Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Paediatric Respiratory Reviews 36 (2020) 118–127

Contents lists available at ScienceDirect

Paediatric Respiratory Reviews

Review

Understanding the immunology of asthma: Pathophysiology,


biomarkers, and treatments for asthma endotypes
Melissa D. Gans ⇑, Tatyana Gavrilova 1
Division of Allergy and Immunology, Montefiore Medical Center, 1525 Blondell Ave, Bronx, NY 10461, USA

Educational aims

The reader will come to appreciate that:

 Patients diagnosed with asthma will soon be classified by asthma endotype using identified risk factors and biomarkers.
 An understanding the pathophysiology of the various asthma endotypes can direct targeted therapy.
 There are many emerging biologic treatments for asthma that can greatly reduce morbidity and mortality in severe asthmatics.

a r t i c l e i n f o a b s t r a c t

Keywords: Asthma is a common disease in paediatrics and adults with a significant morbidity, mortality, and finan-
Asthma cial burden worldwide. Asthma is now recognized as a heterogeneous disease and emerging clinical and
Immunology laboratory research has elucidated understanding of asthma’s underlying immunology. The future of
Pathophysiology asthma is classifying asthma by endotype through connecting discernible characteristics with immuno-
Phenotypes
logical mechanisms. This comprehensive review of the immunology of asthma details the currently
Review
known pathophysiology and clinical practice biomarkers in addition to forefront biologic and targeted
Biomarkers
therapies for all of the asthma endotypes. By understanding the immunology of asthma, practitioners will
be able to diagnose patients by asthma endotype and provide personalized, biomarker-driven treatments
to effectively control patients’ asthma.
Ó 2019 Elsevier Ltd. All rights reserved.

INTRODUCTION There is a critical need to treat asthma more effectively. Asthma


was previously understood to be a single diagnosis with standard-
There are 24 million people in the United States suffering from ized treatments for all patients; however, asthma is now accepted
asthma [1]. Asthma prevalence is increasing, affecting 3.1% of the to be a heterogeneous, multifactorial disorder with a variety of
United States population in 1980 and 8.3% of the population in genetic and environmental factors where targeted therapies result
2016 [1]. Asthma is one of the leading chronic illnesses of child- in improved asthma control.
hood and disproportionally affects African Americans and those The Global Initiative for Asthma (GINA) has recognized demo-
living below the poverty level [1]. Asthma exacerbations cause graphic, clinical, and pathophysiological characteristics that are
missed days from school and work, hospitalizations, emergency clustered into distinct asthma phenotypes: allergic asthma, non-
department visits, and more than 3000 deaths annually [1]. allergic asthma, late-onset asthma, asthma with fixed airflow lim-
Asthma also entails a great financial burden, with a cost burden itation, and asthma with obesity [2]. However, utility of pheno-
greatest for those with poorly controlled asthma and in low- types is limited as these phenotypes are grouped by observable
income countries. characteristics with no connection to the underlying disease
process.
The PRACTALL consensus report by experts from the European
⇑ Corresponding author. Fax: +1 718 405 8532. Academy of Allergy and Clinical Immunology and the American
E-mail addresses: mstone@montefiore.org (M.D. Gans), tgavrilo@montefiore.org Academy of Allergy, Asthma & Immunology in 2011 proposed that
(T. Gavrilova). in addition to phenotypes, asthma should be grouped by endotypes
1
Fax: +1 718 405 8532.

https://doi.org/10.1016/j.prrv.2019.08.002
1526-0542/Ó 2019 Elsevier Ltd. All rights reserved.
M.D. Gans, T. Gavrilova / Paediatric Respiratory Reviews 36 (2020) 118–127 119

Table 1
Risk factors, pathophysiology, and biomarkers used to diagnose the following asthma endotypes: allergic asthma, asthma-predictive indices (API)-positive preschool wheezer,
infection-induced asthma, viral-exacerbated asthma, allergic bronchopulmonary mycosis (ABPM), aspirin-sensitive asthma, airflow obstruction caused by obesity, premenstrual
asthma, neutrophilic asthma, elite-athlete asthma, cross-country skiers’ asthma, steroid-insensitive eosinophilic asthma, late-onset hypereosinophilic asthma, and severe steroid-
dependent asthma. Early-onset eosinophilic asthma and preschooler with episodic viral wheeze are pediatric subendotypes of allergic asthma and infection-induced asthma,
respectively, while exercise-induced asthma is a subendotype of elite-athlete asthma.

Asthma endotype Risk factors Pathophysiology Biomarkers


Allergic asthma Environmental allergen sensitivity Th2 response Eosinophils, IgE, periostin, FeNO,
Early-onset eosinophilic asthma Environmental allergen sensitivity; onset LTE4
in childhood; no systemic eosinophilic
manifestations
API-positive preschool wheezer Environmental allergen sensitivity; Eosinophils, IgE, LTE4
positive API score based on history of
eczema, allergic rhinitis, wheezing
episodes per year, eosinophilia, wheezing
apart from colds, and family history of
asthma; many different
bronchoconstriction triggers; onset in
childhood
Infection-induced asthma Tobacco smoke exposure, prematurity, Th1 and Th2 response; viruses Neutrophils, LTE4
Preschooler with episodic viral wheeze prior intubation, and recurrent viral also cause direct damage to
infections; non-allergic; typically only airway epithelium which allow
have bronchoconstriction during viral allergen penetration into
illness subepithelial tissues
Viral-exacerbated asthma Viral infection; can have Eosinophils, neutrophils, IgE,
bronchoconstriction apart from viral periostin, FeNO, LTE4
illnesses
ABPM Fungal airway colonization typically by Th2 response; fungi release toxic Eosinophils, IgE, FeNO
Aspergillus fumigatus or Candida albicans; proteases to pulmonary
fungi hypersensitivity demonstrated by epithelium that can progress to
elevated specific IgE or positive skin test; bronchiectasis and fixed airflow
patients usually also have pulmonary obstruction if not treated
opacities and peripheral eosinophilia
Aspirin-sensitive asthma Aspirin and nonsteroidal anti- Th2 response; overproduction of Eosinophils, periostin, FeNO,
inflammatory drug hypersensitivity leukotrienes LTE4
demonstrated by challenge; patients with
aspirin-exacerbated respiratory disease
also have chronic rhinosinusitis with
recurrent nasal polyps
Airflow obstruction caused by obesity Obesity, gastroesophageal reflux disease, Th2 response; systemic Unknown
and sleep disordered breathing inflammation from
proinflammatory circulating
cytokines, increased oxidative
stress, and increased leptin
Premenstrual asthma Menstruation triggers Th2 response; sex hormone Eosinophils, FeNO
bronchoconstriction in females surge during premenstrual
phase stimulates exaggerated
Th2 response
Neutrophilic asthma Tobacco smoke and diesel exhaust particle Th17 response Neutrophils
exposure
Elite-athlete asthma Elite-level exercise and environmental Th2 response; airway Neutrophils
exposures such as chlorine in swimming dehydration from increased
and ultrafine particles in ice skating; fixed ventilation causes increased
bronchoconstriction airway fluid osmolarity and
Exercise-induced asthma Amateur-level exercise; reversible airway epithelial damage
bronchoconstriction
Cross-country skiers’ asthma Elite-level exercise and cold air exposure; Th2 response; airway Neutrophils
fixed bronchoconstriction dehydration from cold air causes
increased airway fluid
osmolarity
Steroid-insensitive eosinophilic asthma Non-allergic; glucocorticoid receptor Mutations cause increased p38 Eosinophils, FeNO
mutations causing steroid resistance mitogen-activated protein
kinase activity which
phosphorylates the
glucocorticoid receptor and
causes decreased binding of
steroid
Late-onset hypereosinophilic asthma Non-allergic; onset in adulthood; Eosinophils directly release Eosinophils, FeNO
systemic eosinophilic manifestations mediators to damage airway
endothelium
Severe steroid-dependent asthma Multi-factorial; possibly related to asthma Likely a combination of Th1, Th2, Unknown
genetic mutations in ADAM33, PHF11, and Th17 responses
DPP10, GPRA, or SPINK5
120 M.D. Gans, T. Gavrilova / Paediatric Respiratory Reviews 36 (2020) 118–127

(Table 1) [3]. Asthma endotypes overlap with many asthma pheno- DPP10, GPRA, and SPINK5 [5]. ADAM33 is involved with airway
types but are divergent in the underlying biological mechanism. remodeling, independent of inflammation [6]. PHF11 has been
These asthma endotypes describe the underlying immunology. implicated in maintaining epithelial structural integrity [7], while
Characterizing asthmatics by endotype can optimize management GPRA has an unknown role in the airway epithelium [8]. There is
and drive personalized therapy targeting specific immunological also a genetic predisposition for allergic clusters. For instance,
mechanisms of underlying disease [4]. DPP10 is a genetic biomarker for AERD [9], while SPINK5 polymor-
This review details the role of pathophysiology in diagnosis, the phisms are associated with both asthma and atopic dermatitis [10].
utility of biomarkers in clinical practice, and emerging biologic and
traditional treatments for the various asthma endotypes. Under- BIOMARKERS
standing the immunology of asthma will allow physicians to diag-
nose asthma by endotype and have more targeted and effective Traditional asthma biomarkers include eosinophils, neutrophils,
treatment strategies to better control their patients’ asthma. IgE, periostin, fraction of exhaled nitric oxide (FeNO), and leuko-
trienes. While there are many other biomarkers being studied in
PATHOPHYSIOLOGY asthma – such as cytokines, dipeptidyl peptidase-4, and volatile
organic compounds – there is limited data for the utility of using
Allergens, infections, obesity, hormones, tobacco smoke, exer- these biomarkers in clinical practice. Evaluating biomarkers in
cise, cold air, genetic mutations, and systemic eosinophilia are patients with asthma can aid in endotype diagnosis (Table 1); how-
among known factors that induce chronic airway inflammation ever, biomarker-directed management is still limited and currently
leading to airway obstruction and hyperresponsiveness. The only useful in select asthma endotypes.
immunopathophysiology of asthma involves the activation of both
the innate and adaptive immune systems to stimulate chronic air- Eosinophils
way inflammation. Chronic airway inflammation subsequently
causes airway oedema, mucus hypersecretion, mucus plugging, Serum and sputum eosinophils can be used to diagnose eosino-
and airway remodeling. The process of airway remodeling is driven philic asthma and can be elevated in both allergic and non-allergic
by subepithelial fibrosis, thickening of sub-basement membrane, asthma. Though sputum eosinophilia is more accurate, serum
increased airway smooth muscle mass, angiogenesis, and mucous eosinophilia can service as a surrogate marker of sputum eosino-
gland hyperplasia – which result in permanent structural changes. philia [11].
The pathophysiology of how these known factors induce perma- A meta-analysis in 2010 concluded that sputum eosinophil
nent structural changes in the various asthma endotypes is count can be used to guide therapy in asthmatics [12]. In particular,
through a combination of T helper (Th) 1, 2, and 17 responses in serum eosinophilia can be used to monitor biochemical response to
addition to underlying genetic predisposition (Fig. 1). anti-IL-5 therapy. However, serum eosinophilia cannot be used to
monitor response to dupilumab (anti IL4R) therapy as the eosino-
Th1 response phil count artificially increases with dupilumab since IL-4 and IL-
13 promote vascular cell adhesion proteins on eosinophils.
The Th1 response is typically activated in infections, particu- For children with asthma, peripheral eosinophilia is associated
larly viruses. Viruses upregulate interferon-c and interleukin (IL) with more frequent asthma exacerbations [13]. Of all the biomark-
27, which aid in eliminating the pathogen but also are involved ers for asthma, serum eosinophil count has the most meaningful
in airway inflammation. data for asthma endotyping.

Th2 response Neutrophils

Dendritic cells in the airway present inhaled allergens to naïve Whereas eosinophils are typically elevated in the Th2 asthma
T-cells, activating the production of Th2 cells. Th2 cells release Th2 endotypes, the Th17 endotype predominates in neutrophilic
cytokines including: IL-4, IL-5, IL-9, and IL-13. IL-4, IL-9, and IL-13 asthma where there are increased serum and sputum neutrophils.
stimulate B cells to release IgE. IgE then instigates mast cell However, some patients with neutrophilic asthma do not have ele-
degranulation and mediator release (histamine and leukotrienes) vated peripheral neutrophilia, though are still suspected to have
causing bronchoconstriction. These mechanisms are perpetuated neutrophilic asthma in the absence of peripheral eosinophilia.
by cytokines such as IL-25, IL-33, and thymic stromal lymphopoi-
etin (TSLP). IL-25 induces expression of IL-4, IL-5, IL-9, and IL-13, IgE
while IL-33 activates dendritic cells to produce IL-5 and IL-13. IL-
5 is important for maintaining eosinophils, while IL-9 and IL-13 Serum IgE levels and allergic sensitization are well known to be
contribute to mucous production. elevated in all forms of allergic asthma [14]. The Th2 cytokines IL-4
and IL-13 activate class-switch recombination of IgM to produce
Th17 response elevated levels of IgE. Anti-IgE immunotherapy is effective treat-
ment in patients with allergic asthma, though monitoring serum
Th17 cells produce both IL-17 and IL-22, which induce asthma IgE levels in evaluating response to therapy is not routinely recom-
airway remodeling. IL-17 promotes neutrophilic airway infiltration mended [15]. IgE also cannot be used to predict asthma disease
and induces the airway epithelial to mesenchymal morphological severity [16].
transition, while IL-22 increases smooth muscle mass.
Periostin
Genetic predisposition
Periostin is an extracellular matrix protein that is produced by
Asthma is well known to run in families; however, the exact eosinophils, epithelial cells, and fibroblasts and has a role in Th2-
genetic predisposition for asthma is complex. There are some type allergic disease. IL-4 and IL-13 activate eosinophils to produce
genes know to be associated with asthma: ADAM33, PHF11, periostin which then provides positive feedback to promote further
M.D. Gans, T. Gavrilova / Paediatric Respiratory Reviews 36 (2020) 118–127 121

Fig. 1. Th1 (green), Th2 (orange), and Th17 (blue) underlying pathophysiology for asthma with potential biomarkers (grey).

eosinophil production. IL-4, IL-13, and TGF-b act on fibroblasts and cooperation from the patient in order to measure and is therefore
epithelial cells to produce periostin. Periostin also activates fibrob- difficult to perform in young or uncooperative patients.
lasts and epithelial cells to produce NF-jB and promote fibrosis,
leading to airway remodeling. Leukotrienes
Periostin has been shown to be a biomarker for asthma with
frequent exacerbations [17]. However, periostin’s clinical utility Cys-LT is one of the leukotrienes released by intracellular 5-
in children is skewed by normally elevated periostin levels early lipoxygenase within inflammatory cells, such as mast cells. The
in life from bone turnover [18]. production of leukotrienes triggers a Th2 response by the immune
system. Urinary leukotriene E4 (LTE4) has many limitations as col-
FeNO lection of a 24-h specimen is logistically difficult, testing is not
widely available, and concurrent administration of leukotriene
Measuring exhaled nitric oxide is advantageous over other receptor antagonists could influence results.
biomarkers in that it is easily and noninvasively measured in the
office. In bronchial epithelial cells, nitric oxide is produced by indu- TREATMENTS
cible nitric oxide synthase, which is upregulated by IL-4 and IL-13.
Nitric oxide can be beneficial in asthma by stimulating bronchodi- Treatment of asthma focuses on rescue and control therapies.
lation and relaxation of smooth muscle in the airway through cyc- This review will focus on control therapies for asthma (Table 2).
lic GMP, but it can also be detrimental by increasing mucus While the GINA has published guidelines for asthma therapy, these
secretion, plasma extravasation, and cellular injury. guidelines are generalized for all patients [26]. With a greater
FeNO is most commonly used as a marker for allergic asthma understanding of underlying pathophysiology and biomarkers,
[19]. However, FeNO has also been shown to decrease with omal- target-directed agents have become available for patients with
izumab treatment [20] and with adherence to inhaled corticos- severe-persistent asthma. In the upcoming years, step-up therapy
teroids [21]. FeNO might be useful to follow in patients on for patients with moderate-persistent asthma will be tailored
dupilumab, as IL-4 and IL-13 promote nitric oxide formation. A according to their asthma endotype.
Cochrane review in 2016 concluded that utilizing FeNO levels to
tailor asthma therapy did significantly reduce asthma exacerba- Corticosteroids
tions, though there was no significant decrease in hospitalizations
or oral corticosteroid courses [22]. Therefore, the Cochrane review Inhaled corticosteroids are typically the first-line asthma con-
concluded that FeNO levels should not be universally used to guide troller medication for all diagnoses of asthma [26]. Systemic corti-
asthma therapy [22]. Another Cochrane review promoted the same costeroids can be used for acute asthma exacerbations but long-
conclusions in the paediatric population [23]. The American Tho- term treatment with systemic corticosteroids should be avoided
racic Society has advocated that FeNO is best utilized to identify due to potential side effects, especially suppression of the hypotha-
Th2-type asthma with eosinophilic airway inflammation [24], lamic–pituitary–adrenal axis in children [26]. Though the dosage
and there is evidence that FeNO can identify controlled asthmatics of inhaled corticosteroids is lower and less likely to be absorbed
who are at risk of progressive loss of lung function [25]. While systemically, there is some evidence of that there is suppression
FeNO is useful in many different asthma endotypes, FeNO requires of the hypothalamic–pituitary–adrenal axis in children receiving
122 M.D. Gans, T. Gavrilova / Paediatric Respiratory Reviews 36 (2020) 118–127

Table 2
Treatments by asthma endotype.

Asthma endotype Steroid- Leukotriene modifier Other pharmacologic treatments


sensitive responsive
Allergic asthma Yes Yes Allergen immunotherapy, Anti-IgE, Anti-IL-4R,
Early-onset eosinophilic asthma Anti-IL-5
API-positive preschool wheezer Yes Yes Unknown
Infection-induced asthma No No Anti-IgE
Preschooler with episodic viral
wheeze
Viral-exacerbated asthma Partial Partial Anti-IgE
ABPM Yes Partial Anti-IgE
Aspirin-sensitive asthma Yes Yes Aspirin desensitization, Anti-IgE, Anti-IL-5
Airflow obstruction caused by No Yes Unknown
obesity
Premenstrual asthma Partial Yes Oral contraception pills
Neutrophilic asthma No Yes Unknown
Elite-athlete asthma Yes Yes Unknown
Exercise-induced asthma
Cross-country skiers’ asthma No Yes Unknown
Steroid-insensitive eosinophilic No Partial Anti-IL-5
asthma
Late-onset hypereosinophilic Yes Partial Anti-IL-5
asthma
Severe steroid-dependent asthma Yes Partial Anti-IL-5

inhaled corticosteroids [27], and that there is a small reduction in smooth muscle relaxation of the airways. LABAs can be used for
linear growth velocity [28]. The Canadian Society of Allergy and patients with frequent exacerbations on inhaled corticosteroids
Clinical Immunology actually recommends that all paediatric [26]. LABAs should only be used in combination with inhaled cor-
patients receiving high dose inhaled corticosteroids for at least ticosteroids, as there is an increased risk of asthma-related death,
6 months are screened for adrenal suppression [29]. particularly in children, when using LABAs alone [35]. There is no
Maintenance inhaled corticosteroids decrease baseline airway increased risk of asthma-related event in children or adults using
inflammation by dampening inflammatory cell infiltration of the inhaled corticosteroid with LABA as opposed to inhaled corticos-
airway tissue, swelling, and mucus production. Corticosteroids teroid alone [36]. While the use of a combined inhaled corticos-
are able to repair epithelial damage by increasing ciliated cells in teroid and LABA is known to be effective in asthma, there is a
the airway, blunting inflammatory mechanisms that damage the dearth of evidence regarding individual efficacy in various asthma
epithelium, and prompting cell–cell adhesion. Therefore, corticos- endotypes.
teroids are helpful in asthmatic endotypes where the mechanism
is endothelial damage. However, there is conflicting data regarding
whether using higher doses of inhaled corticosteroids for acute Leukotriene modifier
exacerbations is beneficial [30,31].
Corticosteroids help treat the allergic and Th2 components of Leukotriene modifiers are competitive antagonists of Cys-LT,
asthma exacerbations, though Th1 and Th17 responses are and blocking Cys-LT can decrease inflammation and the Th2
steroid-resistant. Th17 cells have high levels of BCL-2 proteins, response. Leukotriene modifiers are less effective alone than with
which are important for regulating cell apoptosis, and protect cells inhaled corticosteroids for patients with mild to moderate asthma
from glucocorticoid-induced apoptosis. [26]. Leukotriene modifiers are most effective in preventing Th2
There are also likely genetic, race, and ethnicity factors that and Th17 pathways for asthma and are ineffective in Th1 path-
influence steroid responsiveness. For instance, a cross-sectional ways. Additionally, leukotriene modifiers are effective in those
study showed that African Americans on inhaled corticosteroids who smoke or have smoke exposure [37], and in those asthmatics
have no significant change in the forced expiratory volume in with small airway disease [38].
one second (FEV1) after bronchodilator administration [32]. Addi-
tionally, predicting systemic corticosteroid responsiveness in chil-
dren using clinical phenotypic features has not been successful, Theophyllines
suggesting that molecular endotyping might be more useful in
making these predictions [33]. In fact, there have been identified Theophylline is another option recommended by the GINA for
single nucleotide polymorphisms (SNPs) that influences inhaled add-on asthma therapy in adults [26]. Theophylline is a methy-
corticosteroid response in children, likely through a mechanism lated xanthine derivative that is a competitive nonselective phos-
of eosinophilic inflammation [34]. These differences in responsive- phodiesterase inhibitor and nonselective adenosine receptor
ness to inhaled corticosteroids – whether by genetics, race, ethnic- antagonist. Molecularly, this causes bronchodilation of the airway
ity, or asthma endotype – are important for practitioners to take smooth muscle. Theophylline also decreases IL-8 secretion, thus
into account when evaluating a patient’s therapeutic response decreasing neutrophil and other inflammatory marker recruit-
and step-up strategy. ment, to have anti-inflammatory effects. Theophylline is typically
reserved for patients who do not respond to other add-on thera-
Long-acting beta agonists pies, often due to concern regarding its low therapeutic index,
though it is safe at low doses. However, its low cost makes theo-
Long-acting beta agonists (LABA) directly activate beta- phylline a potential option for asthma particularly in low-income
adrenergic receptors to decrease bronchoconstriction through and middle-income countries.
M.D. Gans, T. Gavrilova / Paediatric Respiratory Reviews 36 (2020) 118–127 123

Long-acting muscarinic agents Allergen immunotherapy should only be given to patients with
confirmed allergic disease, and there are risks of adverse events to
Long-acting muscarinic agents (LAMAs) are recommended by therapy. Systemic allergic reactions occur in approximately 0.1% of
GINA as potential add-on therapy after inhaled corticosteroids, all injection visits [44]. And in fact, a past or present history of
LABA, and leukotriene modifiers have been trialed [26]. LAMAs uncontrolled or severe asthma is a significant risk factor for a fatal
alleviate bronchoconstriction by blocking acetylcholine. Acetyl- reaction to allergen immunotherapy [44]. However, allergen
choline is released by cholinergic nerves and stimulates bron- immunotherapy is the only treatment for asthma that actually
choconstriction. LAMAs are well known to be effective treatment changes the course of asthma, as it is well known to cause
in chronic obstruction pulmonary disease (COPD), and could be decreased symptoms for years after cessation of therapy [45].
helpful in COPD/asthma overlap syndrome. There could be a role
for LAMAs in neutrophilic asthma, as this is also triggered by Aspirin desensitization
tobacco smoke, though there has not been any confirmatory
research on effectiveness of LAMAs in different asthma endotypes. For patients with aspirin-sensitive asthma, aspirin desensitiza-
tion and continued daily aspirin administration can improve
Macrolide antibiotics long-term symptoms of aspirin-exacerbated respiratory disease
and improve quality of life [46]. Sinus surgery to remove nasal
The macrolide antibiotic azithromycin is typically used to inhi- polyps and topical corticosteroids are typically used in addition
bit bacterial protein synthesis. However, azithromycin assists in to aspirin desensitization for aspirin-sensitive asthma.
decreasing inflammation in asthma through bronchial reepithe-
lization. The AMAZES trial showed that azithromycin could Dihydrofolate reductase inhibitor
decrease asthma exacerbations in adults with uncontrolled asthma
on inhaled corticosteroid and LABA therapy [39]. There is also data For severe steroid-dependent asthma, there have been few ther-
based on subgroup analyses in a Cochrane Review that macrolides apies showing any benefit besides steroids. However, there has
might be potentially beneficial in patients with non-eosinophilic been some promising research showing that methotrexate,
asthma [40]. However, data is still limited and there are significant through its anti-inflammatory effects, can reduce the steroid use
concerns regarding chronic antibiotic use in asthmatics. in these patients, but this is rarely used as there are significant side
effects to this agent [47].

Vitamin D Hormones

Vitamin D likely influences asthma, though its utility as a treat- There is evidence to suggest that oral contraception pills pre-
ment agent is not well established. Vitamin D levels have a nega- vent premenstrual hormone surges and improve premenstrual
tive correlation with Th2 type cytokines, and IL-5, IL-9, and IL-13 asthma symptoms [48]. This is a viable option for those patients
have been shown to decrease after vitamin D supplementation in with premenstrual asthma symptoms who have other medical
patients with asthma [41]. This suggests that vitamin D supple- indications for hormonal therapy.
mentation might decrease the allergic component of asthma and
could alleviate symptoms for those with allergic asthma. A Anti-IgE
Cochrane Review showed that in children and adults with asthma,
vitamin D can reduce the risk of asthma exacerbation [42]. While Omalizumab is a monoclonal antibody administered by subcu-
more trials are needed to elucidate how baseline vitamin D values taneous injection that targets IgE. Omalizumab binds to circulating
and supplementation can be used to help asthmatics, there are IgE and blocks it from binding to IgE receptors on mast cells, result-
many other well-known positive effects of vitamin D supplementa- ing in a decreased release of inflammatory mediators. Omalizumab
tion and insuring an asthmatic patient has adequate vitamin D is effective as an add-on treatment in patients with allergic asthma,
stores is a low-cost and low-risk adjuvant treatment. elevated IgE levels, and severe-persistent severity [26]. There is
limited data on omalizumab’s effectiveness in non-allergic
Allergen immunotherapy patients.
Other targets of IgE, such as quilizumab (NCT01582503) and
Immunotherapy can be administered either by subcutaneous ligelizumab (NCTCQGE031B2201) have not been shown as effec-
immunotherapy (SCIT) or sublingual immunotherapy (SLIT) for tive in patients with severe refractory asthma, while lumiliximab
those with known sensitization to allergens. Allergen immunother- only has preliminary results [49]. However, of all of the emerging
apy can decrease long-term asthma controller medication use and biologic agents for asthma (Table 3), omalizumab has been used
improve FEV1 [43]. the longest and most widely, so it consequently has the longest
The overall mechanism of allergen immunotherapy is that the follow-up for its safety and efficacy (Fig. 2).
treatment shifts the immune response from a Th2 to a Th1 process.
When an allergen is initially exposed to the body, epithelial cells Anti-IL-4R
activate dendritic cells, which present the allergen antigen to naïve
T cells to cause an IgE-mediated Th2 response. However, when Dupilumab is a subcutaneous injection that blocks both IL-4
allergens are repeatedly exposed to the mucosa, the dendritic cells and IL-13 from binding to IL-4Ra. Dupilumab has been shown to
instead produce IL-10, IL-12, and IL-27, which stimulate a Th1 be effective as add-on therapy in patients with poorly controlled
response. T-regulatory cells and B-regulatory cells are also acti- severe asthma, irrespective of eosinophil counts [50].
vated by these cytokines, subsequently blocking the Th2 response
directly and producing antibodies to further block the Th2 Anti-IL-5
response. IgG4 also greatly increases during allergen immunother-
apy, possibly due to it down regulating activation of mast cells and Mepolizumab, reslizumab, and benralizumab are all anti-IL-5
basophils, though its exact function in immune tolerance is therapy. Mepolizumab and reslizumab directly bind to IL-5 and
unknown. prevent IL-5 from binding to IL-5Ra on eosinophils, while benral-
124 M.D. Gans, T. Gavrilova / Paediatric Respiratory Reviews 36 (2020) 118–127

Table 3 and are most effective in patients with elevated absolute eosino-
FDA approved biologic treatments for asthma. phil counts [26,51–53].
Drug Dosage Minimum Other approved With multiple options for eosinophilic asthma, practitioners are
approved indications often faced with the challenge of choosing a biologic agent. A net-
age for work meta-analysis comparing benralizumab and reslizumab sug-
asthma
gests that reslizumab may be more efficacious than benralizumab
Omalizumab 75–375 mg SC every 2 or 6 years Chronic in select patients [54]. While logistical factors, such as administra-
4 weeks idiopathic
urticaria
tion route and frequency often influence a provider’s choice, there
Dupilumab 400 mg SC followed by 12 years Atopic are no head-to-head studies to date examining efficacy of the dif-
200 mg SC every other Dermatitis ferent anti-IL-5 therapies or comparing anti-IL-5 therapy to the
week or 600 mg SC other FDA approved biologic treatments for asthma: anti-IgE and
followed by 300 mg SC
anti-IL-4R.
every other week
Mepolizumab 100 mg SC every 4 weeks 12 years Eosinophilic
granulomatosis Anti-IL-1R
with
polyangiitis
Reslizumab 3 mg/kg IV every 4 weeks 18 years N/A
Anakinra binds to the IL-1 type 1 receptor to prevent IL-1a and
Benralizumab 30 mg SC every 4 weeks 12 years N/A IL-1b from binding to their receptor. In neutrophilic asthma, IL-1b
for first 3 doses followed supports Th17 differentiation. Initial studies have shown anakinra
by 30 mg SC every 8 weeks to be effective in reducing neutrophilic airway inflammation [55].

Anti-IL-13
izumab binds directly to IL-5Ra to block IL-5 from binding to its
receptor. While IL-5 typically stimulates terminal differentiation Lebrikizumab and tralokinumab block IL-13, a Th2 cytokine.
of eosinophils in the bone marrow, anti-IL-5 targets eosinophil However, both lebrikizumab (NCT01867125, NCT01868061) and
and basophils for apoptosis by antibody-dependent cell-mediated tralokinumab (NCT02281357, NCT02194699, NCT02161757) have
cytotoxicity. Mepolizumab, benralizumab, and reslizumab are rec- not been shown to significantly decrease exacerbation rates in
ommended as add-on treatment for severe eosinophilic asthma, asthmatics.

Fig. 2. All biologic treatments for asthma that are currently FDA approved or in development. Solid line is research completed and dotted line is research underway. Arrows
indicate studies showed favorable results, while circles indicate that the studies either showed unfavorable results or were terminated.
M.D. Gans, T. Gavrilova / Paediatric Respiratory Reviews 36 (2020) 118–127 125

Anti-IL-17 described endotypes. This review demonstrates the importance


of the shift in the practice of asthma from a catchall diagnosis with
There is currently no approved biologic agent targeting neu- standardized treatments to the classification of asthma endotypes
trophilic asthma, which is driven by a predominant Th17 response. with targeted therapy. While all of the tools needed for asthma
Secukinumab, an antibody against IL-17A, (NCT01478360) has not endotyping are not yet widely available to the common practi-
been shown to significantly decrease asthma symptoms, while bro- tioner, practitioners can attempt to endotype patients with the
dalumab, an antibody against IL-17R, (NCT01199289) has only had information currently known. Endotyping is the future of asthma.
moderate success.
DIRECTIONS FOR FUTURE RESEARCH
Anti-IL-25
 This article summarizes the research to date on asthma endo-
ABM125 has been hypothesized as a treatment for asthma by types but there is much to discover.
targeting IL-25, though there have only been mouse studies to date  Genome-wide association studies will help identify genetic
[56]. variants associated with underlying asthma pathophysiology.
 There are also many clinical trials underway to evaluate optimal
Anti-IL-33 therapies for various asthma endotypes.
 In the upcoming years, we can expect an increase in safety and
ANB020, which targets IL-33, has also been proposed as a clinical efficacy data for emerging biologic therapies for asthma.
potential asthma treatment, though only mouse studies have been
performed thus far [57].
ACKNOWLEDGEMENTS
Anti-TSLP
None.
Tezepelumab is an anti-TSLP antibody that binds to TSLP and
prevents it from binding to its receptor. Initial studies have been FUNDING SOURCES
promising (NCT02054130), showing decreased asthma
exacerbations. This research did not receive any specific grant from funding
agencies in the public, commercial, or not-for-profit sectors.
CRTh2 antagonist
Practice points
CRTh2 is a prostaglandin receptor that perpetuates chemotaxis
of Th2 cells and anti-CRTh2 is available as a daily oral medication.  Asthma is caused by an interplay of Th1, Th2, and Th17
While clinical trials have not yet shown effectiveness for AZD1981 immunologic mechanisms in addition to a genetic
in treating asthma (NCT01197794), there has been some efficacy predisposition.
for fevipiprant (NCT01437735).  Eosinophils, neutrophils, IgE, periostin, FeNO, and leuko-
trienes are biomarkers that can be used in clinical practice
Bronchial thermoplasty to guide selection of treatment and track response to
treatment.
The FDA approved bronchial thermoplasty in 2010 for patients  For severe persistent asthmatics, there is a range of differ-
with severe asthma. Bronchial thermoplasty is a minimally inva- ent treatments available to step-up therapy.
sive outpatient procedure where thermal energy is applied to the
airway wall to reduce smooth muscle mass. Research has shown
that bronchial thermoplasty can greatly improve asthma control
References
up to 5 years post-procedure in severe asthmatics, and results
are similar between allergic and non-allergic patients [58]. Histo- [1] National Center for Health Statistics. National Health Interview Survey, 2016.
logically, there is no evidence of scarring post-procedure [59]. Public-use data file and documentation. http://www.cdc.gov/nchs/nhis/
There is, however, a lack of data on the efficacy of bronchial ther- quest_data_related_1997_forward.htm. 2018.
[2] Global Initiative for Asthma, National Heart Lung and Blood Institute. Global
moplasty in different asthma endotypes. There are also potential initiative for asthma: global strategy for asthma management and prevention
short and long-term adverse effects from bronchial thermoplasty. (2016 update). Bethsda, MD: U.S. Dept. of Health and Human Services, Public
Immediately after the procedure, patients can have shortness of Health Service; 2016.
[3] Lotvall J, Akdis CA, Bacharier LB, et al. Asthma endotypes: a new
breath and lobe collapse. The long-term adverse effects beyond approach to classification of disease entities within the asthma syndrome.
5 years post-bronchial thermoplasty are largely unknown. J Allergy Clin Immunol 2011;127(2):355–60. https://doi.org/10.1016/
j.jaci.2010.11.037.
[4] Corren J. Asthma phenotypes and endotypes: an evolving paradigm for
CONCLUSIONS classification. Discov Med 2013;15(83):243–9.
[5] World Health Organization. Genetics and asthma: World Health Organization;
[Available from: http://www.who.int/genomics/about/Asthma.pdf accessed
Asthma is a common heterogeneous disease with complex April 4 2018.
underlying pathophysiology. Understanding the immunology of [6] Davies ER, Kelly JF, Howarth PH, et al. Soluble ADAM33 initiates airway
this disease can greatly aid in personalized, specific, directed ther- remodeling to promote susceptibility for allergic asthma in early life. JCI
Insight 2016;1(11). https://doi.org/10.1172/jci.insight.87632.
apy for all patients suffering from asthma. Additionally, the knowl-
[7] Muscat P, Mercado K, Payne K, et al. PHF11 expression and cellular distribution
edge of a patient’s endotype can help to determine the patient’s is regulated by the toll-like receptor 3 ligand polyinosinic: polycytidylic Acid in
asthma prognosis and in counseling regarding prevention strate- HaCaT keratinocytes. BMC Immunol 2015;16:69. https://doi.org/10.1186/
gies. However, there are limitations to asthma endotyping. Not s12865-015-0131-y.
[8] Postma DS, Koppelman GH. Confirmation of GPRA: a putative drug target for
all patients perfectly fit into a described endotype and many asthma. Am J Respir Crit Care Med 2005;171(12):1323–4. https://doi.org/
patients likely have underlying immunology overlapping several 10.1164/rccm.2503006.
126 M.D. Gans, T. Gavrilova / Paediatric Respiratory Reviews 36 (2020) 118–127

[9] Kim SH, Choi H, Yoon MG, et al. Dipeptidyl-peptidase 10 as a genetic biomarker [33] Fitzpatrick AM, Stephenson ST, Brown MR, et al. Systemic corticosteroid
for the aspirin-exacerbated respiratory disease phenotype. Ann Allergy responses in children with severe asthma: phenotypic and endotypic features
Asthma Immunol 2015;114(3):208–13. https://doi.org/10.1016/j. 410–419 e4. J Allergy Clin Immunol Pract 2017;5(2). https://doi.org/10.1016/
anai.2014.12.003. j.jaip.2016.08.001.
[10] Kabesch M, Carr D, Weiland SK, et al. Association between polymorphisms in [34] Levin AM, Gui H, Hernandez-Pacheco N, et al. Integrative approach identifies
serine protease inhibitor, kazal type 5 and asthma phenotypes in a large corticosteroid response variant in diverse populations with asthma. J Allergy
German population sample. Clin Exp Allergy 2004;34(3):340–5. Clin Immunol 2019;143(5):1791–802. https://doi.org/10.1016/
[11] de Groot JC, Ten Brinke A, Bel EH. Management of the patient with eosinophilic j.jaci.2018.09.034.
asthma: a new era begins. ERJ Open Res 2015;1(1). https://doi.org/10.1183/ [35] McMahon AW, Levenson MS, McEvoy BW, et al. Age and risks of FDA-approved
23120541.00024-2015. long-acting beta(2)-adrenergic receptor agonists. Pediatrics 2011;128(5):
[12] Petsky HL, Cates CJ, Lasserson TJ, et al. A systematic review and meta-analysis: e1147–54. https://doi.org/10.1542/peds.2010-1720.
tailoring asthma treatment on eosinophilic markers (exhaled nitric oxide or [36] Busse WW, Bateman ED, Caplan AL, et al. Combined analysis of asthma safety
sputum eosinophils). Thorax 2012;67(3):199–208. https://doi.org/10.1136/ trials of long-acting beta2-agonists. N Engl J Med 2018;378(26):2497–505.
thx.2010.135574. https://doi.org/10.1056/NEJMoa1716868.
[13] Tran TN, Khatry DB, Ke X, et al. High blood eosinophil count is associated with [37] Pacheco Y, Freymond N, Devouassoux G. Impact of montelukast on asthma
more frequent asthma attacks in asthma patients. Ann Allergy Asthma associated with rhinitis, and other triggers and co-morbidities. J Asthma
Immunol 2014;113(1):19–24. https://doi.org/10.1016/j.anai.2014.04.011. 2014;51(1):1–17. https://doi.org/10.3109/02770903.2013.822081.
[14] Burrows B, Martinez FD, Halonen M, et al. Association of asthma with serum [38] Marcello C, Carlo L. Asthma phenotypes: the intriguing selective intervention
IgE levels and skin-test reactivity to allergens. N Engl J Med 1989;320 with Montelukast. Asthma Res Pract 2016;2:11. https://doi.org/10.1186/
(5):271–7. https://doi.org/10.1056/NEJM198902023200502. s40733-016-0026-6.
[15] Hamilton RG. Monitoring allergic patients on omalizumab with free and total [39] Gibson PG, Yang IA, Upham JW, et al. Effect of azithromycin on asthma
serum IgE measurements. J Allergy Clin Immunol Pract 2016;4(2):366–8. exacerbations and quality of life in adults with persistent uncontrolled asthma
https://doi.org/10.1016/j.jaip.2015.12.002. (AMAZES): a randomised, double-blind, placebo-controlled trial. Lancet
[16] Davila I, Valero A, Entrenas LM, et al. Relationship between serum total IgE and 2017;390(10095):659–68. https://doi.org/10.1016/S0140-6736(17)31281-3.
disease severity in patients with allergic asthma in Spain. J Investig Allergol [40] Kew KM, Undela K, Kotortsi I, et al. Macrolides for chronic asthma. Cochrane
Clin Immunol 2015;25(2):120–7. Database Syst Rev 2015;9:. https://doi.org/10.1002/14651858.CD002997.
[17] Scichilone N, Crimi C, Benfante A, et al. Higher serum levels of periostin and pub4CD002997.
the risk of exacerbations in moderate asthmatics. Asthma Res Pract 2016;2:1. [41] Batmaz SB, Arikoglu T, Tamer L, et al. Seasonal variation of asthma control,
https://doi.org/10.1186/s40733-015-0019-x. lung function tests and allergic inflammation in relation to vitamin D levels: a
[18] Anderson HM, Lemanske Jr RF, Arron JR, et al. Relationships among prospective annual study. Postepy Dermatol Allergol 2018;35(1):99–105.
aeroallergen sensitization, peripheral blood eosinophils, and periostin in https://doi.org/10.5114/ada.2017.71421.
pediatric asthma development. J Allergy Clin Immunol 2017;139(3):790–6. [42] Whitehead L. Vitamin D for the management of asthma: a Cochrane review
https://doi.org/10.1016/j.jaci.2016.05.033. summary. Int J Nurs Stud 2017;73:105–6. https://doi.org/10.1016/j.
[19] Djuric-Filipovic I, Caminati M, Filipovic D, et al. Effects of specific allergen ijnurstu.2017.04.003.
immunotherapy on biological markers and clinical parameters in asthmatic [43] Rice JL, Diette GB, Suarez-Cuervo C, et al. Allergen-specific immunotherapy in
children: a controlled-real life study. Clin Mol Allergy 2017;15:7. https://doi. the treatment of pediatric asthma: a systematic review. Pediatrics 2018.
org/10.1186/s12948-017-0064-5. https://doi.org/10.1542/peds.2017-3833.
[20] Mansur AH, Srivastava S, Mitchell V, et al. Longterm clinical outcomes of [44] Epstein TG, Liss GM, Berendts KM, et al. AAAAI/ACAAI subcutaneous
omalizumab therapy in severe allergic asthma: Study of efficacy and safety. immunotherapy surveillance study (2013–2017): fatalities, infections,
Respir Med 2017;124:36–43. https://doi.org/10.1016/j.rmed.2017.01.008. delayed reactions, and use of epinephrine autoinjectors. J Allergy Clin
[21] Demarche SF, Schleich FN, Henket MA, et al. Effectiveness of inhaled Immunol Pract 2019. https://doi.org/10.1016/j.jaip.2019.01.058.
corticosteroids in real life on clinical outcomes, sputum cells and systemic [45] Canonica GW, Cox L, Pawankar R, et al. Sublingual immunotherapy: World
inflammation in asthmatics: a retrospective cohort study in a secondary care Allergy Organization position paper 2013 update. World Allergy Organ J
centre. BMJ Open 2017;7(11):. https://doi.org/10.1136/bmjopen-2017- 2014;7(1):6. https://doi.org/10.1186/1939-4551-7-6.
018186e018186. [46] Walters KM, Waldram JD, Woessner KM, et al. Long-term clinical outcomes of
[22] Petsky HL, Kew KM, Turner C, et al. Exhaled nitric oxide levels to guide aspirin desensitization with continuous daily aspirin therapy in aspirin-
treatment for adults with asthma. Cochrane Database Syst Rev 2016;9:. exacerbated respiratory disease. Am J Rhinol Allergy 2018;32(4):280–6.
https://doi.org/10.1002/14651858.CD011440.pub2CD011440. https://doi.org/10.1177/1945892418770260.
[23] Petsky HL, Kew KM, Chang AB. Exhaled nitric oxide levels to guide treatment [47] Bilocca D, Hargadon B, Pavord ID, et al. The role of oral methotrexate as a
for children with asthma. Cochrane Database Syst Rev 2016;11:. https://doi. steroid sparing agent in refractory eosinophilic asthma. Chron Respir Dis
org/10.1002/14651858.CD011439.pub2CD011439. 2018;15(1):85–7. https://doi.org/10.1177/1479972317709650.
[24] Dweik RA, Boggs PB, Erzurum SC, et al. An official ATS clinical practice [48] Graziottin A, Serafini A. Perimenstrual asthma: from pathophysiology to
guideline: interpretation of exhaled nitric oxide levels (FENO) for clinical treatment strategies. Multidiscip Respir Med 2016;11:30. https://doi.org/
applications. Am J Respir Crit Care Med 2011;184(5):602–15. https://doi.org/ 10.1186/s40248-016-0065-0.
10.1164/rccm.9120-11ST. [49] Rosenwasser LJ, Busse WW, Lizambri RG, et al. Allergic asthma and an anti-
[25] Matsunaga K, Hirano T, Oka A, et al. Persistently high exhaled nitric oxide and CD23 mAb (IDEC-152): results of a phase I, single-dose, dose-escalating
loss of lung function in controlled asthma. Allergol Int 2016;65(3):266–71. clinical trial. J Allergy Clin Immunol 2003;112(3):563–70.
https://doi.org/10.1016/j.alit.2015.12.006. [50] Rabe KF, Nair P, Brusselle G, et al. Efficacy and safety of dupilumab in
[26] Bateman ED, Hurd SS, Barnes PJ, Bousquet J, Drazen JM, FitzGerald JM, et al. glucocorticoid-dependent severe asthma. N Engl J Med 2018;378
Global strategy for asthma management and prevention: GINA executive (26):2475–85. https://doi.org/10.1056/NEJMoa1804093.
summary Eur Respir J 2008; 31: 143–178. Eur Respir J 2018;51(2). https://doi. [51] Corren J, Weinstein S, Janka L, et al. Phase 3 study of reslizumab in patients with
org/10.1183/13993003.51387-2007. poorly controlled asthma: effects across a broad range of eosinophil counts.
[27] Zollner EW, Lombard CJ, Galal U, et al. Hypothalamic-pituitary-adrenal axis Chest 2016;150(4):799–810. https://doi.org/10.1016/j.chest.2016.03.018.
suppression in asthmatic school children. Pediatrics 2012;130(6):e1512–9. [52] Bleecker ER, FitzGerald JM, Chanez P, et al. Efficacy and safety of benralizumab
https://doi.org/10.1542/peds.2012-1147. for patients with severe asthma uncontrolled with high-dosage inhaled
[28] Gradman J, Wolthers OD, Kan H, et al. A randomized trial of lower leg and corticosteroids and long-acting beta2-agonists (SIROCCO): a randomised,
height growth in children with asthma treated with inhaled budesonide from multicentre, placebo-controlled phase 3 trial. Lancet 2016;388
a new dry powder inhaler. Pediatr Allergy Immunol. 2010;21(1 Pt 2):e206–12. (10056):2115–27. https://doi.org/10.1016/S0140-6736(16)31324-1.
https://doi.org/10.1111/j.1399-3038.2009.00875.x. [53] Lugogo N, Domingo C, Chanez P, et al. Long-term efficacy and safety of
[29] Issa-El-Khoury K, Kim H, Chan ES, et al. CSACI position statement: systemic mepolizumab in patients with severe eosinophilic asthma: a multi-center,
effect of inhaled corticosteroids on adrenal suppression in the management of open-label, phase IIIb study 2058–2070 e1. Clin Ther 2016;38(9). https://doi.
pediatric asthma. Allergy Asthma Clin Immunol 2015;11(1):9. https://doi.org/ org/10.1016/j.clinthera.2016.07.010.
10.1186/s13223-015-0075-z. [54] Casale TB, Pacou M, Mesana L, et al. Reslizumab compared with benralizumab
[30] Jackson DJ, Bacharier LB, Mauger DT, et al. Quintupling inhaled glucocorticoids in patients with eosinophilic asthma: a systematic literature review and
to prevent childhood asthma exacerbations. N Engl J Med 2018;378 network meta-analysis. J Allergy Clin Immunol Pract 2018. https://doi.org/
(10):891–901. https://doi.org/10.1056/NEJMoa1710988. 10.1016/j.jaip.2018.08.036.
[31] McKeever T, Mortimer K, Wilson A, et al. Quadrupling inhaled glucocorticoid [55] Hernandez ML, Mills K, Almond M, et al. IL-1 receptor antagonist reduces
dose to abort asthma exacerbations. N Engl J Med 2018;378(10):902–10. endotoxin-induced airway inflammation in healthy volunteers. J Allergy Clin
https://doi.org/10.1056/NEJMoa1714257. Immunol 2015;135(2):379–85. https://doi.org/10.1016/j.jaci.2014.07.039.
[32] Naqvi M, Tcheurekdjian H, DeBoard JA, et al. Inhaled corticosteroids and [56] Bartlett N, Girkin J, Williams T, et al. ABM125 anti-IL-25 antibody pre-clinical
augmented bronchodilator responsiveness in Latino and African American development for viral asthma exacerbations identifies IL-25 mediated
asthmatic patients. Ann Allergy Asthma Immunol 2008;100(6):551–7. https:// regulation of type-2- and anti-viral immunity. Am J Respir Crit Care Med
doi.org/10.1016/S1081-1206(10)60055-5. 2018;197(A7759).
M.D. Gans, T. Gavrilova / Paediatric Respiratory Reviews 36 (2020) 118–127 127

[57] Lei Y, Boinapally V, Zoltowska A, et al. Vaccination against IL-33 inhibits Clin Immunol 2013;132(6):1295–302. https://doi.org/10.1016/
airway hyperresponsiveness and inflammation in a house dust mite model of j.jaci.2013.08.009.
asthma. PLoS ONE 2015;10(7):. https://doi.org/10.1371/journal. [59] Miller JD, Cox G, Vincic L, et al. A prospective feasibility study of bronchial
pone.0133774e0133774. thermoplasty in the human airway. Chest 2005;127(6):1999–2006. https://
[58] Wechsler ME, Laviolette M, Rubin AS, et al. Bronchial thermoplasty: Long-term doi.org/10.1378/chest.127.6.1999.
safety and effectiveness in patients with severe persistent asthma. J Allergy

You might also like