infection and exposure to allergens, air pollutants and certain drugs such as aspirin and other NSAIDs 7 . In addition, certain types of asthma can enter spontaneous remission (that is, patients become symptom-free), such as during late childhood and adolescence 8 , and can respond to allergen-specific immunotherapy through the acquisition of immunological tolerance 9 .

In both adults and children, asthma has been traditionally classified by either symptom severity or the extent of disease control achieved using a stepwise management process, in which patients are grouped into one of four or five categories that are used to determine treatment requirements with controller drugs. These drugs include inhaled corticosteroids (ICSs), long-acting β 2 -adrenergic receptor agonists (LABAs), long-acting muscarinic antagonists, leukotriene receptor antagonists (LTRAs) and, for the most severe disease, the IgE-specific monoclonal antibody omalizumab 1 . Although this stepwise approach has improved the management of asthma and reduced dependency on inhaled short-acting bronchodilators (SABAs) for symptom relief, none of these treatments have been shown to alter the natural history of the disease 10 , 11 . Cluster and other non-hierarchical analyses have identified subtypes of asthma associated with differing causal pathways, natural histories and responses to interventions 12 ( Fig. 1 ). In this Primer, we discuss the epidemiology, origins, pathophysiology, diagnosis and treatment of asthma with specific reference to disease heterogeneity and stratification according to causal pathways, and show how this is shaping a new personalized or stratified approach to treatment.

figure 1

New subphenotypes and associated causal pathways, or endotypes, of asthma are being discovered through the application of non-hierarchical statistical analyses of clinical, physiological and laboratory characteristics. Figure from Ref. 163 , Nature Publishing Group.

PowerPoint slide

Epidemiology

There are >300 million people in the world who are affected by asthma, making it one of the most common chronic diseases 13 . Although the prevalence of asthma is greatest in countries with a high gross domestic product, the disease is recognized worldwide. In the lowest-income and most rural countries 14 , the prevalence of asthma tends to be ≤1%, far lower than the 10% usually seen in developed western countries ( Fig. 2 ). Within populations of a given gross domestic product, the prevalence of asthma follows an urban–rural gradient and a weak latitudinal gradient, that is, there is greater disease prevalence with greater distance from the equator and asthma is more common in urban areas 14 . Despite the low prevalence of asthma in low-income and middle-income countries, underdiagnosis and misdiagnosis together with inadequate treatment in these regions leads to considerable, and potentially avoidable, disease morbidity and mortality.

figure 2

The prevalence of asthma has plateaued in recent decades in high-prevalence countries, which have a high gross domestic product (GDP). Conversely, there has been a steep increase in the prevalence of asthma in low-prevalence and intermediate-prevalence countries, which have low-ranking and middle-ranking GDPs, respectively. These steep increases have occurred alongside acquisition aspects of the western lifestyle in these countries 254 . Reprinted from Bulletin of World Health Organisation , 83, Bousquet, J., Bousquet, P. J., Godard, P. & Dyres, J.-P., The public health implications of asthma, 548–554, Copyright (2005).

The prevalence of asthma has increased in many parts of the world over the past few decades, and, until recently, asthma prevalence was increasing on a year-by-year basis in developed western countries ( Fig. 2 ). The cause of the epidemic that began in the late 1970s is unclear, but the rise in asthma prevalence is consistent with a rise in other immune-mediated diseases, such as type 1 diabetes mellitus, inflammatory bowel disease and multiple sclerosis. Recent epidemiological research has focused on changes in maternal diet during pregnancy — particularly on the levels of micronutrients such as ω-3 fatty acids, folate and vitamin D (the latter two modifying methylation), and hence fetal programming — along with the gut and airway microbiota, prematurity and maternal paracetamol use during pregnancy 15 .

Immunological factors, age and sex all influence the development of asthma. The disease is closely linked to the presence of immediate hypersensitivity, and 50% of children who are diagnosed with asthma by 3 years of age and 80% of those diagnosed by the time they are 6 years of age are atopic — that is, they are genetically predisposed to allergic hypersensitivity 16 . The prevalence of wheezing exceeds the prevalence of asthma in children up to 6 years of age. This observation suggests that factors other than asthma, such as physician diagnostic bias and lower respiratory tract infections, can drive the onset and persistence of wheezing. Asthma is not constant across the lifecourse of the patient, and patients can experience periods of remission and the onset of new asthma 17 . Whereas asthma is more common in boys than girls in early childhood, throughout puberty and early adulthood, boys experience asthma remission at a higher rate than in girls. In addition, girls acquire asthma more often than boys in this age period. Consequently, the sex ratio of asthma during childhood reverses in adolescence and in young adulthood 16 , 18 , 19 ( Fig. 3 ). The reasons for this variability in asthma across the lifecourse of the patient are not clear, but evidence is mounting in support of a key role for hormones in this process. Furthermore, asthma remission during adolescence is associated with lower initial airway hyper-responsiveness and greater gain in the function of small airways compared with asthma that begins after childhood 18 . The reasons for this variability between early-onset and late-onset asthma are probably complex, but differing environmental exposures (exposome) — including those that occur in occupational settings — are thought to be important 20 .

figure 3

Bars represent the proportion of either boys or girls at particular ages who have asthma, with light-shaded sections representing the proportion of new cases that did not carry over from the previous age group (positive transition); the boxes between bars indicate the percentage of children who grew out of asthma in the intervening years (negative transition or remission). Data from Ref. 18 .

The heterogeneity of asthma can pose challenges for epidemiological research. Cross-sectional studies of asthma can be difficult to interpret given that both recall bias and the fact that most patients with asthma will have had a varied disease course. As such, at any one time, individuals at different stages of their disease and with different pathophysiological mechanisms for their asthma might be affected. The exception to this is severe asthma, which usually has an onset in early childhood, is associated with multi-allergen sensitization 21 and persists across the lifecourse 22 . In addition, new asthma in older adults tends to be more severe from the onset than asthma that develops in younger age groups 23 . Finally, asthma might co-occur with chronic obstructive pulmonary disease (COPD) to cause asthma COPD overlap syndrome 1 . The prevalence of this overlap is substantial.

Mechanisms/pathophysiology

Pathophysiology.

Airway inflammation . Airway inflammation is a prominent feature of asthma ( Fig. 4 ). T2-type inflammation occurs in >80% of children and in the majority of adults with asthma in association with sensitization to environmental allergens, such as those from dust mites, fungi, pets and pollens 24 – 26 . This sensitization is often associated with other clinical manifestations of atopy such as atopic dermatitis (eczema), allergic rhinoconjunctivitis and food allergy. The inflammatory infiltrate that accompanies T helper 2 (T H 2) lymphocyte responses is mainly composed of eosinophils but also includes mast cells, basophils, neutrophils, monocytes and macrophages 27 . Cellular activation and release of inflammatory mediators in asthma is evidenced by mast cell degranulation and eosinophil vacuolation. The majority of mucosal mast cells in mild-to-moderate allergic-type asthma are of the T H 2 cell-dependant tryptase-expressing type (MC T ) 28 . In the more intractable forms of asthma, mast cells containing both tryptase and chymase (MC TC ) predominate, which are more dependent on stem cell factor (also known as KIT ligand) for their survival than are MC T cells 29 , 30 .

figure 4

Cross section of a severe asthmatic airway (right) compared with a normal airway (left). Asthma involves mucosal inflammation that most frequently consists of activated eosinophils, mast cells and T lymphocytes within the context of a remodelled airway with mucous metaplasia, an increase in smooth muscle (Sm), fibrosis and angiogenesis. Bm, basement membrane; Bv, blood vessel; Ep, epithelium. Republished with permission of Dove Medical Press, from Clinical update on the use of biomarkers of airway inflammation in the management of asthma. Wadsworth, S., Sin, D. & Dorscheid, D., 4, 2011; permission conveyed through Copyright Clearance Center, Inc.

The principal role of T cells in asthmatic airways is in controlling the inflammatory cell profile 31 . Whereas the activity of T H 2 CD4 + lymphocytes predominate in classic allergic-type asthma 32 , roles for a range of other T cells in different asthma subtypes have been described, including the association of T H 1 cells 33 and T H 17 cells 34 with neutrophilic asthma ( Fig. 5 ). In eosinophilic allergic asthma 35 , and potentially non-allergic asthma 36 , the initiation of T2-type immune responses occurs through secretion of the epithelial cell-derived cytokines IL-25, IL-33 and thymic stromal lymphopoietin (TSLP). These cytokines induce a new innate lymphoid subset (nuocytes, a type of group 2 innate lymphoid cells (ILC2s)) to produce the T2-type cytokines IL-5, IL-9 and IL-13 (Ref. 37 ) ( Fig. 6 ).

figure 5

Dendritic cells (DCs) and the thymic epithelium together trigger the immune response that drives the development of asthma. In response to a combination of signals transmitted by cytokines and direct contact, the DCs and thymic epithelium promote the differentiation of an array of different leukocyte subsets (inner, yellow circle). Before differentiation, these subsets either augment or protect the airways from inflammatory responses linked to asthma, whereas following differentiation, they generate cytokines that promote the development of asthma. These cytokines influence various different cell types and the attendant inflammatory responses to drive allergic airway inflammation and airway hyper-responsiveness. GM-CSF, granulocyte–macrophage colony-stimulating factor; ILC2, group 2 innate lymphoid cell; iT Reg , inhibitory regulatory T; NKT, natural killer T; TGFβ, transforming growth factor-β T H , T helper; TNF, tumour necrosis factor. Figure from Ref. 31 , Nature Publishing Group.

figure 6

Infection (bacterial and viral) and pollutants perturb the airway epithelium, which leads to initial danger signalling and activation of innate signalling receptors. This signalling causes airway epithelial cells (ECs) to secrete chemokines and leads to trafficking of immature dendritic cells (DCs) to the mucosal epithelium. These DCs respond to danger signals through pattern recognition receptors (PRRs), which leads to their maturation into competent antigen-presenting myeloid-type DCs. Allergen detection and processing by these activated DCs is mediated by the extension of cellular processes into the airways or by the capture of allergens that have breached the epithelium. Allergen-loaded DCs then drive T cell differentiation by migrating to local lymph nodes where they interact with naive T cells (T N ) via the T cell receptor (TCR), major histocompatibility complex (MHC) class II and co-stimulatory molecules. DC activation and T helper 2 (T H 2) cell maturation and migration into the mucosa are influenced by additional epithelial-derived cytokines and chemokines, including IL-25, IL-33, CC-chemokine ligand 17 (CCL17) and CCL22. CCR, CC-chemokine receptor; GM-CSF, granulocyte–macrophage colony-stimulating factor; mDC, mucosal DC; TNF, tumour necrosis factor; TSLP, thymic stromal lymphopoietin. Figure from Ref. 31 , Nature Publishing Group.

Allergen sensitization also requires an interaction between specialized antigen-presenting airway dendritic cells (DCs) and T cells. This mechanism involves processing of allergen into small peptides and the selective major histocompatibility complex (MHC) class II presentation of these processed peptides to the T cell receptors of naive T cells 31 ( Fig. 6 ). Effective allergen signalling also requires co-stimulatory interactions between DCs and T cells 38 that take place in local lymphoid collections 39 , resulting in T cell differentiation into T H 2-type T cells. These T H 2-type T cells secrete the pro-allergic cytokines, IL-3, IL-4, IL-5, IL-9, IL-13 and granulocyte–macrophage colony-stimulating factor (GM-CSF), which in turn leads to the IgE, mast cell and eosinophilic responses that are characteristic of allergic asthma 40 ( Fig. 5 ). Many of the asthma-related allergens — such as those from dust mite, cockroach, animal and fungal sources — exhibit enzymatic properties that enable them to penetrate the epithelial barrier and directly interact with mucosal DCs 41 ( Fig. 6 ). During this time, quiescent DCs transform to express an array of cell adhesion and co-stimulatory molecules. These molecules are recognized by naive T cells, which interact with DCs to create an immunological synapse that facilitates allergen presentation 42 . Whereas a minority of allergen-specific T H 2 cells migrate to the B cell follicle to initiate immunoglobulin class switching from IgM to IgE 43 , others relocate to the airway mucosa, under the influence of chemoattractants, to elicit the T2-type inflammatory response and the associated coordinated secretion of pro-allergic cytokines 44 .

Once sensitized, further exposure of the airways to allergen results in a mast-cell-driven early-type bronchoconstrictor response (EAR) that lasts for 5–90 minutes and involves IgE-dependent release of histamine, prostaglandin D 2 and the leukotriene C 4 (LTC 4 ), which is subsequently converted to LTD 4 and LTE 4 (Ref. 45 ) ( Fig. 5 ). The EAR is followed by a late-phase response (LAR) that evolves over 3–12 hours, and is linked to infiltration and activation of leukocytes (especially eosinophils) with further LTC 4 generation, T H 2 cytokine release from mast cells and T cells 46 and an increase in airway responsiveness 47 . Although EAR and LAR have been studied in humans and animal models to dissect the allergic mechanisms of asthma, these two responses are not accompanied by the chronic persistent inflammation that characterizes most cases of asthma. Consequently, EAR and LAR do not provide an adequate mechanistic setting within which to investigate exacerbations other than those triggered by allergen exposure 48 .

Although much of the focus in asthma pathophysiology has been on positive drivers of inflammation, the defective resolution of inflammation is emerging as a mechanism that might also be involved in asthma. The failure to adequately downregulate the inflammatory response could result in the prolonged survival of mast cells and eosinophils as a result of the cytokine milieu of the asthmatic airway. In addition, an important new paradigm in asthma pathophysiology is the potential role of lipoxins and resolvins as mediators of the endogenous resolution of inflammation. For instance, lipoxin A 4 can induce apoptosis of eosinophils and decrease the activity of ILC2s and natural killer lymphocytes, and the production of lipoxin A 4 is reduced in asthmatic airways 49 .

Airway remodelling . In asthma, the airway wall thickens in proportion to disease severity and duration 50 , 51 . This remodelling involves an increase in airway smooth muscle, thickening of the subepithelial reticular lamina, matrix deposition throughout the airway wall, angiogenesis, neuronal proliferation and epithelial mucous metaplasia — a process that involves the appearance of mucous cells in new areas of the airways and increased production of mucus ( Fig. 4 ). These events are thought to underlie airway hyper-responsiveness, whereas mucus forms plugs that can extend into the small airways and lead to air trapping and hyperinflation 52 . In addition, epithelial goblet cell metaplasia results from the actions of IL-4, IL-9 and IL-13, as well as from the secretion of growth factors such as members of the epidermal growth factor family, which cause epithelial cell stress and injury 53 .

Epithelial damage results from the separation of columnar cells from basal cells. This can be detected by staining of sputum from patients with asthma, showing detached columnar cells as Creola bodies. Thickening of the subepithelial basement membrane is confined to the reticular lamina and results from deposition of ‘repair-type’ collagens I, III, V and VI together with periostin, tenascin, osteopontin and fibronectin 54 – 56 . Subepithelial collagen is produced by a sheath of myofibroblasts that lie beneath the epithelium. An epithelial–mesenchymal trophic unit, located between the epithelial and smooth muscle layers of the airway, is established in response to epithelial cell injury 57 ( Fig. 7 ). Within the epithelial–mesenchymal trophic unit, the epithelium is a potent source of growth factors, including functionally active periostin 33 , platelet-derived growth factors 58 , fibroblast growth factors and members of the transforming growth factor-β (TGFβ) family. In addition, the epithelium is a source of members of the epidermal growth factor family 59 — which are capable of driving both fibrosis and smooth muscle proliferation — as well as neurotrophins 60 and angiogenic factors, such as vascular endothelial growth factors 61 . Together, these growth factors promote the neuronal and microvascular proliferation that accompany airway remodelling 62 . Enhanced growth factor production occurs as a direct consequence of epithelial injury and delayed repair, and in this respect resembles a chronic wound scenario 63 , 64 . Finally, migration of subepithelial microvascular pericytes 65 and proliferation of fibroblasts followed by their differentiation into myofibroblasts contribute to mucosal fibrosis, muscle hyperplasia 54 , 66 and the reduction in distance between airway smooth muscle cells and the epithelium 67 .

figure 7

In chronic moderate-to-severe asthma, the behaviour of the epithelium resembles that in chronic wound scenarios: it is more susceptible to environmental and viral injury than usual and exhibits impaired repair. In addition, the epithelium no longer undergoes healing by ‘primary intention’ but instead undergoes ‘secondary intention’ — a process that involves the production of growth factors that drive remodelling responses in the underlying airway wall. Similarly, the damaged and stimulated epithelium generates growth factors that contribute to goblet cell metaplasia. The augmented communication between the epithelium and the underlying mesenchyme resembles the activation of the epithelium–mesenchymal trophic unit that drives airway morphogenesis in the developing fetal lung. The resulting cytokine milieu also provides a favourable environment for sustaining chronic inflammation. This research was originally published in Clin. Sci. (Lond.) . Holgate, S. T., Arshad, H. S., Roberts, G. C., Howarth, P. H., Thurner, P. & Davies, D. E., A new look at the pathogenesis of asthma. Clin. Sci. (Lond.) . 2009; 118: 439–450 © Portland Press.

Disease onset

Childhood viral illness and lung function . A key trigger for the onset of asthma in children is severe wheezing in early life in response to viral infections, especially respiratory infection with syncytial virus (RSV) or rhinovirus. A second trigger is the emergence and then persistence of a T2-type allergic immune response in the airways ( Fig. 8 ). In the first 2 years of life, all children become infected with RSV and rhinovirus 68 , so the question is not whether infection is a causal factor in the onset of asthma, but whether there is an underlying developmental defect of the lungs and/or the innate immune system that confers asthma susceptibility.

figure 8

Asthma is caused by failure of the respiratory and immune systems to develop normally. This schematic represents asthma risk factors that operate at different stages of life. Asthma risk at birth is influence by genetic predispositions, impaired lung function and delayed immune maturation. Postnatal risk factors that increase asthma risk include reduced lung growth resulting in low lung function, the timing of acquisition of specific components of the pulmonary microbiota, repeated episodes of viral upper respiratory tract infections (URTIs) that spread to the lower airway and result in severe lower respiratory tract infections (LRTIs), maturational deficiencies in the innate and adaptive immune systems that increase the risk of severe LRTIs and favour primary allergic sensitization and repeated allergen exposure, resulting in persistent airway inflammation. The maternal gastrointestinal tract (GIT) microbiota is thought to influence priming of the fetal immune system, and the postnatal development of the infant GIT microbiota is influenced by early-life exposures. Although each individual pathway increases the risk of asthma, the major risk is produced when a child progresses through multiple risk pathways simultaneously.

For instance, an important risk factor for persistent asthma is low lung function, and most longitudinal cohort studies have found a deficit in lung function in children with asthma when lung function was first measured 69 – 71 . Lung function is partially influenced by genetics, and inherited factors might contribute to low lung function in those with asthma 72 , 73 . Exposure to environmental toxicants, such as cigarette smoke and ambient air pollution during pregnancy, is associated with reduced lung function at birth 74 – 76 and an increased risk of subsequent asthma ( Fig. 8 ). The debate over whether early-life infections damage the lungs (viral-induced effect) or whether such infections unmask vulnerable individuals (susceptible host) to cause asthma has not yet been settled. Severe viral infections that require hospitalization, especially infection with adenoviruses and RSV, can damage the developing lung and lead to recurrent respiratory problems including recurrent wheeze in childhood 77 , 78 . Longitudinal birth cohort studies have shown that wheezing in early life associated with rhinovirus infection is a risk factor for both subsequent asthma and lower lung function in childhood compared with infants who did not show rhinovirus-related wheezing 79 – 81 . However, none of these studies provide definitive evidence to determine whether these postnatal exposures increase the risk for asthma by limiting lung growth during early childhood.

Another key factor in the relationship between viral infection and developing asthma is the existence of impaired antiviral innate immunity in the airway epithelium. This impairment might include deficiency in mounting a robust type 1 (IFNβ) and type 3 (IFNλ) protective response when confronted by common respiratory viruses, such as rhinovirus, RSV, influenza virus, coronaviruses and adenoviruses 82 , 83 , and will be discussed in further detail in relation to viral exacerbations of asthma.

Prenatal and postnatal risk factors . Prenatal risk factors for the development of asthma include ethnicity, low socioeconomic status, stress, caesarean section and maternal tobacco smoking, whereas postnatal risk factors include the levels of endotoxins and allergens within the home, viral and bacterial infection, air pollution, antibiotic use, paracetamol exposure and obesity 84 . For example, prematurity confers a fourfold increase in the risk of developing asthma 85 , representing the largest effect of any known epidemiological risk factor for this disease 86 . In addition, increased airway responsiveness is present at birth. Given that this phenotype is known to be associated with prematurity and low birth weight, this physiological marker of asthma susceptibility is thus present in at-risk babies before any viral illness 74 , 85 , 87 . As such, airway responsiveness is an important determinant of the response to viral infections early in life and of ultimate asthma risk. However, with the exception of smoking, the identification of these risk factors has failed to translate into implementable public health policy.

Microbiota and the ‘hygiene hypothesis’ . Epidemiologists have also had difficulty in integrating knowledge about putative risk factors into a comprehensive theory about the developmental origins of the disease and the relationship of asthma to patient susceptibility to infection. One such attempt is the ‘hygiene hypothesis’, which was developed almost 25 years ago. This hypothesis posited that respiratory infections ‘protected’ against asthma and allergies by ‘educating’ the immature infant immune system 88 . However, although the potential protective effect of microorganisms on the development of allergy is an attractive idea, the mechanisms that underlie the negative association between microorganisms and allergy acquisition have proven to be complex and elusive 89 . Recent interest has focused on vitamin D 90 and the developing infant gut microbiota 91 , both of which might influence developmental asthma susceptibility and immune function through epigenetic mechanisms 92 , 93 . Evidence that supports a role for the maternal gastrointestinal microbiota in asthma development is also emerging. The maternal microbiota might affect the developing fetal immune system during pregnancy and influence respiratory health during infancy 94 . The infant gastrointestinal microbiota develops postnatally and is influenced by factors linked to asthma risk, such as mode of delivery, infant feeding practices, antibiotic exposure and exposure to siblings and pets. Whether the infant gastrointestinal microbiota is the ‘missing link’ between early-life environmental exposures and asthma risk is yet to be determined.

Bacterial pathogens . Along with a potential involvement of the intestinal microbiota in asthma, there is also increasing recognition that the presence of microorganisms in the respiratory tract, including the upper airways, and the way that the immune system responds to these, is likely to have an effect on respiratory health 95 ( Fig. 8 ). The presence of bacterial pathogens per se is not necessarily associated with disease risk, suggesting that an effective mechanism must normally operate to protect against trans -epithelial invasion in situations where local homeostasis is disturbed and barrier function is compromised, such as during viral infections. Immune recognition of invading bacteria has the potential to play such a part. For example, longitudinal cohort studies have determined that the development of IgG1 (T1-type immunity) and IgE (T2-type immunity) antibodies against bacterial antigens might limit tissue-damaging inflammatory responses to bacterial invasion across the respiratory epithelium 96 . These data indicate that a delayed postnatal rise in serum titres of IgG1 against Haemophilus influenzae and Streptococcus pneumoniae in children with a family history of atopy is associated with increased risk of sensitization to perennial aeroallergens and of persistent asthma at 5 years of age 97 , 98 . In this context, the likely role of specific IgG1 is to accelerate phagocytic clearance of microorganisms that breach the mucosal barrier, and thus limit potential tissue damage 99 . The presence of potentially pathogenic bacteria in the lower airways is likely to generate a vigorous inflammatory response associated with substantial lower respiratory symptoms, and might result in unbalanced T1-type immunity that increases the risk of developing asthma.

T2-type immunity against respiratory mucosal dwelling bacteria such as H. influenzae and S. pneunomiae , detectable as bacterial-specific serum IgE, is present in teenagers regardless of their atopic status. The strength of this immunity is inversely associated with asthma risk 100 . This bacterial-specific IgE is probably a surrogate marker for underlying populations of bacterial-specific T H 2 memory cells that secrete IL-4 and IL-13, which are pluripotent cytokines that probably have a role in downregulating bacterial-induced macrophage activation and thus limiting local tissue damage 96 . Exaggerated and unbalanced T2-type immunity is also probably associated with the eosinophilic inflammation and tissue damage observed in chronic asthma. Thus, deficiencies in the normal immune recognition of bacterial incursion across the respiratory epithelium, especially at times of respiratory viral infection that spreads to the lower airways, probably results in an unbalanced immune response that translates into an increased risk of subsequently developing asthma.

Airway modelling and remodelling . Although histological evidence of airway inflammation and modelling (or remodelling) in infants and young children with recurrent wheeze or asthma is limited, what little evidence there is suggests that recurrent wheeze is associated with thickening of the epithelial reticular basement membrane and T2-type airway inflammation 101 . These changes relate to symptom severity, the need for anti-asthma medication 102 and long-term evidence of airway inflammation 103 . It is noteworthy that thickening of the reticular lamina only begins with the onset of asthma, and in infants this thickening is not a feature of viral wheezing per se 104 . Perplexing questions are how and when the airways smooth muscle bulk increases and what is the relationship of these increases to the development of inflammation. There is evidence in support of the notion that smooth muscle hyperplasia accompanies early-onset childhood asthma 104 . However, studies in older children with asthma, cystic fibrosis or non-cystic fibrosis bronchiectasis have shown that the increases in smooth muscle mass in these children were similar to those observed in children without a lung disease 105 . This similarity suggests that a proportion of changes in smooth muscle mass might be related to chronic inflammation generally rather than being disease-specific. However, the remarkable hyper-responsive behaviour of asthmatic smooth muscle does suggest that there are highly specific changes in morphology linked with muscle function, such as an increase in the expression of oxidant pathways that changes the behaviour of muscle fibre contractility 106 . Recent data from challenge tests in adults with atopic asthma indicate that bronchoconstriction per se, regardless of the mechanism inducing it, increases the secretion of pro-fibrotic cytokines and the deposition of subepithelial collagen 107 . Thus, in addition to chronic inflammation and responses to tissue injury, altered mechanotransduction might be a further cause of airway wall remodelling. In this setting, remodelling might be promoted by induction of factors in the epithelium such as resistin-like-β (RELMβ), which accentuates collagen deposition 108 , members of the plasminogen activator system and chitinase-3-like protein 1 (CHI3L1; also known as YKL40) 109 , 110 .

Disease chronicity and exacerbations

Through a continuing cycle of epithelial injury and repair, chronic inflammation and airway remodelling occur in parallel to create the disease chronicity that is characteristic of asthma 57 , 111 . Superimposed on this chronicity is the acute worsening of asthma (also termed asthma exacerbation), which is most often driven by common respiratory viruses, allergen exposure and air pollutants. One possible explanation for why the asthmatic airways are so susceptible to usually innocuous viruses such as rhinovirus is that the epithelium, which undergoes repeated damage and repair, also exhibits an impaired innate immune response that involves a reduced capacity for induction of the protective interferons IFNβ and IFNλ in response to viruses. This defect is linked to the chronic wound status of the epithelium 112 , and inhaled IFNβ has recently been shown to attenuate viral exacerbations in moderate-to-severe asthma by restoring defective antiviral innate immunity 113 .

Remission of asthma

Up to one-third of children with asthma become disease free in young adulthood, with boys outgrowing asthma more frequently than girls 18 , 19 , 114 ( Fig. 3 ). Up to one-third of those who become asthma-free as young adults remain in remission throughout their adult life — that is, they remain symptom-free and have normal lung function in the absence of medication use. However, given that some evidence of airway inflammation remains in these individuals, it is debatable whether their lack of obvious symptoms indicates complete recovery as opposed to signifying clinical remission only 18 , 115 . Sensitization to small-particle allergens from animals with fur, especially from cats, is a risk factor for the persistence of asthma into adulthood. These allergens, such as major allergen I polypeptide chain 1 (allergen Fel dI) found in the saliva of cats 116 , can penetrate the small airways.

Complete remission from asthma (cure) can also occur, but in this case, airway hyper-responsiveness also resolves 117 – 119 . Although complete remission of asthma is accompanied by the normalization of lung function, which is usually measured by spirometry as forced expiratory volume in 1 second (FEV1), the small airways, which are known to contribute to asthma, have not been well studied in this context. In addition, bronchial biopsy studies have revealed that airway remodelling, defined by thickening of the reticular lamina of the epithelial basement membrane, persists in adults who experience complete remission of asthma, and these individuals also show some residual increase in the number of eosinophils compared with those who do not have asthma. However, the function of these residual eosinophils is important to note. In contrast to the eosinophils of patients with asthma, those present in individuals in complete remission are neither ‘activated’ nor are they easily attracted to the airway lumen in the absence of ICSs or by indirect stimuli such as inhaled adenosine 120 . These differences indicate that eosinophil activation and associated mediator secretion, rather than the mere presence of these inflammatory cells, is linked to the persistence of asthma.

Diagnosis, screening and prevention

Risk factors.

Environmental allergens . Children sensitized to aeroallergens early in childhood are more likely than their non-sensitized peers to experience persistent asthma later in life. In addition, seasonal allergic rhinitis and exposure to indoor allergens are associated with asthma development, severity and morbidity 121 . In particular, the relationship between asthma and exposure to dust mites is especially strong 121 – 123 , with >50% of children and adolescents with asthma sensitized to dust mites, whereas generally <20% of children without asthma are sensitized to dust mites.

Consequently, to prevent asthma onset, allergen avoidance is strongly recommended in cases of existing respiratory allergies. However, the role of allergen avoidance in preventing allergic asthma is poor, unless it is undertaken as part of a complex and multifaceted prevention effort 124 , 125 . The disappointing outcomes of indoor allergen reduction programmes either in pregnancy and early life (referred to as primary prevention) or once asthma is established (termed secondary prevention) are difficult to explain 126 – 128 . One potential reason for the failure of such efforts is that they might have simultaneously reduced exposure to protective factors, such as microbial products, which shape innate immunity by interacting with pattern recognition receptors, such as Toll-like receptors 129 . Indeed, microbial products present in indoor environments are protective against allergen sensitization if exposure occurs prenatally or in the first few years of life 130 , 131 . The mechanisms that underlie this protection might involve altered signalling through Toll-like receptors in the placenta and induction of tolerogenic regulatory T (T Reg ) cells in infants 89 , 132 .

Viral infection . Childhood viral infections, especially with rhinovirus and RSV, are associated with the development of asthma ( Fig. 8 ) and are the most common cause of asthma exacerbations ( Fig. 9 ). RSV is a major cause of bronchiolitis in the first year of life and an independent predictor of recurrent wheeze and early-childhood asthma 133 . With the advent of modern diagnostic technologies, rhinovirus-induced wheezing has also been recognized as a potential predictor of asthma development 134 . Several longitudinal and birth-cohort studies provide evidence that respiratory viral infections, particularly with rhinovirus that led to wheezing events during the first 3 years of life, are strong predictors of later asthma development 80 , 81 . A relative lack of production of type 1 and type 3 interferons together with the release of the T2-type alarmin cytokines TSLP, IL-25 and IL-33 by the airway epithelium provides a link between epithelial injury, enhanced allergen sensitization and the acquisition of asthma 82 , 83 , 135 ( Fig. 6 ). Initiation of asthma also involves preferential expression of susceptibility genes in different regions of the airway. These genes include protocadherin 1 (PCDH1) and cadherin 3 (CDH3), which are preferentially expressed in the airway epithelium 136 , 137 , and genes that are preferentially expressed in airway mesenchymal cells including ADAM33 (which encodes disintegrin and metalloproteinase domain-containing protein 33) (Ref. 138 ).

figure 9

Solid outline indicates that the role of these risk factors has been firmly established. Dotted outline indicates that there are controversial and/or preliminary results for the contribution of these risk factors. ETS, environmental tobacco smoke; HRV, human rhinovirus; NO 2 , nitric dioxide; O 3 , ozone; PM, particulate matter; RSV, respiratory syncytial virus; TRAP, traffic-related air pollution; VOCs, volatile organic compounds.

Air pollution . Indoor and outdoor air pollution, which have increased alongside urbanization and population growth, are important contributing factors to the development and exacerbation of asthma, particularly in the developing world 139 , 140 ( Fig. 9 ). The pollutants implicated in asthma are nitrogen dioxide, ozone, volatile organic compounds, particulate matter (PM 10 and PM 2.5 ) and traffic-related air pollution, which contains fresh vehicle exhaust pollutants and non-combustion-derived particles. These pollutants induce oxidative stress and epithelial damage to initiate or augment airway inflammation 141 , 142 and reduce inhibitory T Reg function 143 . Although controversial, such pathways are being linked to the initiation of asthma in those who otherwise would not experience the disease, as all of these pathways could contribute to enhanced respiratory sensitization to aeroallergens 144 and increased airway hyper-responsiveness and remodelling 145 . It has even been suggested that prenatal exposure to pollutants might contribute to postnatal asthma 146 . One possible mechanism in which pollutants could induce asthma involves interaction with genetic risk factors such as polymorphisms in antioxidant genes, for instance in those that encode various glutathione S-transferases, and in the TNF promoter 147 . In addition, pollutants might influence gene function through epigenetic pathways by affecting promoter methylation and histone acetylation status 148 .

Tobacco smoke . Exposure to tobacco smoke, which comprises a complex mixture of many volatile organic compounds and nitrogen dioxide, is an independent risk factor for the development of asthma 149 , 150 and acts, in part, by augmenting T2-type responses 151 ( Fig. 9 ). Even grandmaternal smoking during pregnancy increases the risk for asthma for the second generation 152 , thereby further incriminating intrauterine epigenetic mechanisms in asthma causation.

Obesity . Obesity during childhood is strongly associated with the incidence and severity of asthma 153 , and maternal obesity and high gestational weight gain are associated with an increased risk of childhood asthma, especially in non-asthmatic mothers 154 ( Fig. 9 ). A combination of mechanical factors and shared causal metabolic, hormonal and low-grade inflammatory pathways might help explain these associations. For instance, the obesity-associated chronic inflammatory response is defined by increased levels of inflammatory cytokines, such as TNF, IL-1 and IL-6, produced by macrophages and adipokines, such as leptin, chemerin and adiponectin, produced by adipocytes that inhibit cell proliferation and cause tissue damage. Leptin is a sentinel mediator of differentiation of lipofibroblasts and controls pulmonary surfactant synthesis in fetal lungs 155 . In mice, leptin infusion enhances airway hyper-responsiveness, increases the levels of IgE following allergen challenge 156 and increases airway levels of IL-6 (Ref. 157 ). A low-grade maternal inflammatory response with increased levels of leptin also contributes to postnatal risk of asthma 158 . However, the exact mechanism linking obesity and asthma still remains to be fully clarified.

Asthma is among the most common chronic diseases in the developed and developing world, but its diagnosis can be difficult. Although symptoms including wheeze, chest tightness and shortness of breath are often considered essential features of asthma in humans, the adage ‘all that is asthma does not wheeze and all that wheezes is not asthma’ holds true. Epidemiological studies that rely on ‘doctor diagnosis’ of asthma overestimate the true disease prevalence owing to misclassification 159 . A diagnosis of asthma usually begins when a child or adult presents with a range of spontaneous respiratory symptoms including recurrent cough and nocturnal awakening, along with symptoms triggered by external stimuli, such as allergens, viral infections, exercise and cold air. In adults, a history of asthma or recurrent ‘bronchitic episodes’ in childhood holds an important clue to diagnosis. However, a diagnosis of asthma should not be made on clinical characteristics alone. The definition of asthma requires a combination of appropriate clinical symptoms in association with documented reversible airflow limitation and/or airway hyper-responsiveness 160 . Although reversibility of airway obstruction and hyper-responsiveness are considered hallmarks of asthma, the sensitivity and specificity of the diagnositic criteria to identify these symptoms are poorly defined. Guidelines have added the important extra dimension of airway inflammation to the diagnosis of asthma, which is measured by eosinophil counts in sputum or blood and/or increased fractional exhaled nitric oxide (FeNO) 161 , 162 .

Physiologically determined abnormalities, such as reduced spirometry, are also of value in establishing an asthma diagnosis early in the course of the condition. Patients should be tested on a spirometer that is equipped with population normal values and, ideally, one that generates a flow-volume loop that can be evaluated for both inspiratory and expiratory effort. FEV1, forced vital capacity (FVC) and the FEV1/FVC ratio should be reported alongside reversibility of lung function with an inhaled SABA. Symptomatic asthma is often associated with a predicted FEV1 of <80% and an age-adjusted FEV1/FVC of <75%. Testing should be repeated after inhalation of a SABA to establish reversibility of airway obstruction, a hallmark of asthma, although as noted, both bronchodilator reversibility and peak expiratory flow (PEF) variability have poor sensitivity and specificity for the diagnosis of asthma. By convention, a diagnosis of asthma requires at least a 12% improvement in FEV1 over baseline and a total improvement of at least 200 ml. As asthma is frequently highly variable, normal spirometry results do not exclude the disease. Additional diagnostic aids include repeat testing over time and diurnal PEF monitoring using a portable PEF meter. If spirometry results remain normal, bronchial provocation testing with inhaled methacholine or mannitol should be considered to establish if airway hyper-responsiveness exists as another characteristic feature of asthma, although some variability in responses can be seen. To perform this test, the patient inhales increasing concentrations of the challenge substance until there is a ≥20% fall in the FEV1 from the saline control value. Each challenge agent has a threshold concentration for the fall that identifies asthma. Exercise testing or, as an alternative, eucapnic hyperventilation, which mimics the volume of air exchanged during exercise, is another method for uncovering hyper-responsiveness and is especially useful in diagnosing asthma in children.

The documentation of asthma-related airway inflammation is an important recent development in asthma diagnosis and is especially useful for ruling out asthma, as many diseases can produce asthma-like symptoms and provide positive results in the tests described above. Recent UK National Institute for Health and Care Excellence (NICE) guidelines for asthma diagnosis highlighted an urgent need to mainstream the use of inflammatory biomarkers, such as FeNO and sputum eosinophil counts, for reliable diagnosis of T2-type asthma 162 ( Fig. 10 ). If diagnosis is still questionable, then a practical approach is to treat the patient with medications that are appropriate to their level of severity defined by national or international asthma guidelines. If the patient's symptoms become markedly better in response to this treatment, then asthma is the likely cause. Possible other causes of wheezing and asthma-like symptoms should always be considered in the face of a poor response to a trial of treatment, and can include COPD, upper airway obstruction and laryngospasm in adults and viral-associated infection in children.

figure 10

This molecular phenotype, or endotype, of asthma can be identified using biomarkers. These include already established biomarkers (blue boxes) and markers currently under clinical evaluation (yellow boxes). iNOS, inducible nitric oxide synthase; NO, nitric oxide; T H , T helper.

Many different ‘subphenotypes’ of asthma with differing characteristics are becoming increasingly recognized 163 ( Fig. 1 ). In terms of disease classification, perhaps the most important distinction to make is whether the patient has evidence of an eosinophilic T2-type inflammatory process. This is important for disease management, as evidence is emerging that responses to ICSs and biologic therapies that target IgE, the IL-4–IL-13 pathway and IL-5 are all greater in patients with evidence of T2-type inflammation than in patients without evidence of T2-type inflammation 32 , 164 , 165 . A characteristic feature of this immune activation profile is the appearance of eosinophils in the blood and induced sputum as well as increased FeNO. ICS treatment monitored by sputum eosinophil testing is highly effective 166 , 167 , and both sputum and blood eosinophil counts are being used to effectively identify patients who might be responsive to biologic therapies that target IL-4, IL-13 and IL-5 (Refs 168 , 169 ). Measurement of sputum and blood eosinophilia is, unfortunately, not widely implemented 162 .

Nitric oxide produced by inducible nitric oxide synthase in the bronchial epithelium increases in response to IL-4 and IL-13, is a marker for T2-type inflammation and is highly corticosteroid sensitive 170 , 171 . Although measurement of the FeNO requires a specific analyser, the test itself is easy to perform, is reproducible and can be measured as a point-of-care biomarker with instant results. Elevated FeNO increases the likelihood of an asthma diagnosis involving T2-type inflammation and can be used as a predictor of and to follow therapeutic responses to biologics that are targeted at IgE, the IL-4 receptor, IL-13 and IL-5 (Refs 172 – 174 ) ( Fig. 10 ).

Expression of periostin, an extracellular matrix protein, is induced by IL-4 and IL-13 in airway epithelial cells and lung fibroblasts 175 , and periostin is secreted as a soluble peptide from the basolateral surface from which it gains access to the circulation 176 . Periostin functions as a ligand for α V β 3 and α V β 5 integrins to promote adhesion and migration of epithelial cells and aids in the crosslinkage of submucosal collagen 177 . As a T2-type immunity biomarker, reduced serum levels of periostin predict the clinical efficacy of biologics targeting the IL-4–IL-13 pathway and discriminate patients with high numbers of eosinophils in their airways 178 , 179 . As a high proportion of patients expressing T2-type airway inflammation are atopic, assessment of allergen-specific IgE in the serum provides information on patient-specific allergic triggers 180 .

Perhaps the most crucial use of these biomarkers will be to identify various molecular phenotypes of asthma, in particular severe asthma, where biologic agents are likely to be targeted. To date, the biomarkers identified are all linked to T2-type inflammatory phenotypes, which might either predict or be responsive to these T2-type-targeted therapies. Blood eosinophil counts of ∼ 150 per microlitre seem to both predict responses to IL-5-targeted therapies and fall in response to these treatments 181 . At present, it is unclear which biomarker will best predict response to IL-4–IL-13 pathway-targeted therapies, as all have shown some predictive ability. However, for IL-4–IL-13 pathway-targeted therapies, whereas blood eosinophil counts predict response, these counts do not decline in response to therapy 164 , 173 . It remains unclear whether any current biomarker, alone or in combination with a second or third biomarker, will identify patients who respond better to IL-5-specific as compared with IL-4–IL-13 pathway-specific or IgE-specific approaches. These biomarkers are linked closely to disease pathways, with less relation to treatment responses. As such, their inhibition by specific targeted biologic approaches and use to produce improved clinically meaningful outcomes might eventually translate what are currently molecular phenotypes into endotypes — which are definitively defined biological processes that underlie particular subgroups of the disease.

Most recently, a cross-sectional study of patients with asthma of varying severity and endobronchial tissue gene expression analysis has revealed three major patient clusters: T H 2-high, T H 17-high, and T H 2/T H 17-low 182 . In individual patient samples, T H 2-high and T H 17-high patterns were mutually exclusive and their gene signatures were inversely correlated and differentially regulated by IL-13 and IL-17A. In a mouse model of allergen-induced lung inflammation, IL-4–IL-13 blockade caused an increase in T H 17 cells and neutrophilic inflammation, whereas neutralization of IL-13 and IL-17 protected mice from eosinophilia, mucus metaplasia and airway hyper-responsiveness as well as causing an attenuation of neutrophilic inflammation. The authors conclude that combination therapy targeting both pathways may maximize therapeutic efficacy across a patient population comprising both T H 2 and T H 17 endotypes.

A crucial question is whether intervening on selective pathways prevents or reverses airway wall remodelling to influence the natural history of asthma. Although there is very limited evidence in support of the notion that anti-asthma therapy influences remodelling processes, a recent report showing that IgE blockade with omalizumab downregulates bronchial smooth muscle proteins in severe asthma provides justification for some optimism in the use of targeted biologics 183 .

Allergen avoidance . Although prevention of asthma, especially asthma associated with allergy, should be straightforward it has in fact proven difficult. Specific allergen avoidance and strategies with the aim to reduce asthma have produced disappointing results. This outcome could be taken to mean that allergic sensitization does not contribute to asthma, or it might also signify that the process of allergic sensitization in general, rather than sensitization to a specific allergen, is key to disease expression. For example, comprehensive (multifaceted) allergen avoidance that includes breastfeeding while the mother is on a low allergen diet or given an extensively hydrolysed formula in conjunction with dust mite reduction strategies in the first year of life seems to be most effective in preventing asthma onset in individuals who are genetically at risk of asthma, with protection extending to 18 years of age 124 . However, as this study was single-blinded in design, more studies are needed to confirm the benefits of multifaceted allergen avoidance.

Dietary measures . Breastfeeding is one of the best-studied asthma prevention measures. Whereas initial studies have provided some evidence for a reduction of the incidence of both allergy and childhood asthma 184 , 185 as a result of breastfeeding, these findings have recently been challenged 186 , 187 . Nonetheless, on the basis of the overwhelming general health benefits of breastfeeding, many guidelines still recommend exclusive breastfeeding during the first months of life 188 , 189 . Vitamin D levels might also influence asthma. A large trial aimed at testing whether vitamin D supplementation for pregnant women prevents their children from developing asthma has recently concluded, but its results are yet to be reported 190 . Finally, dietary supplementation with fish oil — an important source of long-chain polyunsaturated fatty acids — has received attention on account of the immune-modulatory activities of the altered and less-active eicosanoid derivatives that are metabolically produced instead of those derived from arachidonic acid. However, despite the theoretical advantages of fish oil supplementation, attempts to use fish oil to prevent asthma have been unsuccessful 191 , 192 .

Exposure to microorganisms . Microbial exposures have an essential role in priming immune responses, particularly early in life. Sources of exposure include certain allergy-protective and asthma-protective lifestyle conditions, such as traditional farms that contain environmental microorganisms — for example, high levels of Gram-positive and Gram-negative bacteria and of fungi 193 — and biodiverse environments found, for example, in rural areas. By contrast, urbanization is associated with both low biodiversity and an increase in the prevalence of asthma. Although epidemiological and experimental studies strongly support the concept that a decrease in exposure to diverse microorganisms causes increases in asthma prevalence 89 , 132 , this now needs to be translated into a clinical application for asthma prevention. Interventions that have aroused recent interest are the use of probiotics, which are live bacteria found in foods such as yoghurt or taken as supplements, and prebiotics, which are specialized plant fibres that promote the growth of ‘good bacteria’ already resident in the colon, especially in those living in rural environments with livestock farming. There have been a range of studies in which the gut microbiota has been changed by probiotics or prebiotics. For instance, prenatal and postnatal administration of certain probiotics decreases the risk of allergen sensitization 194 . However, although there have been claims there are some benefits of using probiotics to prevent atopic dermatitis 195 , results in asthma prevention have so far been disappointing 196 , 197 .

Finally, prevention strategies that can address other asthma risk factors include avoidance of active and passive tobacco smoke exposure, interventions that target obesity and achievement of a balanced diet comprising adequate micronutrients such as vitamin D. However, for these strategies to have their greatest effect, major public health measures that begin early in life and run as lifelong programmes are needed 198 .

Over the past decade, understanding of asthma has changed considerably, with better insights into its heterogeneity. Consequently, treatment has changed from being based on a ‘one-size-fits-all’ model to a more patient-centred or personalized approach 199 . Central to this development is supported self-management and provision of an individualized written asthma action plan, referred to in guidelines as the co-management of asthma 200 . This approach ensures that interventions are tailored to the needs of patients, such as prioritizing minimal absences from school and work, creation of a supportive partnership with their doctor, family support and use of the appropriate drug regimen and inhaler devices. Such co-management plans should provide the best quality of life (QOL) through minimizing disease symptoms and abolishing disease exacerbations 201 . As far as is possible, drug treatment should only be initiated after removal of the stimuli known to exacerbate the disease ( Box 1 ).

This management situation becomes more complex in children, especially in those <5 years of age, as many children who present with asthma symptoms have one of the various wheezing syndromes that, compared with asthma, are less likely to persist into adult life and do not require treatment with ICSs 202 . Unfortunately, the clinician has few tools available to determine which children are likely to have or develop persistent asthma and therefore benefit from early treatment.

In adults, GINA guidelines, which were updated in 2015, have provided sound recommendations to be used worldwide 1 , 203 . However, health services and treatment facilities, including the availability of drugs, differ between countries, and these disparities need to be taken into account when constructing action plans for asthma 204 .

Standard asthma treatment . Asthma guidelines such as those provided by GINA 1 , the US National Asthma Education and Prevention Program 205 and the British Thoracic Society 206 classify asthma by severity and extent of disease control. This classification is done in 4 or 5 steps that are used to assess the use of controller drugs, such as inhaled ICSs, LABAs, LTRAs and, for the most severe disease, the IgE-specific monoclonal antibody omalizumab ( Fig. 11 ). By starting ICSs as early as step 1 of the management plan and scaling controller therapy up in a stepwise manner, dependency on inhaled SABAs for symptom relief has been reduced, exacerbations prevented, asthma-related mortality reduced and QOL improved in the majority of patients with asthma. Combination ICS–LABA therapy in moderate-to-severe asthma has achieved considerable success, as has omalizumab in severe allergic asthma. Moreover, maintenance and rescue therapy with the combination of ICS and the rapidly acting LABA formoterol has shown beneficial effects when used as maintenance and reliever treatment in adult patients with asthma 207 . In those patients who remain symptomatic despite taking an ICS–LABA combination, adding a long-acting anti-muscarinic antagonist is beneficial 208 . Inhaled drugs are most frequently used on demand or twice daily. Novel ultra-long-acting bronchodilators have been developed to be used once daily 209 but their effectiveness is yet to be fully evaluated.

figure 11

The Global Initiative for Asthma (GINA) recommends that asthma is classified and treated according to 5 steps that reflect symptom severity and patient characteristics. The relative widths of each step reflect the number of patients who receive each treatment. In particular, dose–response studies demonstrate that the majority of patients with asthma should be able to be treated with low-dose inhaled corticosteroids (ICSs; step 2). The relative heights of each step reflect disease severity, with treatment in step 5 reserved for those with the most severe disease. *For children 6–11 years of age, theophylline is not recommended and the preferred step 3 treatment with is medium-dose ICSs. ‡ Tiotropium by soft-mist inhaler is indicated as add-on treatment for patients with a history of exacerbations; it is not indicated in patients <18 years of age. § For patients prescribed inhaled beclomethasone dipropionate/formoterol or budesonid/formoterol maintenance and reliever therapy. LABA, long-acting β 2 -adrenergic receptor agonist; LTRA, leukotriene receptor antagonist; OCS, oral corticosteroid; SABA, short-acting β 2 -receptor agonist. Adapted from the Global Strategy for Asthma Management and Prevention 2015, © Global Initiative for Asthma (GINA) all rights reserved. Available from http://www.ginasthma.org .

Despite the availability of a wide range of controller and reliever therapies, uncontrolled asthma remains a challenge. This problem reflects the need for new therapeutic options, especially as the number of deaths due to asthma has been reduced drastically since the introduction of anti-inflammatory treatments but, importantly, significant asthma-related mortality persists. Figure 12 shows that treatment should be based on both current control of a patient's asthma and reduction of future asthma risk. A recent meta-analysis compared methods for preventing severe disease exacerbations 210 and found that, for this purpose, both combined maintenance and reliever treatment and combined fixed-dose treatment with ICSs and a LABA performed equally well and were ranked first for effectiveness. All other treatment strategies that used ICSs with another agent, whether by single or separate inhalers, tended to perform better than low-dose ICSs, but the difference failed to achieve significance. In patients sensitized to a single allergen, for instance allergen Fel dI from cats or pectate lyase 1 (also known as antigen Amb a I) from ragweed, subcutaneous allergen immunotherapy is an option 211 . The effect of sublingual immunotherapy on asthma is still being evaluated 212 .

figure 12

Asthma management should aim to both control the current symptoms of asthma and reduce the risk of future adverse outcomes for patients. Adapted with permission from Ref. 256 , Elsevier.

Before deciding whether to start a new treatment or change treatment, health practitioners should reconsider whether there are any factors that are modifiable. These include incorrect use of an inhaler, which needs repeat instruction especially in cases of uncontrolled asthma, active smoking or passive smoke exposure, obesity, rhinitis, sinusitis, diabetes mellitus, gastro-oesophageal reflux, anxiety and depression, low vitamin D status, limited language proficiency, absence of specialty care, lack of an asthma action plan and lack of self-management education. Special consideration also needs to be given to managing asthma in pregnancy 213 .

Improvement in the treatment of asthma in adults requires that, in the future, study designs take into account a range of considerations. For example, they need to ensure that their findings have applicability for various settings and patient populations and that they have a patient-centred focus, such as that taken in pragmatic comparative effectiveness studies 214 . Nonetheless, although there is an ongoing need for further research in some areas, the evidence base in support of patient-centred and co-management approaches to asthma is strong 215 .

Novel therapeutic approaches . Cluster analyses have identified subtypes of asthma, one of which is severe asthma that is unresponsive to ICS and oral corticosteroid treatment 12 , 23 , 216 . Despite extensive research, the underlying pathobiology of severe asthma has not been established. Nonetheless the aetiology of severe asthma is likely to be heterogeneous rather than based on a single unifying process ( Fig. 1 ). As a result of increased awareness of this heterogeneity, the identification of appropriate biomarkers in individual patients with asthma is becoming crucial in guiding the use of therapies that target the specific causative pathways or endotypes of asthma 217 – 220 . Individual T2-type cytokine biologics that target the IL-4–IL-13, IL-5 and TSLP pathways and IgE itself are promising treatment options for severe disease, especially when used with biomarkers such as sputum and blood eosinophil numbers, FeNO, serum periostin levels and total IgE levels to differentiate ‘responders’ from ‘non-responders’. Blockade of a single pathway has clear clinical benefits and few associated adverse effects. However, whereas this strategy leads to improvement in some asthma outcomes, such as a reduction in asthma exacerbations produced with IL-5-specific therapy, outcomes for other measures, such as lung function and hyper-responsiveness, are less favourable. As a result, single pathway blockade has only partial efficacy 169 . Additional research is needed to define how such agents should be used in a stratified or personalized approach to management. The same is true for pharmacogenomics, in which single-nucleotide polymorphisms in genes contribute to treatment responsiveness. The proportion of phenotypic variability accounted for by genetic variation (heritability) is estimated to be 28.5% for bronchodilator responsiveness and 50% for lung function 221 . For example, the Arg16 allele of the gene encoding the β 2 -adrenergic receptor has been shown to associate with worsening of asthma when children with the disease receive continuous SABA or LABA monotherapy 222 , 223 . This and other variants of the β2-adrenergic receptor have different effects on the disease in different ethnic populations 224 . Polymorphisms also influence responsiveness to ICSs and LTRAs and deserve further study 225 .

Bronchial thermoplasty, delivered by the Alair ™ System, is a treatment for severe asthma that was approved by the US FDA in 2010. This treatment involves the delivery of controlled, therapeutic radiofrequency energy to the airway wall to heat the tissue and thereby reduce the amount of smooth muscle in the airway wall 226 . This procedure causes epithelial injury followed by regeneration of the epithelium, blood vessels, mucosa and nerves but not airway smooth muscle 227 , 228 . Indeed, there is preliminary evidence that this procedure is able to reduce airway smooth muscle mass in patients with severe asthma 228 . Although bronchial thermoplasty is an exciting development for managing some types of severe asthma, its place in management guidelines is still being evaluated 229 , 230 .

Although early treatment reduces asthma symptoms in those at high risk of developing persistent asthma, there is no evidence that early treatment decreases the risk of subsequent asthma or alters its natural history 10 , 11 , 202 , 231 , 232 . In addition, with the exceptions of treatment with omalizumab for children >12 years of age who have severe atopic asthma that is not controlled by maximal conventional therapy 233 and treatment with oral montelukast (a cysteinyl LTRA) for exercise-induced asthma 234 , so far, there is little evidence that ‘phenotype-specific’ therapy is helpful in children 235 . Treatment of asthma in school-aged children is perhaps the most straightforward, with most patients responding to simple first-line therapy that forms the basis of most asthma management plans. If first-line therapy is not successful, there is no single ‘best’ add-on therapy for all children. Thus, if the first add-on is not sufficient, it should be stopped and an alternative therapy of a different drug class tried in its place 236 .

The diagnosis and treatment of asthma in infants and preschool-aged children is much more problematic than it is for older children. As outlined earlier, although most severe childhood asthma starts early in life, the majority of infants and young children who have wheezing episodes do not progress to persistent asthma 237 . A further problem arises with the apparent lack of clinical or physiological response to bronchodilators in infants and young children with established asthma-related risk factors that predict long-term outcomes 238 . The aim of asthma management at all ages is to achieve good clinical control for as long as possible. Young children with persistent symptoms should be managed in a similar manner to that of older children with persistent symptoms. As pointed out in the GINA guidelines, for children ≤5 years of age 1 , 169 , a combination of increased daytime cough, daytime wheeze and night-time use of an inhaled SABA is a strong predictor of acute exacerbations in these children. Inhaled medications are the cornerstone of asthma management in younger children; however, substantial importance must be placed on the ability of the child to use the inhalation device. The use of ICSs as first-line management for children with persistent symptoms is supported by placebo-controlled clinical trials 239 . In contrast with adults, there is no evidence for the use of combination ICS–LABA therapy in this age group, with the GINA guidelines specifically advising against this in children ≤5 years of age.

The treatment of intermittent, usually viral-induced, symptoms in young children is more controversial. As discussed earlier, the use of ICSs or the LTRA montelukast as maintenance therapy does not reduce the frequency of acute intermittent symptoms in young children. Montelukast but not ICSs 169 , 239 , 240 or oral corticosteroids 241 given as a short-course treatment at the onset of symptoms can reduce the severity and duration of asthma symptoms, but do not reduce the need for additional medication or hospitalization. There is no justification for using a LTRA in infants 169 , 242 . Many young children can be treated with SABAs alone to relieve symptoms provided that symptoms are troublesome enough to warrant any treatment.

Box 1: Asthma triggers

Wherever possible, avoidance of triggers should be part of every patient's written asthma action plan. Common triggers include 253 :

Inflammatory factors

Respiratory infections

Temperature change

Strong odours

Stress and emotions

Food additives

Gastric reflux

Quality of life

For the vast majority of patients with asthma, life expectancy should not differ from the general population. Thus, treatment of asthma is primarily focused on improving the day-to-day symptoms of the patient, preventing exacerbations and generally improving their QOL ( Fig. 12 ). Indeed, GINA 1 identifies asthma control as the main outcome of asthma management. Asthma control incorporates level of symptoms, number and severity of exacerbations, as well as lung function. The effect of these factors on patients' activities and daily living is generally referred to as a patient's QOL. To address this more holistic approach, patient-reported outcome measures (PROMs; defined in Box 2 ) are becoming more attractive in the assessment of treatment success. These are typically short, self-completed questionnaires, most commonly used to measure the health status or health-related QOL of a patient before and after an intervention. The concept that PROMs are necessary to assess the impact of asthma treatment has contributed to their increasing use in clinical practice 243 , 244 . The PROMs generally used for the assessment of asthma are the asthma QOL questionnaire (AQLQ), the paediatric version of the AQLQ (PAQLQ), the Asthma Control Test (ACT) and the Asthma Control Questionnaire (ACQ) 245 . The ACT and ACQ focus on symptoms and functional status rather than how these affect the patient's personal perceptions of the effect of asthma on their day-to-day QOL. However, these outcomes are intimately related as studies show strong correlations between the level of asthma control and QOL, even when controlling for disease severity 246 .

QOL in patients with asthma is likely to be affected by various factors and co-morbidities associated with asthma, including asthma control (even when controlling for asthma severity), duration of disease and the concurrent need for chronic therapies, frequent exacerbations, obesity or weight gain and psychosocial factors 241 , 247 , 248 . In addition, sex and socioeconomic factors, as well as co-morbidities such as gastro-oesophageal reflux and rhinoinusitis also probably have a role in determining QOL in patients with asthma. These co-morbidities are particularly prevalent in more-severe asthma 6 . Patients with more-severe asthma experience an increase in symptom severity, more frequent and severe exacerbations and more co-morbidity, all of which contribute to an overall poor QOL.

Treatment with ICSs, usually with SABAs or ICSs in combination with a LABA, montelukast or omalizumab have all been reported to improve QOL, with varying effects on asthma control, when compared with a placebo. Interestingly, not all therapies affect these parameters in the same manner. For instance, although IgE-specific treatment improves exacerbations in patients with more-severe asthma, the mean improvement in QOL as measured by the AQLQ, while substantial, does not achieve the 0.5 points threshold change for clinical significance on the 7-point scale of this questionnaire 249 . The intensity of the intervention also seems to influence the magnitude of improvement in the AQLQ. This improvement is demonstrated by the invasive treatment bronchial thermoplasty; however, the difference in improvement between active and sham thermoplasty also did not achieve clinical significance 250 . Finally, it should be recognized that the mere label of asthma as a chronic disease, with lifelong required treatment and daily confrontation with the disease, even though controlled to some extent, might affect the QOL of an individual with asthma considerably, irrespective of disease severity 251 . Hence, all efforts should be made to prevent disease chronicity.

Box 2: Definition of a patient-reported outcome measure

A patient-reported outcome measure (PROM) is reported directly by a patient or their carer. It is a measure used to assess the outcome and/or the effect that a particular treatment for a long-term condition has on the quality of life of a patient. These reports might include how the condition affects health-related quality of life, a patient's perceptions of their health or functional status in relation to their long-term condition and the effect that treatment or care strategies have on the quality of life of the patient 244 .

Although substantial progress has been made in our understanding of asthma mechanisms and epidemiology, these are only just being translated into novel approaches for disease management. However, the prevention of asthma in at-risk infants and cure of the fully developed disease seem distant goals. All of our current therapies are still based on damping down airway inflammation and on relieving airway obstruction. To make further progress and recognizing that the majority of the disease begins early in life, we need to understand what the genetic, prenatal, maternal and early-life environmental factors are that initiate asthma or at least render particular infants more susceptible to it. A most promising field connecting these two drivers is the uncovering of gene functions by environmental exposures through epigenetics. There are already novel data about the relationship of the epigenome to maternal smoking; similar epigenetic studies looking at the plethora of chemicals that humans are now exposed to, such as air pollutants and endocrine disruptors, as well as diet might cast new light on preterm susceptibility. In terms of established asthma and disease severity, novel mechanisms are emerging to explain the way that cells adapt to environmental cues. These mechanisms include changes in DNA methylation, histone modifications and regulation of transcription and translation by non-coding RNAs and, when coupled with recent advances in lung delivery of oligonucleotides and small molecules, are opening up new opportunities for intervention 219 , 252 .

An important question stemming from lifecourse epidemiology is what limits the development of asthma in high-risk infants and, in particular, what role the maternal gut microbiota has in priming fetal immune development, predisposition to atopy and allergy and preventing viral-induced respiratory wheezing from progressing into subsequent asthma? Of equal importance is the infant upper airway and lung microbiota and understanding how different microbial signalling pathways modify innate immunity towards protection against or susceptibility to asthma. The role of metabolic pathways relating to diet, micronutrients, obesity and the microbiota and how all these interact with environmental exposures are also likely to be of great relevance.

From a mechanistic standpoint, the pathobiology of asthma is in large part restricted to the central and peripheral conducting airways, including inflammation and the structural changes referred to as airway modelling or remodelling, depending on the age of the patient. This has implications for lung development in utero and its association with large and small airway dysfunction and hyper-responsiveness that might already be present at birth in those likely to develop asthma. As the majority of asthma cases, at least in childhood, adopt a T2-type immune pattern, a key question is what drives the development of this immunological pathway as opposed to others. Adaptive immunity is just that. It has to be initiated by some other event (or events). Where does the initial defect (or defects) lie? Have we overemphasized altered immunity towards the allergic phenotype at the expense of abnormalities of the airway structural cells, such as epithelial cells, fibroblasts, nerves, microvasculature and smooth muscle, along with the modelling or remodelling components? Given that these airway components of adult asthma increase in response to mechanotransductive as well as inflammatory stimuli, is it possible that repeated wheeze in infancy can similarly initiate asthma in susceptible individuals?

Bronchial thermoplasty is an example of a disruptive technology that looks promising but needs further study to identify which patients benefit most from this invasive intervention. There is early evidence that this procedure targets smooth muscle, possibly suggesting that to overcome asthma more attention needs to be given to influencing modelling or remodelling pathways as well as halting inflammation. A crucial question that still needs answering is whether there is a mutual interdependency of airway inflammation (T2 type) and the onset of smooth muscle and other remodelling characteristics of asthma. If so, how and when does this occur? If interventions were able to alter the airway structural components (the ‘soil’) in which the T2-type inflammatory response (the ‘seed’) takes hold at the initiation of asthma, would we stand a better chance of preventing or even curing established disease?

Lifecourse epidemiology also indicates that severe asthma in early childhood continues throughout life. Low lung function at birth is a primary risk factor for asthma and, therefore, research into the causes of this both at birth and in early life is needed to uncover potentially modifiable exposures. Understanding the heterogeneity of severe asthma and using modern biomarker and statistical tools are opening up the field into asthma subtypes. This is a key focus of current research as differing causal pathways (endotypes) might be involved in different subtypes of asthma. New diagnostic tests to identify these subtypes and specific treatments, especially biologics, which target particular pathways represent new directions for asthma research where the focus is on identifying treatable disease traits. Thus, the proliferation of biologics now being trialled in severe asthma, although at present these only target components of the T2-type pathway (including IgE, IL-4, IL-13, IL-5 and TSLP), herald a new era in highly targeted, pathway-directed therapy that will require companion diagnostics to make them cost effective. Progress is also being made on the diagnostics front, with sputum and blood eosinophil counts, FeNO and levels of IgE and serum periostin all emerging as candidates. It now seems crucial that, even with current biomarkers, great attempts are made to place inflammation at the forefront of asthma diagnosis and management as a treatable trait that complements PROMs, symptoms and lung function. Implementation research is required to embed such a change so that laboratories and practices routinely offer such tests. Targeted therapy will increase in use as new causative endotypes are discovered, but even with current anti-inflammatory therapy (ICSs and LTRAs), biomarker monitoring needs to become an integral part of assessing the patients' treatment response.

As in adults, the availability of these novel, mostly biologic, therapies offers new opportunities for managing asthma in children. If intervention on the T2-type pathway took place early in life, could the natural history of asthma be influenced or the disease cured? Only clinical trials dedicated to asthma in children will answer this key question. There are already encouraging results for IgE-specific and allergen immunotherapy, with the possibility of halting the ‘allergic march’. If only we knew more about what happens when children lose their asthma in adolescence, and likewise how adults can lose their asthma, then new tractable therapeutic pathways might emerge.

Turning to asthma in adults, we need better treatments for asthma exacerbations that are linked in large part to viral infections. Death from asthma is almost always avoidable as it is usually owing to undertreatment and lack of adherence to the use of anti-inflammatory controller medications. It is clear that better integrative management, based on chronic diseases education for patients, family doctors, nurses and pharmacists, and possibly supported by shared decision making and smartphone or tablet computer applications for the delivery of asthma self-management programmes, is required.

At a more mechanistic level, increased understanding of the underlying cellular and molecular events that predispose individuals with asthma to exacerbations is required. This should involve further study not only on viral infection but also on the role of air pollution and other environmental exposures, including stress in exacerbating asthma symptoms. In addition, although late-onset asthma remains enigmatic, asthma subphenotyping has identified several different phenotypic clusters linked to different causal pathways. Very little progress has been made to date on understanding how these types of asthma begin, although, as in children, there seems to be a link to an initial or persistent viral infection. There is an urgent need to understand more about this role of viral infection, the interactions with sex hormones, including why and how women and girls are more affected by asthma over the lifecourse, and the different inflammatory asthma profiles, including T2 type, T17 type and T2/T17-low type.

The future for asthma treatment very much looks as if it is moving along the personalized (P4 or stratified) approach to health care. If successful, this approach of targeting the right intervention to the right patient at the right time will create new opportunities for both prevention and cure.

Global Initiative for Asthma. Global strategy for asthma management and prevention GINA [online] , (2015).

Lange, P., Parner, J., Vestbo, J., Schnohr, P. & Jensen, G. A 15-year follow-up study of ventilatory function in adults with asthma. N. Engl. J. Med. 339 , 1194–1200 (1998).

Article   CAS   PubMed   Google Scholar  

Porsbjerg, C., Lange, P. & Ulrik, C. S. Lung function impairment increases with age of diagnosis in adult onset asthma. Respir. Med. 109 , 821–827 (2015).

Article   PubMed   Google Scholar  

Custovic, A. To what extent is allergen exposure a risk factor for the development of allergic disease? Clin. Exp. Allergy 45 , 54–62 (2015).

Amelink, M. et al . Severe adult-onset asthma: a distinct phenotype. J. Allergy Clin. Immunol. 132 , 336–341 (2013).

Ledford, D. K. & Lockey, R. F. Asthma and comorbidities. Curr. Opin. Allergy Clin. Immunol. 13 , 78–86 (2013).

Custovic, A. et al . EAACI position statement on asthma exacerbations and severe asthma. Allergy 68 , 1520–1531 (2013).

Fu, L. et al . Natural progression of childhood asthma symptoms and strong influence of sex and puberty. Ann. Am. Thorac. Soc. 11 , 939–944 (2014).

Article   PubMed   PubMed Central   Google Scholar  

Soyka, M. B., van de Veen, W., Holzmann, D., Akdis, M. & Akdis, C. A. Scientific foundations of allergen-specific immunotherapy for allergic disease. Chest 146 , 1347–1357 (2014).

Murray, C. S., Woodcock, A., Langley, S. J., Morris, J. & Custovic, A. Secondary prevention of asthma by the use of Inhaled Fluticasone propionate in Wheezy INfants (IFWIN): double-blind, randomised, controlled study. Lancet 368 , 754–762 (2006). This longitudinal birth-cohort study, along with similar studies, failed to show that regular ICSs administered to high-risk infants with viral-associated wheeze before they fully developed asthma had any effect in protecting children from developing asthma by 5 years of age.

Murray, C. S. Can inhaled corticosteroids influence the natural history of asthma? Curr. Opin. Allergy Clin. Immunol. 8 , 77–81 (2008).

Moore, W. C. et al . Identification of asthma phenotypes using cluster analysis in the Severe Asthma Research Program. Am. J. Respir. Crit. Care Med. 181 , 315–323 (2010).

World Health Organization. Global surveillance, prevention and control of chronic respiratory diseases: a comprehensive approach. WHO [online] , (2007).

Wang, D. et al . Cross-sectional epidemiological survey of asthma in Jinan, China. Respirology 18 , 313–322 (2013).

Jackson, D. J., Hartert, T. V., Martinez, F. D., Weiss, S. T. & Fahy, J. V. Asthma: NHLBI workshop on the primary prevention of chronic lung diseases. Ann. Am. Thorac. Soc. 11 , S139–S145 (2014).

Yunginger, J. W. et al . A community-based study of the epidemiology of asthma. Incidence rates,1964–1983. Am. Rev. Respir. Dis. 146 , 888–894 (1992). This population-based survey showed that the median age of onset of asthma was 3 years for males and 8 years for females. These findings demonstrated that asthma begins in early childhood, with a higher incidence and earlier onset in males, and that the increase in incidence rates from 1964 to 1983 occurred only in children and adolescents.

Butland, B. K. & Strachan, D. P. Asthma onset and relapse in adult life: the British 1958 birth cohort study. Ann. Allergy Asthma Immunol. 98 , 337–343 (2007).

Arshad, S. H. et al . Pathophysiological characterization of asthma transitions across adolescence. Respir. Res. 15 , 153 (2014).

Szefler, S. J. Advances in pediatric asthma in 2014: moving toward a population health perspective. J. Allergy Clin. Immunol. 135 , 644–652 (2015).

Dumas, O. et al . Occupational irritants and asthma: an Estonian cross-sectional study of 34,000 adults. Eur. Respir. J. 44 , 647–656 (2014).

Simpson, A. et al . Beyond atopy: multiple patterns of sensitization in relation to asthma in a birth cohort study. Am. J. Respir. Crit. Care Med. 181 , 1200–1206 (2010).

Tai, A. et al . Outcomes of childhood asthma to the age of 50 years. J. Allergy Clin. Immunol. 133 , 1572–1578.e3 (2014).

Wu, W. et al . Unsupervised phenotyping of Severe Asthma Research Program participants using expanded lung data. J. Allergy Clin. Immunol. 133 , 1280–1288 (2014). In this study, unsupervised clustering approaches were applied to clinical, physiological and inflammatory variables from 378 individuals with asthma. Ten variable clusters and six subject clusters were identified, which differed and overlapped with previous clusters.

Sporik, R., Holgate, S. T., Platts-Mills, T. A. & Cogswell, J. J. Exposure to house-dust mite allergen ( Der p I) and the development of asthma in childhood — a prospective study. N. Engl. J. Med. 323 , 502–507 (1990). This highly cited study of 67 children from south England carried out in 1989 showed that the level of exposure in early childhood to household dust mite allergens is an important determinant of the subsequent development of asthma.

Permaul, P. et al . Allergens in urban schools and homes of children with asthma. Pediatr. Allergy Immunol. 23 , 543–549 (2012).

Olivieri, M. et al . Risk factors for new-onset cat sensitization among adults: a population-based international cohort study. J. Allergy Clin. Immunol. 129 , 420–425 (2012).

Djukanovic, R. et al . Mucosal inflammation in asthma. Am. Rev. Respir. Dis. 142 , 434–457 (1990).

Fajt, M. L. & Wenzel, S. E. Mast cells, their subtypes, and relation to asthma phenotypes. Ann. Am. Thorac. Soc. 10 , S158–S164 (2013).

Brightling, C. E. et al . Mast-cell infiltration of airway smooth muscle in asthma. N. Engl. J. Med. 346 , 1699–1705 (2002). Asthma and eosinophilic bronchitis are characterized by similar inflammatory infiltrates in the submucosa of the lower airway. In this airway biopsy study, the main differentiating feature between the two conditions was the presence in asthma of MCTs between muscle bundles, whereas the location and number of eosinophils and T cells were similar in the two diseases.

Balzar, S. et al . Mast cell phenotype, location, and activation in severe asthma. Data from the Severe Asthma Research Program. Am. J. Respir. Crit. Care Med. 183 , 299–309 (2011).

Holgate, S. T. Innate and adaptive immune responses in asthma. Nat. Med. 18 , 673–683 (2012).

Fahy, J. V. Type 2 inflammation in asthma — present in most, absent in many. Nat. Rev. Immunol. 15 , 57–65 (2015).

Article   CAS   PubMed   PubMed Central   Google Scholar  

Truyen, E. et al . Evaluation of airway inflammation by quantitative Th1/Th2 cytokine mRNA measurement in sputum of asthma patients. Thorax 61 , 202–208 (2006).

Chesné, J. et al . IL-17 in severe asthma. Where do we stand? Am. J. Respir. Crit. Care Med. 190 , 1094–1101 (2014).

Neill, D. R. et al . Nuocytes represent a new innate effector leukocyte that mediates type-2 immunity. Nature 464 , 1367–1370 (2010). This important publication shows that nuocyte (ILC2) numbers expand in vivo in response to the T2-type-inducing cytokines IL-25 and IL-33, and represent the predominant early source of IL-13 during helminth infection with Nippostrongylus brasiliensis. In the combined absence of IL-25 and IL-33 signalling, nuocytes fail to expand, resulting in a severe defect in worm expulsion that is rescued by the adoptive transfer of in vitro-cultured wild-type, but not IL-13-deficient, nuocytes.

Kim, H. Y., DeKruyff, R. H. & Umetsu, D. T. The many paths to asthma: phenotype shaped by innate and adaptive immunity. Nat. Immunol. 11 , 577–584 (2010).

Martinez-Gonzalez, I., Steer, C. A. & Takei, F. Lung ILC2s link innate and adaptive responses in allergic inflammation. Trends Immunol. 36 , 189–195 (2015).

Jain, V. V., Perkins, D. L. & Finn, P. W. Costimulation and allergic responses: immune and bioinformatic analyses. Pharmacol. Ther. 117 , 385–392 (2008).

Lambrecht, B. N. & Hammad, H. The immunology of asthma. Nat. Immunol. 16 , 45–56 (2014).

Article   CAS   Google Scholar  

Kay, A. B. The role of T lymphocytes in asthma. Chem. Immunol. Allergy 91 , 59–75 (2006).

Jacquet, A. Interactions of airway epithelium with protease allergens in the allergic response. Clin. Exp. Allergy 41 , 305–311 (2011). This excellent review draws attention to the interactions of protease allergens with the respiratory epithelium in facilitating their antigen presentation through epithelial barrier degradation, activating airway mucosal surfaces to recruit inflammatory cells and initiating airway remodelling. The authors suggest that a greater understanding of the effects of protease allergens on airway inflammation and on the relevant targets could define novel therapeutic strategies for the treatment of allergic asthma.

Lambrecht, B. N. & Hammad, H. Biology of lung dendritic cells at the origin of asthma. Immunity 31 , 412–424 (2009).

Kemeny, D. M. The role of the T follicular helper cells in allergic disease. Cell. Mol. Immunol. 9 , 386–389 (2012).

Perros, F., Hoogsteden, H. C., Coyle, A. J., Lambrecht, B. N. & Hammad, H. Blockade of CCR4 in a humanized model of asthma reveals a critical role for DC-derived CCL17 and CCL22 in attracting Th2 cells and inducing airway inflammation. Allergy 64 , 995–1002 (2009).

Bradding, P., Walls, A. F. & Holgate, S. T. The role of the mast cell in the pathophysiology of asthma. J. Allergy Clin. Immunol. 117 , 1277–1284 (2006).

Ma, L. L. & O'Byrne, P. M. The pharmacological modulation of allergen-induced asthma. Inflammopharmacology 21 , 113–124 (2013).

Hargreave, F. E., Dolovich, J., O'Byrne, P. M., Ramsdale, E. H. & Daniel, E. E. The origin of airway hyperresponsiveness. J. Allergy Clin. Immunol. 78 , 825–832 (1986).

Diamant, Z. et al . Inhaled allergen bronchoprovocation tests. J. Allergy Clin. Immunol. 132 , 1045–1055.e6 (2013). This is a comprehensive review of the allergen provocation tests used to measure allergen-related bronchial hyper-responsiveness.

Barnig, C. et al . Lipoxin A4 regulates natural killer cell and type 2 innate lymphoid cell activation in asthma. Sci. Transl. Med. 5 , 174ra26 (2013).

Barbers, R. G. et al . Near fatal asthma: clinical and airway biopsy characteristics. Pulm. Med. 2012 , 829608 (2012).

Al-Muhsen, S., Johnson, J. R. & Hamid, Q. Remodeling in asthma. J. Allergy Clin. Immunol. 128 , 451–462 (2011).

Bjermer, L. The role of small airway disease in asthma. Curr. Opin. Pulm. Med. 20 , 23–30 (2014). This is a review summarizing the literature showing that inflammation and remodelling occur from the central to peripheral airways in asthma.

Boucherat, O., Boczkowski, J., Jeannotte, L. & Delacourt, C. Cellular and molecular mechanisms of goblet cell metaplasia in the respiratory airways. Exp. Lung Res. 39 , 207–216 (2013).

Roche, W. R., Beasley, R., Williams, J. H. & Holgate, S. T. Subepithelial fibrosis in the bronchi of asthmatics. Lancet 1 , 520–524 (1989).

Kanemitsu, Y. et al . Osteopontin and periostin are associated with a 20-year decline of pulmonary function in patients with asthma. Am. J. Respir. Crit. Care Med. 190 , 472–474 (2014).

Liesker, J. J. W. et al . Reticular basement membrane in asthma and COPD: similar thickness, yet different composition. Int. J. Chron. Obstruct. Pulmon. Dis. 4 , 127–135 (2009).

CAS   PubMed   PubMed Central   Google Scholar  

Holgate, S. T. et al . Epithelial–mesenchymal communication in the pathogenesis of chronic asthma. Proc. Am. Thorac. Soc. 1 , 93–98 (2004). This review proposes the hypothesis that epithelial damage in patients with asthma initiates aberrant communication, mediated by growth factors, between the epithelium and the underlying myofibroblast sheath to drive airway remodelling, which is termed activation of the epithelial–mesenchymal trophic unit.

Chanez, P. et al . Platelet-derived growth factor in asthma. Allergy 50 , 878–883 (1995).

Puddicombe, S. M. et al . Involvement of the epidermal growth factor receptor in epithelial repair in asthma. FASEB J. 14 , 1362–1374 (2000).

Olgart Höglund, C. et al . Nerve growth factor levels and localisation in human asthmatic bronchi. Eur. Respir. J. 20 , 1110–1116 (2002).

Lopez-Guisa, J. M. et al . Airway epithelial cells from asthmatic children differentially express proremodeling factors. J. Allergy Clin. Immunol. 129 , 990.e6–997.e6 (2012). Using nasal epithelial cells, these investigators showed that, in asthma, such cells cultured in vitro differentially express the growth factors TGFβ2, vascular endothelial growth factor and periostin when compared with cells from atopic non-asthmatic and healthy children. They suggest that nasal epithelial cells might be a suitable surrogate for bronchial cells that could facilitate investigation of the airway epithelium in future longitudinal paediatric studies.

Harkness, L. M., Ashton, A. W. & Burgess, J. K. Asthma is not only an airway disease, but also a vascular disease. Pharmacol. Ther. 148 , 17–33 (2015).

Puddicombe, S. M. et al . Increased expression of p21 waf cyclin-dependent kinase inhibitor in asthmatic bronchial epithelium. Am. J. Respir. Cell. Mol. Biol. 28 , 61–68 (2003).

Alcala, S. E. et al . Mitotic asynchrony induces transforming growth factor-β1 secretion from airway epithelium. Am. J. Respir. Cell. Mol. Biol. 51 , 363–369 (2014).

Johnson, J. R. et al . Pericytes contribute to airway remodeling in a mouse model of chronic allergic asthma. Am. J. Physiol. Lung Cell. Mol. Physiol. 308 , L658–L671 (2015).

Descalzi, D. et al . Importance of fibroblasts–myofibroblasts in asthma-induced airway remodeling. Recent Pat. Inflamm. Allergy Drug Discov. 1 , 237–241 (2007).

Pepe, C. et al . Differences in airway remodeling between subjects with severe and moderate asthma. J. Allergy Clin. Immunol. 116 , 544–549 (2005).

Glezen, W. P., Taber, L. H., Frank, A. L. & Kasel, J. A. Risk of primary infection and reinfection with respiratory syncytial virus. Am. J. Dis. Child. 140 , 543–546 (1986).

CAS   PubMed   Google Scholar  

Turner, S. et al . A longitudinal study of lung function from 1 month to 18 years of age. Thorax 69 , 1015–1020 (2014). This article describes longitudinal tracking of lung function from birth to young adult life. This comes from the family of studies linking lung function and airway responsiveness in infancy to asthma risk in later life.

Morgan, W. J. et al . Outcome of asthma and wheezing in the first 6 years of life: follow-up through adolescence. Am. J. Respir. Crit. Care Med. 172 , 1253–1258 (2005).

Robertson, C. F. Long-term outcome of childhood asthma. Med. J. Aust. 177 , S42–S44 (2002).

PubMed   Google Scholar  

Van Putte-Katier, N. et al . Relationship between parental lung function and their children's lung function early in life. Eur. Respir. J. 38 , 664–671 (2011).

Soler Artigas, M. et al . Genome-wide association and large-scale follow up identifies 16 new loci influencing lung function. Nat. Genet. 43 , 1082–1090 (2011).

Young, S. et al . The influence of a family history of asthma and parental smoking on airway responsiveness in early infancy. N. Engl. J. Med. 324 , 1168–1173 (1991). This landmark paper demonstrated airway responsiveness in infancy related to a family history of asthma. Subsequent studies in this cohort have shown this responsiveness to be highly predictive of persistent asthma in later life.

Latzin, P., Röösli, M., Huss, A., Kuehni, C. E. & Frey, U. Air pollution during pregnancy and lung function in newborns: a birth cohort study. Eur. Respir. J. 33 , 594–603 (2009).

Stick, S. M., Burton, P. R., Gurrin, L., Sly, P. D. & LeSouëf, P. N. Effects of maternal smoking during pregnancy and a family history of asthma on respiratory function in newborn infants. Lancet 348 , 1060–1064 (1996).

Sly, P. D., Soto-Quiros, M. E., Landau, L. I., Hudson, I. & Newton-John, H. Factors predisposing to abnormal pulmonary function after adenovirus type 7 pneumonia. Arch. Dis. Child. 59 , 935–939 (1984). This study links infections to reduced lung function in childhood, a factor that was later shown to be associated with the development of asthma.

Sly, P. D. & Hibbert, M. E. Childhood asthma following hospitalization with acute viral bronchiolitis in infancy. Pediatr. Pulmonol. 7 , 153–158 (1989).

Guilbert, T. W. et al . Decreased lung function after preschool wheezing rhinovirus illnesses in children at risk to develop asthma. J. Allergy Clin. Immunol. 128 , 532–538.e10 (2011).

Jackson, D. J. et al . Wheezing rhinovirus illnesses in early life predict asthma development in high-risk children. Am. J. Respir. Crit. Care Med. 178 , 667–672 (2008). This study highlights the major risk for asthma in children who continue to wheeze with acute rhinovirus infections into their third year of life.

Jackson, D. J. et al . Evidence for a causal relationship between allergic sensitization and rhinovirus wheezing in early life. Am. J. Respir. Crit. Care Med. 185 , 281–285 (2012).

Wark, P. A. B. et al . Asthmatic bronchial epithelial cells have a deficient innate immune response to infection with rhinovirus. J. Exp. Med. 201 , 937–947 (2005).

Edwards, M. R. et al . Impaired innate interferon induction in severe therapy resistant atopic asthmatic children. Mucosal Immunol. 6 , 797–806 (2013).

Belsky, D. W. & Sears, M. R. The potential to predict the course of childhood asthma. Expert Rev. Respir. Med. 8 , 137–141 (2014).

Chan, K. N. et al . Airway responsiveness in low birthweight children and their mothers. Arch. Dis. Child. 63 , 905–910 (1988).

Sonnenschein-van der Voort, A. M. et al . Preterm birth, infant weight gain, and childhood asthma risk: a meta-analysis of 147,000 European children. J. Allergy Clin. Immunol. 133 , 1317–1329 (2014).

Kennedy, J. D. Lung function outcome in children of premature birth. J. Paediatr. Child Health 35 , 516–521 (1999).

Strachan, D. P. Hay fever, hygiene, and household size. BMJ 299 , 1259–1260 (1989).

Renz, H. et al . Gene–environment interactions in chronic inflammatory disease. Nat. Immunol. 12 , 273–277 (2011).

Litonjua, A. A. & Weiss, S. T. Is vitamin D deficiency to blame for the asthma epidemic? J. Allergy Clin. Immunol. 120 , 1031–1035 (2007).

Weiss, S. T. Bacterial components plus vitamin D: the ultimate solution to the asthma (autoimmune disease) epidemic? J. Allergy Clin. Immunol. 127 , 1128–1130 (2011).

Sundar, I. K. & Rahman, I. Vitamin D and susceptibility of chronic lung diseases: role of epigenetics. Front. Pharmacol. 2 , 50 (2011).

Shaheen, S. O. & Adcock, I. M. The developmental origins of asthma: does epigenetics hold the key? Am. J. Respir. Crit. Care Med. 180 , 690–691 (2009).

Huang, Y. J. & Boushey, H. A. The microbiome in asthma. J. Allergy Clin. Immunol. 135 , 25–30 (2015).

Holt, P. G., Strickland, D. H., Hales, B. J. & Sly, P. D. Defective respiratory tract immune surveillance in asthma: a primary causal factor in disease onset and progression. Chest 145 , 370–378 (2014).

Holt, P. G. Prevention — what is the most promising approach? Pediatr. Allergy Immunol. 25 , 12–14 (2014). This editorial summarizes a substantial body of evidence gained from longitudinal studies of birth cohorts that suggest that a balanced immune response to bacteria resident on airway mucosal surfaces might limit the risk of asthma.

Hales, B. J. et al . Antibacterial antibody responses associated with the development of asthma in house dust mite-sensitised and non-sensitised children. Thorax 67 , 321–327 (2012).

Hales, B. J. et al . Differences in the antibody response to a mucosal bacterial antigen between allergic and non-allergic subjects. Thorax 63 , 221–227 (2008).

Holt, P. G. & Sly, P. D. Viral infections and atopy in asthma pathogenesis: new rationales for asthma prevention and treatment. Nat. Med. 18 , 726–735 (2012).

Hollams, E. M. et al . Th2-associated immunity to bacteria in teenagers and susceptibility to asthma. Eur. Respir. J. 36 , 509–516 (2010).

Saglani, S. et al . Early detection of airway wall remodeling and eosinophilic inflammation in preschool wheezers. Am. J. Respir. Crit. Care Med. 176 , 858–864 (2007).

Malmström, K. et al . Lung function, airway remodelling and inflammation in symptomatic infants: outcome at 3 years. Thorax 66 , 157–162 (2011).

Sonnappa, S., Bastardo, C. M., Saglani, S., Bush, A. & Aurora, P. Relationship between past airway pathology and current lung function in preschool wheezers. Eur. Respir. J. 38 , 1431–1436 (2011).

O'Reilly, R. et al . Increased airway smooth muscle in preschool wheezers who have asthma at school age. J. Allergy Clin. Immunol. 131 , 1024–1032.e16 (2013). This is one of only a few studies on airway tissue from preschool-aged children with asthma that shows increased airway smooth muscle in these children. The results led the authors to the conclusion that a focus on early changes in smooth muscle might be important in understanding the subsequent development of childhood asthma.

Regamey, N. et al . Increased airway smooth muscle mass in children with asthma, cystic fibrosis, and non-cystic fibrosis bronchiectasis. Am. J. Respir. Crit. Care Med. 177 , 837–843 (2008).

An, S. S. & Fredberg, J. J. Biophysical basis for airway hyperresponsiveness. Can. J. Physiol. Pharmacol. 85 , 700–714 (2007).

Grainge, C. L. et al . Effect of bronchoconstriction on airway remodeling in asthma. N. Engl. J. Med. 364 , 2006–2015 (2011). This landmark study provides proof of principle for the concept that the mechanical stress on the airways imposed by bronchoconstriction might contribute to airway remodelling.

Grainge, C. et al . Resistin-like molecule-β is induced following bronchoconstriction of asthmatic airways. Respirology 17 , 1094–1100 (2012).

Chu, E. K. et al . Induction of the plasminogen activator system by mechanical stimulation of human bronchial epithelial cells. Am. J. Respir. Cell. Mol. Biol. 35 , 628–638 (2006).

Park, J.-A., Drazen, J. M. & Tschumperlin, D. J. The chitinase-like protein YKL-40 is secreted by airway epithelial cells at base line and in response to compressive mechanical stress. J. Biol. Chem. 285 , 29817–29825 (2010).

Holgate, S. T. The sentinel role of the airway epithelium in asthma pathogenesis. Immunol. Rev. 242 , 205–219 (2011).

Bedke, N. et al . Transforming growth factor-beta promotes rhinovirus replication in bronchial epithelial cells by suppressing the innate immune response. PLoS ONE 7 , e44580 (2012).

Djukanovic, R. et al . The effect of inhaled IFN-β on worsening of asthma symptoms caused by viral infections. A randomized trial. Am. J. Respir. Crit. Care Med. 190 , 145–154 (2014). Following the discovery that in moderate-to-severe asthma there is a deficiency in epithelial interferon production upon infection with common respiratory viruses, this randomized controlled trial showed that, in such individuals, inhaled IFNβ at the start of an upper respiratory tract infection can protect against exacerbation of asthma.

Vink, N. M., Postma, D. S., Schouten, J. P., Rosmalen, J. G. & Boezen, H. M. Gender differences in asthma development and remission during transition through puberty: the TRacking Adolescents' Individual Lives Survey (TRAILS) study. J. Allergy Clin. Immunol. 126 , 498–504.e6 (2010).

Van den Toorn, L. M. et al . Airway inflammation is present during clinical remission of atopic asthma. Am. J. Respir. Crit. Care Med. 164 , 2107–2113 (2001).

Wood, R. A., Laheri, A. N. & Eggleston, P. A. The aerodynamic characteristics of cat allergen. Clin. Exp. Allergy 23 , 733–739 (1993).

Vonk, J. M. et al . Childhood factors associated with asthma remission after 30 year follow up. Thorax 59 , 925–929 (2004).

Broekema, M. et al . Airway eosinophilia in remission and progression of asthma: accumulation with a fast decline of FEV1 . Respir. Med. 104 , 1254–1262 (2010).

Broekema, M. et al . Persisting remodeling and less airway wall eosinophil activation in complete remission of asthma. Am. J. Respir. Crit. Care Med. 183 , 310–316 (2011).

Volbeda, F. et al . Can AMP induce sputum eosinophils, even in subjects with complete asthma remission? Respir. Res. 11 , 106 (2010).

Shaaban, R. et al . Rhinitis and onset of asthma: a longitudinal population-based study. Lancet 372 , 1049–1057 (2008).

Platts-Mills, T., Vaughan, J., Squillace, S., Woodfolk, J. & Sporik, R. Sensitisation, asthma, and a modified Th2 response in children exposed to cat allergen: a population-based cross-sectional study. Lancet 357 , 752–756 (2001).

Torrent, M. et al . Early-life allergen exposure and atopy, asthma, and wheeze up to 6 years of age. Am. J. Respir. Crit. Care Med. 176 , 446–453 (2007).

Scott, M. et al . Multifaceted allergen avoidance during infancy reduces asthma during childhood with the effect persisting until age 18 years. Thorax 67 , 1046–1051 (2012). This longitudinal randomized controlled trial on babies considered at high risk of asthma owing to heredity showed that, within the first 12 months of life, intervention with either breastfeeding with the mother on a low-allergen diet or an extensively hydrolysed formula combined with reduced exposure to house dust mite allergen prevented the onset of asthma. The effect occurred in the early years but persisted through to adulthood.

Kovac, K., Dodig, S., Tjesic-Drinkovic, D. & Raos, M. Correlation between asthma severity and serum IgE in asthmatic children sensitized to Dermatophagoides pteronyssinus . Arch. Med. Res. 38 , 99–105 (2007).

Adgate, J. L., Ramachandran, G., Cho, S. J., Ryan, A. D. & Grengs, J. Allergen levels in inner city homes: baseline concentrations and evaluation of intervention effectiveness. J. Expo. Sci. Environ. Epidemiol. 18 , 430–440 (2008).

MacDonald, C., Sternberg, A. & Hunter, P. R. A systematic review and meta-analysis of interventions used to reduce exposure to house dust and their effect on the development and severity of asthma. Environ. Health Perspect. 115 , 1691–1695 (2007). This meta-analysis of 14 studies concluded that there was not sufficient evidence to support implementing hygiene measures to improve outcomes in existing atopic disease. However, interventions from birth in those at high risk of atopy were found to be useful in preventing diagnosed asthma but not parental-reported wheeze.

Van Schayck, O. C., Maas, T., Kaper, J., Knottnerus, A. J. & Sheikh, A. Is there any role for allergen avoidance in the primary prevention of childhood asthma? J. Allergy Clin. Immunol. 119 , 1323–1328 (2007).

Braun-Fahrländer, C. et al . Environmental exposure to endotoxin and its relation to asthma in school-age children. N. Engl. J. Med. 347 , 869–877 (2002).

Ege, M. J. et al . Exposure to environmental microorganisms and childhood asthma. N. Engl. J. Med. 364 , 701–709 (2011).

Heederik, D. & von Mutius, E. Does diversity of environmental microbial exposure matter for the occurrence of allergy and asthma? J. Allergy Clin. Immunol. 130 , 44–50 (2012).

Renz, H., Brandtzaeg, P. & Hornef, M. The impact of perinatal immune development on mucosal homeostasis and chronic inflammation. Nat. Rev. Immunol. 12 , 9–23 (2011).

Carroll, K. N. et al . Season of infant bronchiolitis and estimates of subsequent risk and burden of early childhood asthma. J. Allergy Clin. Immunol. 123 , 964–966 (2009).

Jackson, D. J. & Lemanske, R. F. The role of respiratory virus infections in childhood asthma inception. Immunol. Allergy Clin. North Am. 30 , 513–522 (2010).

Durrani, S. R. et al . Innate immune responses to rhinovirus are reduced by the high-affinity IgE receptor in allergic asthmatic children. J. Allergy Clin. Immunol. 130 , 489–495 (2012).

Mortensen, L. J., Kreiner-Møller, E., Hakonarson, H., Bønnelykke, K. & Bisgaard, H. The PCDH1 gene and asthma in early childhood. Eur. Respir. J. 43 , 792–800 (2014).

Bønnelykke, K. et al . A genome-wide association study identifies CDHR3 as a susceptibility locus for early childhood asthma with severe exacerbations. Nat. Genet. 46 , 51–55 (2014).

Klaassen, E. M. et al . An ADAM33 polymorphism associates with progression of preschool wheeze into childhood asthma: a prospective case–control study with replication in a birth cohort study. PLoS ONE 10 , e0119349 (2015).

Huang, S.-K., Zhang, Q., Qiu, Z. & Chung, K. F. Mechanistic impact of outdoor air pollution on asthma and allergic diseases. J. Thorac. Dis. 7 , 23–33 (2015).

PubMed   PubMed Central   Google Scholar  

Guarnieri, M. & Balmes, J. R. Outdoor air pollution and asthma. Lancet 383 , 1581–1592 (2014). This recent review confirms the link between outdoor air pollution and exacerbations of pre-existing asthma. It also suggests that air pollution contributes to new-onset asthma.

Chan, J. K. et al . Combustion-derived flame generated ultrafine soot generates reactive oxygen species and activates Nrf2 antioxidants differently in neonatal and adult rat lungs. Part. Fibre Toxicol. 10 , 34 (2013).

Wang, P., Thevenot, P., Saravia, J., Ahlert, T. & Cormier, S. A. Radical-containing particles activate dendritic cells and enhance Th17 inflammation in a mouse model of asthma. Am. J. Respir. Cell. Mol. Biol. 45 , 977–983 (2011).

Nadeau, K. et al . Ambient air pollution impairs regulatory T-cell function in asthma. J. Allergy Clin. Immunol. 126 , 845–852.e10 (2010).

Gowers, A. M. et al . Does outdoor air pollution induce new cases of asthma? Biological plausibility and evidence; a review. Respirology 17 , 887–898 (2012).

Goodman, J. E., Chandalia, J. K., Thakali, S. & Seeley, M. Meta-analysis of nitrogen dioxide exposure and airway hyper-responsiveness in asthmatics. Crit. Rev. Toxicol. 39 , 719–742 (2009).

Gauderman, W. J. et al . Association of improved air quality with lung development in children. N. Engl. J. Med. 372 , 905–913 (2015). Levels of air pollution have been trending progressively downward over the past several decades in southern California, as a result of the implementation of air quality-control policies. Here, the authors show that long-term reductions in air pollution were associated with improvements in lung growth and respiratory health among children.

Yang, I. A., Fong, K. M., Zimmerman, P. V., Holgate, S. T. & Holloway, J. W. Genetic susceptibility to the respiratory effects of air pollution. Postgrad. Med. J. 85 , 428–436 (2009).

Esposito, S. et al . Possible molecular mechanisms linking air pollution and asthma in children. BMC Pulm. Med. 14 , 31 (2014).

Vork, K. L., Broadwin, R. L. & Blaisdell, R. J. Developing asthma in childhood from exposure to secondhand tobacco smoke: insights from a meta-regression. Environ. Health Perspect. 115 , 1394–1400 (2007).

Burke, H. et al . Prenatal and passive smoke exposure and incidence of asthma and wheeze: systematic review and meta-analysis. Pediatrics 129 , 735–744 (2012).

Feleszko, W. et al . Parental tobacco smoking is associated with augmented IL-13 secretion in children with allergic asthma. J. Allergy Clin. Immunol. 117 , 97–102 (2006).

Li, Y.-F., Langholz, B., Salam, M. T. & Gilliland, F. D. Maternal and grandmaternal smoking patterns are associated with early childhood asthma. Chest 127 , 1232–1241 (2005). In this key study on generational influences on asthma, a case–control study nested within the Children's Health Study in southern California comprising 338 children with asthma that had been diagnosed in the first 5 years of life, and 570 control individuals, was conducted. The study showed that maternal and grandmaternal smoking during pregnancy might increase the risk of childhood asthma, possibly through epigenetic mechanisms.

Mebrahtu, T. F., Feltbower, R. G., Greenwood, D. C. & Parslow, R. C. Childhood body mass index and wheezing disorders: a systematic review and meta-analysis. Pediatr. Allergy Immunol. 26 , 62–72 (2015).

Forno, E., Young, O. M., Kumar, R., Simhan, H. & Celedon, J. C. Maternal obesity in pregnancy, gestational weight gain, and risk of childhood asthma. Pediatrics 134 , e535–e546 (2014).

Torday, J. S. et al . Leptin mediates the parathyroid hormone-related protein paracrine stimulation of fetal lung maturation. Am. J. Physiol. Lung Cell. Mol. Physiol. 282 , L405–L410 (2002).

Shore, S. A. et al . Effect of leptin on allergic airway responses in mice. J. Allergy Clin. Immunol. 115 , 103–109 (2005).

McLachlan, C. R. et al . Adiposity, asthma, and airway inflammation. J. Allergy Clin. Immunol. 119 , 634–639 (2007).

Kattan, M. et al . Asthma control, adiposity, and adipokines among inner-city adolescents. J. Allergy Clin. Immunol. 125 , 584–592 (2010).

Aaron, S. D. et al . Overdiagnosis of asthma in obese and nonobese adults. CMAJ 179 , 1121–1131 (2008).

[No authors listed.] Statement on standards for the diagnosis and care of patients with chronic obstructive pulmonary disease (COPD) and asthma. Am. Rev. Respir. Dis. 136 , 225–244 (1987).

National Institute for Health and Care Excellence. Asthma: diagnosis and monitoring of asthma in adults, children and young people. NICE [online] , (2015).

Pavord, I. D., Bush, A. & Holgate, S. Asthma diagnosis: addressing the challenges. Lancet Respir. Med. 3 , 339–341 (2015). This editorial highlights the lack of uptake of diagnostic tests to identify inflammation of the airways in patients with asthma in both diagnosis and long-term management, which leads to suboptimal disease control.

Wenzel, S. E. Asthma phenotypes: the evolution from clinical to molecular approaches. Nat. Med. 18 , 716–725 (2012).

Corren, J. et al . Lebrikizumab treatment in adults with asthma. N. Engl. J. Med. 365 , 1088–1098 (2011).

Woodruff, P. G. et al . T-helper type 2-driven inflammation defines major subphenotypes of asthma. Am. J. Respir. Crit. Care Med. 180 , 388–395 (2009).

Green, R. H. et al . Asthma exacerbations and sputum eosinophil counts: a randomised controlled trial. Lancet 360 , 1715–1721 (2002). This landmark study demonstrated that control of asthma using sputum eosinophil numbers to titre ICS use as an adjunct to symptoms and lung function produced better disease control and less overall corticosteroid use. On the basis of these findings, the authors concluded that an asthma treatment strategy directed at normalization of the induced sputum eosinophil count reduces asthma exacerbations and admissions without the need for additional anti-inflammatory treatment.

Jayaram, L. et al . Determining asthma treatment by monitoring sputum cell counts: effect on exacerbations. Eur. Respir. J. 27 , 483–494 (2006).

Bel, E. H. et al . Oral glucocorticoid-sparing effect of mepolizumab in eosinophilic asthma. N. Engl. J. Med. 371 , 1189–1197 (2014).

Pavord, I. D. et al . Mepolizumab for severe eosinophilic asthma (DREAM): a multicentre, double-blind, placebo-controlled trial. Lancet 380 , 651–659 (2012).

Guo, F. H. et al . Interferon γ and interleukin 4 stimulate prolonged expression of inducible nitric oxide synthase in human airway epithelium through synthesis of soluble mediators. J. Clin. Invest. 100 , 829–838 (1997).

Modena, B. D. et al . Gene expression in relation to exhaled nitric oxide identifies novel asthma phenotypes with unique biomolecular pathways. Am. J. Respir. Crit. Care Med. 190 , 1363–1372 (2014).

Smith, A. D. et al . Diagnosing asthma: comparisons between exhaled nitric oxide measurements and conventional tests. Am. J. Respir. Crit. Care Med. 169 , 473–478 (2004).

Wenzel, S. et al . Dupilumab in persistent asthma with elevated eosinophil levels. N. Engl. J. Med. 368 , 2455–2466 (2013). In this paper, which describes one of several biologic interventions in severe asthma, the authors investigated the efficacy and safety of dupilumab, a fully human monoclonal antibody raised against the α-subunit of the IL-4 receptor, in patients with persistent, moderate-to-severe asthma and increased eosinophil counts. Patients received the study drug or placebo for 12 weeks or until a protocol-defined asthma exacerbation occurred. The results showed that dupilumab therapy, as compared with placebo, was associated with fewer asthma exacerbations when LABAs and ICSs were withdrawn accompanied by improved lung function and reduced levels of TH2-associated inflammatory markers.

Hanania, N. A. et al . Exploring the effects of omalizumab in allergic asthma: an analysis of biomarkers in the EXTRA study. Am. J. Respir. Crit. Care Med. 187 , 804–811 (2013).

Takayama, G. et al . Periostin: a novel component of subepithelial fibrosis of bronchial asthma downstream of IL-4 and IL-13 signals. J. Allergy Clin. Immunol. 118 , 98–104 (2006).

Sidhu, S. S. et al . Roles of epithelial cell-derived periostin in TGF-β activation, collagen production, and collagen gel elasticity in asthma. Proc. Natl Acad. Sci. USA 107 , 14170–14175 (2010).

Conway, S. J. et al . The role of periostin in tissue remodeling across health and disease. Cell. Mol. Life Sci. 71 , 1279–1288 (2014).

Jia, G. et al . Periostin is a systemic biomarker of eosinophilic airway inflammation in asthmatic patients. J. Allergy Clin. Immunol. 130 , 647–654.e10 (2012).

Izuhara, K. et al . Periostin in allergic inflammation. Allergol. Int. 63 , 143–151 (2014). Periostin is an extracellular matrix protein involved in remodelling during tissue and/or organ development or repair that is under the control of IL-4 and IL-13 in the airways. In this review, not only do the authors point to the usefulness of serum periostin as a TH2 biomarker but they also suggest that, at least in skin, periostin is crucial for amplification and persistence of allergic inflammation by communicating between fibroblasts and keratinocytes. In addition, they suggest that blocking the interaction between periostin and its receptor, αv integrin, or downregulating periostin expression might be a novel asthma intervention.

Schulman, E. S. & Pohlig, C. Rationale for specific allergen testing of patients with asthma in the clinical pulmonary office setting. Chest 147 , 251–258 (2015).

Ortega, H. G. et al . Mepolizumab treatment in patients with severe eosinophilic asthma. N. Engl. J. Med. 371 , 1198–1207 (2014).

Choy, D. F. et al . TH2 and TH17 inflammatory pathways are reciprocally regulated in asthma. Sci. Transl. Med. 7 , 301ra129 (2015).

Mauri, P. et al . Proteomics of bronchial biopsies: galectin-3 as a predictive biomarker of airway remodelling modulation in omalizumab-treated severe asthma patients. Immunol. Lett. 162 , 2–10 (2014).

Duncan, J. M. & Sears, M. R. Breastfeeding and allergies: time for a change in paradigm? Curr. Opin. Allergy Clin. Immunol. 8 , 398–405 (2008).

Kramer, M. S. Breastfeeding and allergy: the evidence. Ann. Nutr. Metab. 59 , S20–S26 (2011).

Oddy, W. H., Peat, J. K. & de Klerk, N. H. Maternal asthma, infant feeding, and the risk of asthma in childhood. J. Allergy Clin. Immunol. 110 , 65–67 (2002).

Sears, M. R. et al . Long-term relation between breastfeeding and development of atopy and asthma in children and young adults: a longitudinal study. Lancet 360 , 901–907 (2002).

Dogaru, C. M., Nyffenegger, D., Pescatore, A. M., Spycher, B. D. & Kuehni, C. E. Breastfeeding and childhood asthma: systematic review and meta-analysis. Am. J. Epidemiol. 179 , 1153–1167 (2014). Although controversial for many years, this systematic review and meta-analysis of studies published between 1983 and 2012 on breastfeeding and asthma in children from the general population found a positive association between breastfeeding and reduced asthma and/or wheezing, and is supported by the combined evidence of existing studies.

Ramos, G. F. et al . Cost-effectiveness of primary prevention of paediatric asthma: a decision-analytic model. Eur. J. Health Econ. 15 , 869–883 (2014).

Litonjua, A. A. et al . The Vitamin D Antenatal Asthma Reduction Trial (VDAART): rationale, design, and methods of a randomized, controlled trial of vitamin D supplementation in pregnancy for the primary prevention of asthma and allergies in children. Contemp. Clin. Trials 38 , 37–50 (2014).

Yang, H., Xun, P. & He, K. Fish and fish oil intake in relation to risk of asthma: a systematic review and meta-analysis. PLoS ONE 8 , e80048 (2013).

Anandan, C., Nurmatov, U. & Sheikh, A. Omega 3 and 6 oils for primary prevention of allergic disease: systematic review and meta-analysis. Allergy 64 , 840–848 (2009).

Von Mutius, E. & Vercelli, D. Farm living: effects on childhood asthma and allergy. Nat. Rev. Immunol. 10 , 861–868 (2010).

Elazab, N. et al . Probiotic administration in early life, atopy, and asthma: a meta-analysis of clinical trials. Pediatrics 132 , e666–e676 (2013).

West, C. E., Hammarströ m, M.-L. & Hernell, O. Probiotics in primary prevention of allergic disease — follow-up at 8–9 years of age. Allergy 68 , 1015–1020 (2013).

Bengmark, S. Gut microbiota, immune development and function. Pharmacol. Res. 69 , 87–113 (2013).

Nieto, A. et al . Allergy and asthma prevention 2014. Pediatr. Allergy Immunol. 25 , 516–533 (2014).

Heine, R. G. Preventing atopy and allergic disease. Nestle Nutr. Inst. Workshop Ser. 78 , 141–153 (2014).

Tamblyn, R. et al . Evaluating the impact of an integrated computer-based decision support with person-centered analytics for the management of asthma in primary care: a randomized controlled trial. J. Am. Med. Inform. Assoc. 22 , 773–783 (2015).

Griffiths, C. & Levy, M. L. Preventing avoidable asthma deaths. Practitioner 258 , 27–31 (2014).

Toelle, B. G. & Ram, F. S. Written individualised management plans for asthma in children and adults. Cochrane Database Syst. Rev. 2 , CD002171 (2004).

Google Scholar  

Depner, M. et al . Clinical and epidemiologic phenotypes of childhood asthma. Am. J. Respir. Crit. Care Med. 189 , 129–138 (2014).

Pedersen, S. E. et al . Global strategy for the diagnosis and management of asthma in children 5 years and younger. Pediatr. Pulmonol. 46 , 1–17 (2011).

Roberts, N. J. et al . The development and comprehensibility of a pictorial asthma action plan. Patient Educ. Couns. 74 , 12–18 (2009).

National Heart Lung and Blood Advisory Council Asthma Expert Working Group. Draft needs assessment report for potential update of the expert panel report-3 (2007): guidelines for the diagnosis and management of asthma January 2015. NIH [online] , (2015).

British Thoracic Society & Scottish Intercollegiate Guidelines Network. British guideline on the management of asthma. Thorax 69 , S1–S192 (2014).

Papi, A. et al . Beclometasone-formoterol as maintenance and reliever treatment in patients with asthma: a double-blind, randomised controlled trial. Lancet Respir. Med. 1 , 23–31 (2013).

Lipworth, B. J. Emerging role of long acting muscarinic antagonists for asthma. Br. J. Clin. Pharmacol. 77 , 55–62 (2014).

Matera, M. G. & Cazzola, M. Ultra-long-acting beta2-adrenoceptor agonists: an emerging therapeutic option for asthma and COPD? Drugs 67 , 503–515 (2007).

Loymans, R. J. B. et al . Comparative effectiveness of long term drug treatment strategies to prevent asthma exacerbations: network meta-analysis. BMJ 348 , g3009 (2014). In this comprehensive study, 64 trials with 59,622 patient years of follow-up comparing 15 strategies and placebo were evaluated for the prevention of severe exacerbations, combined ICSs and LABAs as maintenance and reliever treatment and combined ICSs alone. It found that LABAs in a fixed daily dose performed equally well and were ranked first for effectiveness. The overall conclusion was that strategies with combined ICSs and long-acting β-adrenergic agonists are most effective and safe in preventing severe exacerbations of asthma.

Abramson, M. J., Puy, R. M. & Weiner, J. M. Injection allergen immunotherapy for asthma. Cochrane Database Syst. Rev. 8 , CD001186 (2010).

Mosbech, H. et al . Standardized quality (SQ) house dust mite sublingual immunotherapy tablet (ALK) reduces inhaled corticosteroid use while maintaining asthma control: a randomized, double-blind, placebo-controlled trial. J. Allergy Clin. Immunol. 134 , 568–575.e7 (2014).

Kelly, W., Massoumi, A. & Lazarus, A. Asthma in pregnancy: physiology, diagnosis, and management. Postgrad. Med. 127 , 349–358 (2015).

Price, D. et al . Reassessing the evidence hierarchy in asthma: evaluating comparative effectiveness. Curr. Allergy Asthma Rep. 11 , 526–538 (2011).

Qamar, N., Pappalardo, A. A., Arora, V. M. & Press, V. G. Patient-centered care and its effect on outcomes in the treatment of asthma. Patient Relat. Outcome Meas. 2 , 81–109 (2011).

Hinks, T. S. C. et al . Innate and adaptive T cells in asthmatic patients: relationship to severity and disease mechanisms. J. Allergy Clin. Immunol. 136 , 323–333 (2015).

Holgate, S. T. Stratified approaches to the treatment of asthma. Br. J. Clin. Pharmacol. 76 , 277–291 (2013).

Campo, P. et al . Phenotypes and endotypes of uncontrolled severe asthma: new treatments. J. Investig. Allergol. Clin. Immunol. 23 , 76–88 (2013).

Krug, N. et al . Allergen-induced asthmatic responses modified by a GATA3-specific DNAzyme. N. Engl. J. Med. 372 , 1987–1995 (2015).

Hambly, N. & Nair, P. Monoclonal antibodies for the treatment of refractory asthma. Curr. Opin. Pulm. Med. 20 , 87–94 (2014).

Lima, J. J. Do genetic polymorphisms alter patient response to inhaled bronchodilators? Expert Opin. Drug Metab. Toxicol. 10 , 1231–1240 (2014).

Zuurhout, M. J. L. et al . Arg16 ADRB2 genotype increases the risk of asthma exacerbation in children with a reported use of long-acting β2-agonists: results of the PACMAN cohort. Pharmacogenomics 14 , 1965–1971 (2013).

Manoharan, A. et al . β2-adrenergic receptor Gly16Arg polymorphism and impaired asthma control in corticosteroid-treated asthmatic adults. Ann. Allergy. Asthma Immunol. 114 , 421–423 (2015).

Ortega, V. E. et al . Effect of rare variants in ADRB2 on risk of severe exacerbations and symptom control during longacting β agonist treatment in a multiethnic asthma population: a genetic study. Lancet Respir. Med. 2 , 204–213 (2014). Although polymorphisms of the β2-adrenergic receptor especially, at amino acids 16 and 27, have been associated with more-active asthma and impaired β-agonist treatment responsiveness, this comprehensive genotypic analysis has shown that the rare adrenoceptor β2 gene (ADRB2) variants Ile164 and -376ins are associated with severe adverse events during LABA therapy. It is suggested that they should be evaluated in large clinical trials, including in the current FDA-mandated safety study.

Meyers, D. A., Bleecker, E. R., Holloway, J. W. & Holgate, S. T. Asthma genetics and personalised medicine. Lancet Respir. Med. 2 , 405–415 (2014).

Miller, R. J. & Murgu, S. D. Interventional pulmonology for asthma and emphysema: bronchial thermoplasty and bronchoscopic lung volume reduction. Semin. Respir. Crit. Care Med. 35 , 655–670 (2014).

Castro, M., Cox, G., Wechsler, M. E. & Niven, R. M. Bronchial thermoplasty: ready for prime time — the evidence is there! Chest 147 , e73–e74 (2015).

Pretolani, M. et al . Reduction of airway smooth muscle mass by bronchial thermoplasty in patients with severe asthma. Am. J. Respir. Crit. Care Med. 190 , 1452–1454 (2014).

Sheshadri, A., McKenzie, M. & Castro, M. Critical review of bronchial thermoplasty: where should it fit into asthma therapy? Curr. Allergy Asthma Rep. 14 , 470 (2014).

Iyer, V. N. & Lim, K. G. Bronchial thermoplasty: reappraising the evidence (or lack thereof). Chest 146 , 17–21 (2014).

Ducharme, F. M., Tse, S. M. & Chauhan, B. Diagnosis, management, and prognosis of preschool wheeze. Lancet 383 , 1593–1604 (2014).

Guilbert, T. W. et al . Long-term inhaled corticosteroids in preschool children at high risk for asthma. N. Engl. J. Med. 354 , 1985–1997 (2006).

Busse, W. W. et al . Randomized trial of omalizumab (anti-IgE) for asthma in inner-city children. N. Engl. J. Med. 364 , 1005–1015 (2011).

Wasfi, Y. S. et al . Onset and duration of attenuation of exercise-induced bronchoconstriction in children by single-dose of montelukast. Allergy Asthma Proc. 32 , 453–459 (2011).

Schultz, A. & Brand, P. L. Phenotype-directed treatment of pre-school-aged children with recurrent wheeze. J. Paediatr. Child Health 48 , E73–E78 (2012).

Lemanske, R. F. et al . Step-up therapy for children with uncontrolled asthma receiving inhaled corticosteroids. N. Engl. J. Med. 362 , 975–985 (2010). The Best Add-on Therapy Giving Effective Responses (BADGER) study demonstrated that there is not a single ‘best’ add-on therapy for children whose asthma is not controlled by first-line therapy.

Martinez, F. D. The connection between early life wheezing and subsequent asthma: the viral march. Allergol. Immunopathol. (Madr.) 37 , 249–251 (2009).

Martinez, F. D. & Vercelli, D. Asthma. Lancet 382 , 1360–1372 (2013).

Van Asperen, P. P., Mellis, C. M., Sly, P. D. & Robertson, C. F. Evidence-based asthma management in children — what's new? Med. J. Aust. 194 , 383–384 (2011). This study reviewed the evidence-base for asthma treatment in children and concluded that early-life intervention is crucial to prevent asthma from evolving over the lifecourse.

Zeiger, R. S. et al . Daily or intermittent budesonide in preschool children with recurrent wheezing. N. Engl. J. Med. 365 , 1990–2001 (2011).

Beigelman, A. et al . Do oral corticosteroids reduce the severity of acute lower respiratory tract illnesses in preschool children with recurrent wheezing? J. Allergy Clin. Immunol. 131 , 1518–1525 (2013).

Pelkonen, A. S. et al . The effect of montelukast on respiratory symptoms and lung function in wheezy infants. Eur. Respir. J. 41 , 664–670 (2013).

Wechsler, M. E. Getting control of uncontrolled asthma. Am. J. Med. 127 , 1049–1059 (2014).

Worth, A. et al . Patient-reported outcome measures for asthma: a systematic review. NPJ Prim. Care Respir. Med. 24 , 14020 (2014). This systematic review of the literature identified the AQLQ and mini-AQLQ for adults and PAQLQ as PROMs that were found to be sufficiently well validated to offer promise for use in clinical settings. Rhinasthma was also considered promising in simultaneously assessing the impact of asthma and rhinitis.

Juniper, E. Measurement of health-related quality of life and asthma control. Quoltech [online] ,

Chen, H. et al . Asthma control, severity, and quality of life: quantifying the effect of uncontrolled disease. J. Allergy Clin. Immunol. 120 , 396–402 (2007).

Lurie, A., Marsala, C., Hartley, S., Bouchon-Meunier, B. & Dusser, D. Patients' perception of asthma severity. Respir. Med. 101 , 2145–2152 (2007).

Haselkorn, T. et al . Effect of weight change on asthma-related health outcomes in patients with severe or difficult-to-treat asthma. Respir. Med. 103 , 274–283 (2009).

Hanania, N. A. et al . Omalizumab in severe allergic asthma inadequately controlled with standard therapy: a randomized trial. Ann. Intern. Med. 154 , 573–582 (2011).

Castro, M. et al . Effectiveness and safety of bronchial thermoplasty in the treatment of severe asthma: a multicenter, randomized, double-blind, sham-controlled clinical trial. Am. J. Respir. Crit. Care Med. 181 , 116–124 (2010).

Earnshaw, V. A. & Quinn, D. M. The impact of stigma in healthcare on people living with chronic illnesses. J. Health Psychol. 17 , 157–168 (2012).

Comer, B. S., Ba, M., Singer, C. A. & Gerthoffer, W. T. Epigenetic targets for novel therapies of lung diseases. Pharmacol. Ther. 147 , 91–110 (2015).

Boutin, H. & Boulet, L.-P. L'asthme au Quotidien (Press Universite Laval, 2005).

Bousquet, J., Bousquet, P. J., Godard, P. & Dyres, J.-P. The public health implications of asthma. Bull. World Health Organ. 93 , 548–554 (2005).

Holgate, S. T. et al . A new look at the pathogenesis of asthma. Clin. Sci. 118 , 439–450 (2009).

Bateman, E. D. et al . Overall asthma control: the relationship between current control and future risk. J. Allergy Clin. Immunol. 125 , 600–608.e6 (2010).

Download references

Author information

Authors and affiliations.

Clinical and Experimental Sciences, Mail Point 810, Level F, Sir Henry Wellcome Building

Stephen T. Holgate

Southampton General Hospital, Southampton, SO16 6YD, UK

Subsection Chief of Allergy, Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Asthma Institute at UPMC/UPSOM, Pittsburgh, Pennsylvania, USA

Sally Wenzel

Department of Pulmonology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands

Dirkje S. Postma

Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA

Scott T. Weiss

Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Philipps University Marburg, University Hospital Giessen and Marburg GmbH, Campus Marburg, Marburg, Germany

Harald Renz

Queensland Children's Medical Research Institute and Centre for Child Health Research, University of Queensland, Brisbane, Australia

Peter D. Sly

You can also search for this author in PubMed   Google Scholar

Contributions

Introduction (S.T.H.); Epidemiology (S.T.W.); Mechanisms/pathophysiology (P.D.S. and S.T.H.); Diagnosis, screening and prevention (D.S.P., H.R., S.T.H. and S.W.); Management (D.S.P., P.D.S. and S.T.H.); Quality of life (D.S.P.); Outlook (D.S.P., H.R., P.D.S., S.T.H., S.T.W. and S.W.); Overview of Primer (S.T.H.).

Corresponding author

Correspondence to Stephen T. Holgate .

Ethics declarations

Competing interests.

S.T.H. is a non-executive director and a consultant for Synairgen, and a consultant for AstraZeneca and Novartis. He is in receipt of a UK Medical Research Council (MRC) programme grant and receives salary support as an MRC clinical professor. S.W. has served as a consultant for Aerocrine, GlaxoSmithKline and AstraZeneca. She has received research funding (paid to her institution) from GlaxoSmithKline, AstraZeneca, Genentech and Sanofi-Aventis. She has also received research support from the NIH National Heart, Lung and Blood Institute (NHLBI) and the National Institute of Allergy and Infectious Diseases (NIAID), which provides her salary support. D.S.P. has received an unrestricted educational grant for research (paid to the University of Groningen) from AstraZeneca. Her travel to the European Respiratory Society (ERS) and/or the American Thoracic Society (ATS) meetings has been partially funded by AstraZeneca, Chiesi, GlaxoSmithKline and Takeda. Fees for consultancies were given to the University of Groningen by AstraZeneca, Boehringer Ingelheim, Chiesi, GlaxoSmithKline, Takeda and Teva. Her travel and lectures in China have been funded by Chiesi. S.T.W. is funded by the NIH NHLBI and is a consultant for the TENOR study for Novartis. H.R. has received research support from the German Research Foundation (DFG), the Federal Ministry of Education and Research (BMBF), the European Union, Land Hessen, the German Academic Exchange Service (DAAD), ALK, Stiftung Pathobiochemie, Ernst-Wendt-Stiftung, Mead Johnson Nutritional and Beckman Coulter. He has also received speakers honoraria from Allergopharma, Novartis, Thermo Fisher Scientific, Danone, Mead Johnson Nutritional and Bencard Allergie. H.R. also serves as a consultant for Bencard Allergie and Sterna Biologicals (for which he is also a cofounder). P.D.S. declares no competing interests.

PowerPoint slides

Powerpoint slide for fig. 1, powerpoint slide for fig. 2, powerpoint slide for fig. 3, powerpoint slide for fig. 4, powerpoint slide for fig. 5, powerpoint slide for fig. 6, powerpoint slide for fig. 7, powerpoint slide for fig. 8, powerpoint slide for fig. 9, powerpoint slide for fig. 10, powerpoint slide for fig. 11, powerpoint slide for fig. 12, rights and permissions.

Reprints and permissions

About this article

Cite this article.

Holgate, S., Wenzel, S., Postma, D. et al. Asthma. Nat Rev Dis Primers 1 , 15025 (2015). https://doi.org/10.1038/nrdp.2015.25

Download citation

Published : 10 September 2015

DOI : https://doi.org/10.1038/nrdp.2015.25

Share this article

Anyone you share the following link with will be able to read this content:

Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative

This article is cited by

Racial differences in length of stay and readmission for asthma in the all of us research program.

  • Esteban Correa-Agudelo
  • Yadu Gautam
  • Tesfaye B. Mersha

Journal of Translational Medicine (2024)

High apoptotic endothelial microparticle levels measured in asthma with elevated IgE and eosinophils

  • Yael Strulovici-Barel
  • Robert J. Kaner
  • Ronald G. Crystal

Respiratory Research (2023)

Association of GAB1 gene with asthma susceptibility and the efficacy of inhaled corticosteroids in children

  • Yuxuan Zhang

BMC Pulmonary Medicine (2023)

Diagnostic value of IL-6 for patients with asthma: a meta-analysis

  • Shougang Kuai

Allergy, Asthma & Clinical Immunology (2023)

Activating transcription factor-2 supports the antioxidant capacity and ability of human mesenchymal stem cells to prevent asthmatic airway inflammation

  • HongDuck Yun
  • Dong-Myung Shin

Experimental & Molecular Medicine (2023)

Quick links

  • Explore articles by subject
  • Guide to authors
  • Editorial policies

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

research articles on asthma pdf

IMAGES

  1. (PDF) Diagnosis of asthma in adults

    research articles on asthma pdf

  2. Asthma research articles available free through June 30, 2019

    research articles on asthma pdf

  3. Are adults with asthma less physically active? A systematic review and

    research articles on asthma pdf

  4. (PDF) Drug utilization study of bronchial asthma in adults at rural

    research articles on asthma pdf

  5. (PDF) Bronchial Asthma: Diagnosis and Long-Term Treatment in Adults

    research articles on asthma pdf

  6. Case Study of Asthma

    research articles on asthma pdf

VIDEO

  1. Asthma 2024 By Dr Dheyaa Jabbar

  2. Asthma Causes, Symptoms || Gyanear

  3. Study finds people with persistent asthma linked to artery blockages; Low, moderate stress has b

  4. Asthma and COVID-19

  5. MANAGEMENT OF PEDIATRIC ASTHMA

  6. #AskAboutAsthma: using medicines to safely treat asthma attacks in children

COMMENTS

  1. Treatment strategies for asthma: reshaping the concept of asthma

    Asthma treatment is based on a stepwise and control-based approach that involves an iterative cycle of assessment, adjustment of the treatment and review of the response aimed to minimize symptom burden and risk of exacerbations. Anti-inflammatory treatment is the mainstay of asthma management.

  2. What is Asthma?

    We know a lot more about asthma today than we did just a decade ago, and we have a much better understanding of how to treat it. BASIC FACTS ABOUT ASTHMA. • Asthma is a lung disease. It’s a physical and medical problem that needs treatment. It’s not something that you imagined or made up.

  3. (PDF) An Overview of Asthma and its treatment

    Asthma is a disorder characterized by chronic airway inflammation, airway hypersensitivity to a variety of stimuli, and airway obstruction. It is at least partially reversible, either...

  4. (PDF) Asthma: pathophysiology, diagnosis and management -

    This article provides an overview of asthma in adults, includin pathophysiology, risk factors and triggers. Assessment, diagn pharmacological therapies are considered.

  5. Update in Adult Asthma 2020

    PDF. In this 2020 Update of publications in American Thoracic Society (ATS) journals, we review breakthroughs in understanding of pathophysiology, treatment, and risk factors for asthma.

  6. Primary prevention of asthma: from risk and protective factors to

    Download PDF. Summary. Asthma is a complex disease that often starts in childhood. Genomic and environmental factors as well as aberrant immune maturation early in life can contribute to the onset of disease, with great disparity over time and geographical regions.

  7. Biologic Therapies for Severe Asthma

    Severe asthma is a heterogeneous syndrome encompassing several clinical pheno-types that differ according to age at the onset of asthma (onset in childhood vs. onset in adulthood), presence or...

  8. Asthma

    The 2015 Global Strategy for Asthma Management and Prevention by the Global Initiative for Asthma (GINA) defined asthma as a heterogeneous disease characterized by chronic airway inflammation...