Next Article in Journal
Evaluating the Quality of Raw Chocolate as an Alternative to Commercial Products
Next Article in Special Issue
A Preliminary Investigation of Mobile Respiratory Function Testing in Western Australian Communities
Previous Article in Journal
A New Method to Detect Splicing Image Forgery Using Convolutional Neural Network
Previous Article in Special Issue
Prevalence and Outcome of Asthma in Adult Patients Admitted to the Emergency Department for COVID-19: A Case-Control Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Diagnosis and Treatment in Asthma and Allergic Rhinitis: Past, Present, and Future

by
Marta Espada-Sánchez
1,†,
Rocío Sáenz de Santa María
1,2,†,
María del Carmen Martín-Astorga
1,3,
Clara Lebrón-Martín
1,
María Jesús Delgado
1,
Ibón Eguiluz-Gracia
1,2,
Carmen Rondón
1,2,
Cristobalina Mayorga
1,2,
María José Torres
1,2,3,
Carlos José Aranda
1,*,‡ and
José Antonio Cañas
1,*,‡
1
Allergy Research Group, Instituto de Investigación Biomédica de Málaga-Plataforma BIONAND (IBIMA-BIONAND), 29009 Málaga, Spain
2
Allergy Unit, Hospital Regional Universitario de Málaga, 29009 Málaga, Spain
3
Medicine Department, Universidad de Málaga (UMA), 29010 Málaga, Spain
*
Authors to whom correspondence should be addressed.
These authors contribute equally to this work.
These authors contribute equally to this work.
Appl. Sci. 2023, 13(3), 1273; https://doi.org/10.3390/app13031273
Submission received: 21 December 2022 / Revised: 13 January 2023 / Accepted: 16 January 2023 / Published: 18 January 2023
(This article belongs to the Special Issue Asthma and Respiratory Disease: Prediction, Diagnosis and Treatment)

Abstract

:
Respiratory diseases are pathological conditions that affect airways, hampering breathing and causing high mortality. In particular, asthma and allergic rhinitis (AR) are two of the most common airway diseases that affect millions of people and have a high prevalence in childhood and adulthood. Asthma is a heterogeneous chronic inflammatory disease characterized by wheezing, chest tightness, shortness of breath, and cough. AR occurs with rhinorrhea, nasal congestion, and sneezing. Indeed, these pathologies share common physiopathological mechanisms such as airway hyperresponsiveness and similar immunopathology such as tissue eosinophilia and T-helper type 2 inflammation. Moreover, AR can be an important risk factor for suffering asthma. Thus, early diagnosis and effective treatment are crucial to improving the health and quality of life of these patients. Classical drugs such as corticosteroids have been used; however, in the last decades, efforts to improve treatments have increased, focusing on biological agents and specific allergen immunotherapy development. Moreover, more precise diagnostic tools have been elaborated, besides classical methods (medical history, physical examination, and pulmonary function tests), such as basophil activation test, and specific cellular and molecular biomarkers (microRNAs, sputum/blood eosinophils, IgE serum, and periostin levels). Therefore, in this review, we compile all these important issues for managing asthma and AR.

1. Introduction

Respiratory diseases are pathological disorders of the airways and other parts of the lungs that represent a major health and economic burden, with high prevalence, and one of the leading causes of mortality and morbidity worldwide [1]. They are caused by multiple triggers, with air pollutants (nitrogen dioxide, carbon monoxide, or some volatile organic compounds), allergens (pollen, dust mite, pet dander, mold), biological agents (viruses, bacteria, fungi), and smoking tobacco being the most common ones [2]. Respiratory diseases comprise a wide spectrum of pathologies, with asthma, chronic obstructive pulmonary disease (COPD), lung cancer, and rhinitis being the most frequent ones [3]. Additionally, COVID-19 has become a common respiratory pathology in the last few years [4].
The management and treatment of respiratory diseases have always been a challenge for clinicians, especially due to the pathological heterogeneity of these diseases. Thus, the correct diagnosis and choice of effective treatment are crucial to improving the health of patients suffering from these diseases. In the present review, we focus on the diagnosis and therapeutic tools for asthma and allergic rhinitis (AR), recapitulating the advances and research performed in the last years.

2. Materials and Methods

The search of literature was performed between November and December 2022 in the PubMed database, using the following terms for asthma: “asthma” AND “clinical diagnosis” OR “miRNAs” OR “metabolomics” OR “proteomics” OR “transcriptomic” OR “inhaled drugs” OR “monoclonal antibody therapy” OR “allergen immunotherapy”. In the same line, for AR we used the following terms: “allergic rhinitis” AND “clinical diagnosis” OR “molecular diagnosis” OR “inhaled therapy” OR “monoclonal antibody therapy” OR “allergen immunotherapy”. We tried to omit general terms as “treatment” to prevent the ambiguity of irrelevant articles.
Original articles, reviews, clinical trials, randomized clinical trials, systematic reviews, and meta-analyses indexed from January 2000 to November 2021 using the search terms described above were searched. Mainly, we focused on eligible studies published since 2015, although earlier studies were also included if they were considered relevant to the topic. The inclusion criteria for the articles were the following: (i) written in English; (ii) performed on human subjects, in both adults and children; and (iii) articles or clinical guidelines for the management of asthma and allergic rhinitis. The exclusion criteria were the following: (i) animal and in silico studies; (ii) articles not written in English; (iii) articles mainly focused on other diseases that only mentioned asthma and AR; (iv) abstracts (not full text); and (v) studies lacking controls such as control case studies. A flowchart of the literature and article search can be observed in Figure 1.

3. Results

3.1. Asthma

Asthma is a chronic and heterogeneous respiratory disease characterized by airway inflammation and hyperresponsiveness, wheezing, shortness of breath, chest tightness, and cough [5]. It affects 300 million people around the world. Although its prevalence, morbidity, and mortality vary globally, asthma produced almost 500,000 deaths in 2019 [6]. Therefore, this respiratory pathology has become an important social and economic problem for multiple countries (high consumption of healthcare resources), making early diagnosis and effective treatment crucial. Despite its heterogeneity, asthma is presented with several phenotypes and endotypes, which makes, in most cases, its diagnosis difficult [7]. Traditionally, asthma has been divided endotypically into type 2 (T2) asthma and non-T2 asthma, and phenotypically into atopic, late-onset, obesity-related, or non-inflammatory phenotypes [7,8]. T2 asthma is triggered by allergic contaminants producing a typical T2 immune response, with cellular components as eosinophils, basophils, dendritic cells (DCs), immunoglobulin E (IgE)-producing cells, T-helper type 2 (Th2) cells, and pro-inflammatory cytokines being key players [9,10]. On the other hand, non-T2 asthma usually manifests itself in more severe states, where Th1 and Th17 cells, neutrophils, and non-Th2 cytokines are commonly present [11,12].
Taking this into account, effective treatments and diagnoses are needed. Hence, in this part of review, we focus on the different methods to diagnose this respiratory pathology and on how it can be treated.

3.1.1. Asthma Diagnosis

Clinical Diagnosis of Asthma

Over many years, the diagnosis of asthmatic pathology has been made by assessing the evaluating airflow limitation, bronchodilator responses, and by bronchial provocation challenges [13]. Additionally, the use of several biomarkers, such as fractional exhaled nitric oxide (FeNO), forced vital capacity (FVC), forced expiratory volume in 1 s (FEV1), and peripheral blood eosinophils, are of great utility (Figure 2). However, due to the heterogeneity in the pathomechanisms involved in this disease, currently, there are many challenges related to the development of more personalized and precise diagnostic tools [14].
The development in recent years of drawing up guidelines updated by societies and working groups has led to an improved approach and clinical management of asthma [15]. An anamnesis should always be carried out to search for possible triggers and a complete allergological study should also be performed: with skin prick tests (SPT), detection of total and specific IgE (sIgE), and an allergen-specific nasal or bronchial challenge test to indicate an atopic state (Figure 2) [16]. Although there is no gold standard test, spirometry is essential, despite it maybe not being sufficient to confirm the diagnosis. Then, the spirometry parameters include FEV1 and FVC, which are variable according to age, height, weight, sex, and ethnic group. The main alterations in asthma are airflow obstruction, reversibility, variability, and bronchial hyperresponsiveness, so a reversible obstruction that improves after short-acting beta-agonists (SABA) administration confirms the diagnosis, although a normal result does not exclude it. Variations in peak expiratory flow (PEF) and FEV1 between exacerbations and stable periods in the same patient are also common. Spirometry with bronchodilator test and PEF measurement should be performed prior to the initiation of treatment, since as lung function improves, variability decreases. In cases where spirometry values comprised the normal range (FEV1/FVC > 0.7, FEV1 increase < 12%), it is possible to confirm the diagnosis by bronchial challenge with direct (methacholine) [17], indirect (mannitol, physical exercise) [18] or allergen-specific (decreasing FEV1 ≥ 20%) bronchoconstrictor agents, measurement of nitric oxide in exhaled air (FeNO > 40ppb) [19], diurnal variability of peak expiratory flow (PEF ≥ 20%) [20], or analysis of induced sputum (levels of sputum eosinophils, elevated values of eosinophil cationic proteins and Creola bodies) [21]. In addition, when a diagnosis cannot be reached, normalization of the pattern after treatment with oral glucocorticoids (GCs) for 14–21 days confirms it [22].
In any case, a correct differential diagnosis should be always reached, especially in treatment-refractory cases and if the evolution is not favorable. Imaging tests such as thorax radiography can rule out other respiratory diseases (signs of air trapping, atelectasis, or bronchial thickening) [15]. Establishing the phenotype (total and specific IgE determination, peripheral eosinophilia, FeNO measurement, and induced sputum), mainly in patients with severe asthma, allows for improvement in the clinical management of these patients and to offer them the most appropriate treatment.

Cellular Diagnostic Parameters and Biomarkers in Asthma

As support to clinical diagnosis, biomarkers and cellular and molecular parameters have been developed to diagnose asthma pathology (Figure 2). Asthma classification (T2 and non-T2) enables the establishment of an effective treatment strategy [23,24,25]. Th2 cells, type 2 innate lymphoid cells (ILC2), eosinophils, mast cells, and M2 macrophages are involved in T2 inflammation, characteristic of T2 asthma, which is primarily caused by the activation of the Th2 cells and ILC2, producing Th2 cytokines (IL-4, IL-5, IL-9, IL-13, and IL-31) [26,27]. At the cellular level, several immune cells (eosinophils, Th2 cells, ILC2, B cells, and mast cells) are present in T2 asthma. On the other hand, it is also known that neutrophils, Th1, and Th17 cells are the main mediators in non-T2 asthma [26]. Although the phenotypes of non-T2 asthma are still poorly known (some associated symptoms include obesity, smoking, and psychological factors), the innate immunological, metabolic, and epigenetic indicators must be considered [8]. Therefore, searching for new biomarkers and cellular diagnostic tools for allergy disorders, including asthma, has been the focus of a plethora of studies in recent years [28,29].
The basophil activation test (BAT) is a functional assay that evaluates the percentage of degranulation of the basophils after allergen incubation by flow cytometry. In the last few years, BAT has become a novel promise diagnostic tool for allergic asthma [30]. Basophil sensitivity, measured by CD-sens, has been demonstrated as a useful parameter in the diagnosis of allergic asthma, correlating with the dose of allergen used in the bronchial challenge that causes a 20% drop in FEV1 [31].
On the other hand, biomarkers are important parameters for precision medicine as they provide information on disease endotypes, clusters, precise diagnoses, identification of therapeutic targets, and tracking of therapy efficacies. Several studies have shown some clinically applicable cell point-of-care biomarkers [11,32,33,34,35], including sputum and blood eosinophils [36,37], circulating ILC2s expressing chemokine receptor (CCR) 10, plasmatic CCL27 levels, and an increase in serum IgE (Figure 2). Moreover, innate epithelial cytokines (IL-25, IL-33, and thymic stromal lymphopoietin [TSLP]), Th2 cytokines (IL-4 and IL-13), and receptor for advanced glycation end products (RAGE) are also important biomarkers in asthma [32,38,39,40,41,42,43]. In contrast, non-T2 asthma typically relies on neutrophilic or paucigranulocytic patterns [11,44,45], so calprotectin and HMGB1 levels are also useful biomarkers for this type of asthma. Although these biomarkers could provide limited information [37], they are used for multiple approaches: disease diagnosis, selection of targeted therapy, disease monitoring, and prediction of prognosis [46].

Molecular Diagnostic Parameters in Asthma: Proteomics, Metabolomics, and Transcriptomics Analysis

Several novel diagnostic tools are under current investigation, such as other biomarkers that may be measured in serum, bodily fluids, and exhaled air, such as pro-inflammatory mediators, micro RNAs (miRNAs), eicosanoid molecules, epithelial barrier integrity, and microbiota alterations [24,47,48] (Figure 2). They are helpful in the diagnosis and monitoring of allergic disorders [24,26,49,50].
New technologies such as imaging, artificial intelligence, and omics models further enable a precision treatment strategy for asthma [8]. Specifically, omics studies [51], such as proteomics [52,53], metabolomics [54], and transcriptomic (real-time PCR, microarrays, and RNA sequencing), have been recently used as tools for the identification of novel biomarkers as diagnostic tools. These are becoming even more necessary because of the use of biologicals in clinical practice for patient selection, outcome prediction, and monitoring, allowing for a suitable choice of how long to administer these expensive and long treatments [24].
On the other hand, the function of metabolites in allergic disorders has recently attracted scientific interest. Several pathophysiological processes in allergic diseases are influenced by eicosanoids, which include thromboxanes, leukotrienes, prostaglandins, and lipoxins [55]. In addition, periostin is an extracellular matrix protein that exhibits surrogate indicators for T2 immunity and tissue remodeling, making it a useful diagnostic marker for the early identification of asthma [56,57,58].
According to transcriptomic analysis, chromosome 17q21 is of interest in genetic epidemiological studies of asthma because it contains important genes (ORMDL3, GSDMB, LRRC3C, GSDMA, ZPBP2, IKZF3, GRB7, ERBB2, and PGAP3) and three single nucleotide polymorphisms (SNPs) (rs4794820, rs807631, and rs2872507) that are strongly associated with the pathogenesis and severity of asthma [26,59]. However, a multitude of factors (innate and adaptive immune response, metabolic pathways, microbiome infections, epithelial barrier, genetic and epigenetic factors, remodeled resident cells, anatomical factors, exposome allergens, irritants, pollutant- and psychosocial factors) can induce or suppress certain genes or pathways and may play a role in the development of certain phenotypes and endotypes as well as in the control of asthma [25]. In addition, differential gene expression related to cytokines and transcription factors, as well as immunological response and elevation of numerous cellular processes, are influenced by DNA methylation [60] and histone modifications [61,62,63]. Therefore, searching for epigenetic markers is also essential to finding out asthma endotypes, phenotypes, individualized treatments, and prevention.
Finally, over the past ten years, the interest in miRNAs has increased [26,38,64]. MiRNAs are short noncoding RNAs that behave as post-transcriptional negative regulators by inhibiting the translation of mRNA or initiating its degradation [65]. MiRNA levels in asthma have been correlated to the expression of the Th2 cytokines [66,67] IL-5 [68], IL-13 [69], as well as the tissue remodeling factor VEGF [70], which are key molecules in asthma pathogenesis [38]. Recent research has revealed that most miRNAs are involved in Th1 and Th2 cytokine secretion, anti-inflammatory responses, macrophage polarization, T cell differentiation towards T2 response, and bronchial smooth muscle cell hyperplasia and hypertrophy [71]. miRNA profiles can be correlated to clinical traits, such as lung function, phenotype, and asthma severity [72]. Moreover, miRNAs have been shown to be signaling molecules released by cells and transported in extracellular vesicles such as exosomes [73,74,75]. According to previous studies [76], asthma is primarily characterized by the upregulation of miR-21 [77,78,79], miR-223 [80], miR-146a [78], miR-142-5p [81], miR-142-3p [82], miR-146b [83], and miR-155 [84], and downregulation of the let-7 family [85], miR-193b [86], and miR-375 [87,88]. It has been shown that miR-155, miR-146a, miR-21, miR-1248, and miR-210 play key roles in controlling eosinophil and T cell activities as well as the release of Th2 cytokines (IL-4, IL-5, IL-13) [76]. However, for the use of miRNAs as biomarkers, they must be specific to each pathology, able to predict asthma phenotypes, and easy to detect in bodily fluids [38]. Therefore, clinical investigations may benefit from the expression of circulating miRNAs [36], because several of these miRNAs were found to distinguish asthmatic from non-asthmatic children [89] and adults [66,67,84,90,91,92,93], and rank asthma severity [94].

3.1.2. Asthma Treatment

Due to asthma heterogeneity, symptoms, and severity, the goal of asthma treatment is to ameliorate this pathology’s symptoms, avoiding future risks. Historically, asthma has been treated with anti-inflammatory drugs and bronchodilators [95]. The reliever treatment with inhaled corticosteroids (ICs)-formoterol reduces the risk of exacerbations as compared to the use of SABA. However, nowadays, new techniques have been developed, comprising immunotherapy and biologics [95,96] (Figure 3). In most cases, personalized asthma treatment is necessary, because each patient develops symptoms in a different way [97]. Therefore, there are two critical factors in the treatment of asthma: the patient’s symptoms and the way that the patient reacts to medication [98].
Currently, two categories of drugs are used for the treatment of asthma: rescue and controller drugs [15] (Figure 3). Rescue drugs are used to alleviate symptoms quickly (within minutes), whereas controller medicines are used daily to treat all asthma symptoms and to achieve asthma remission [15,99].
The main rescue drugs are inhaled, rapid-acting β2 adrenergic agonists (SABAs), such as albuterol and levalbuterol [100]. They are recommended for patients with mild or intermittent asthma [15]. SABAs bind to β-adrenergic receptors from the smooth muscle cells of the lung, causing the release of Ca2+ and, subsequently, muscle relaxation, mitigating asthma symptoms in a few minutes [101]. The use of these medicines is still limited because of their adverse effects: tremors, tachycardia, and hypocalcemia [100].
Severe asthma is defined as uncontrolled asthma despite optimized treatment with high-dose of ICs and long-acting β2 adrenergic agonists (LABAs), or requires this treatment to prevent exacerbations [15]. So, ICs are the first option as controller drugs, such as mometasone, budesonide, and ciclesonide [100]. These medications act as anti-inflammatory molecules, producing a long-term reduction in asthma symptoms [100,102]. However, ICs have negative side effects such as oral infections, hoarseness, reduction in growth velocity, osteoporosis, or hypothalamic–pituitary–adrenal axis suppression [103].
In addition to ICs, oral corticosteroids (OC) can be also used in asthma management [15]. They have more anti-inflammatory power than ICs, but also more adverse effects (loss of bone density, hyperglycemia, weight gain, cataracts, hypothalamic–pituitary–adrenal axis suppression, diabetes, sleep apnea, etc.) [104,105]. Nevertheless, an optimal concentration of OCs can be helpful in relieving symptoms of moderate–severe asthma [100].
Another type of drug employed in asthma control treatment is an LABA, such as salmeterol, formoterol, and vilanterol, having a similar mechanism of action to SABAs, blocking β-adrenergic receptors on the smooth muscle of the lung, and relaxing smooth muscle [100]. Although LABAs have a longer half-life than SABAs, they are considered controller drugs. LABAs are recommended to be used in combination with other corticosteroids [15].
On the other hand, other drugs used in the treatment of asthma are antileukotrienes, such as montelukast and zafirlukast. Leukotrienes are inflammatory molecules produced during the Th2 response by the degranulation of basophils, mast cells, and eosinophils. These molecules are responsible for airway inflammation and subsequent asthma symptoms [106,107]. Anti-leukotrienes are anti-inflammatory drugs that interfere with leukotrienes synthesis by inhibiting the 5-lipoxygenase activity, and they also act as cysteinyl leukotriene receptor 1 (CysLT1) inhibitors [108,109], blocking leukotrienes inflammatory activity. Several studies have found that these drugs have less anti-inflammatory capacity than the others mentioned above. Bleecker et al. demonstrated that inhaled fluticasone propionate was more effective than montelukast in improving some asthma exacerbations: FEV1 (0.42 L vs. 0.20 L over baseline, p < 0.001), morning PEF (49.94 L/min vs. 11.68 L/min over baseline, p < 0.001), and evening PEF (38.91 L/min vs. 10.50 L/min over baseline, p < 0.001) [110]. Thus, antileukotrienes are only used in the treatment of mild asthma [15]. However, anti-leukotrienes have several side effects such as elevated levels of transaminases, which indicates hepatocellular injury [109].
Finally, long-acting muscarinic antagonists (LAMAs) are used in the treatment of severe asthma, such as tiotropium, which is among the most used ones [15]. They are suppressors of acetylcholine, which acts as a bronchoconstrictor during asthmatic responses [111]. LAMAs inhibit the muscarinic receptors of the bronchioles (there are five muscarinic receptors, but only M1, M2, and M3 are enrolled in asthma exacerbations); thus, acetylcholine cannot bind to these receptors and muscle relaxation occurs [112].

3.2. Allergic Rhinitis

One of the most common comorbidities associated with asthma is AR, which also contributes to asthma severity [113]. AR is a heterogeneous disease that affects the upper airway and nose, and it is characterized by itching, sneezing, watery rhinorrhea, and nasal congestion [114]. AR also contains an allergic component characterized by an IgE-mediated response against specific allergens, producing inflammation by several mediators released by Th2 cells, eosinophils, and mast cells [115]. Although nasal allergen challenge (NAC) is the gold standard method to diagnose AR (with optimal sensitivity, specificity, and safety) [116], the development of novel diagnostic methods is needed, such as BAT [117] (Figure 2). Treatment options comprise ICs, antihistamines (AHs), allergen immunotherapy [114], and, during the last years, the study of treatment using biologic therapies in severe AR have increased exponentially [118,119,120,121] (Figure 3).
Symptoms begin after exposure to the sensitized allergen, which triggers within minutes rhinorrhea, sneezing, nasal itching, and nasal obstruction, which can also occur at a later time. AR can present ocular symptoms in up to 60–80% of cases, bronchial symptoms (75–80%) [122], and nasal polyposis (1.5%) [123].
AR is commonly classified based on the Allergic Rhinitis and its Impact on Asthma (ARIA) guidelines, which has a good clinical correlation with the visual analogue scale (VAS) [124]. According to its duration, it can be classified into intermittent/persistent (taking 4 days/week and/or 4 consecutive weeks as the cutoff point) and according to the impact on quality of life (impairment of activities of daily living, disturbance of night-time rest, disturbance of work/study, bothersome symptoms) into mild and moderate/severe.

3.2.1. Diagnosis of Allergic Rhinitis

Clinical Tools to Diagnose Allergic Rhinitis

The allergological study begins with a detailed clinical history [125] (symptoms, seasonality, relationship with triggers, associated symptoms, need for treatment, and response) as the diagnosis is fundamentally clinical. Complementary tests are necessary to study its etiology and phenotype, as well as a correct physical examination [126]. Physical examination by anterior rhinoscopy or nasal endoscopy provides information (mucosal coloration, turbinate morphology, or the presence of polyps) that may indicate the presence of other associated pathologies, although there are no pathognomonic signs.
To confirm the presence of allergen-specific IgE, intraepidermal SPT and/or detection of specific IgE in serum against allergens that are considered clinically relevant for the patient can be performed. Skin tests can diagnose two out of three allergic diseases and up to 90% of respiratory allergies [127]. Skin prick tests are considered to be more sensitive, fast, and cost-effective than serum IgE tests [128].
Circulating sIgE against whole allergens or components allows us to differentiate between genuine sensitization or cross-reactivity, which is essential information for the indication of treatments such as immunotherapy [129,130].
NAC can be performed by acoustic rhinometry, anterior or posterior rhinomanometry, or measurement of nasal peak inspiratory flow (NPIF). Although its use is becoming increasingly widespread, NAC is not routinely performed. NAC can be used to assess the clinical relevance in polysensitized patients [126] or when it is not possible to determine sIgE (serum and/or skin tests) [131]. In addition, NAC is currently the gold standard for the diagnosis of local allergic rhinitis (LAR), as it is a safe and reproducible technique [116].
The diagnosis is, therefore, clinical; therefore, it is important to differentiate between sensitization and allergy [132]. In addition, the infiltration of cells and inflammatory mediators can be studied by the detection of sIgE in nasal secretions, which is common in research, as well as BAT [133]. Once a diagnosis is reached, we will use methods to assess control: the VAS, considering uncontrolled rhinitis ≥5, and specific validated questionnaires, commonly used in research, such as the Rhinitis Control Assessment Test (RCAT) and Allergic Rhinitis Control Test (ARCT), or the more recently published ARIA-c, with the same parameters used for the staging of severity.

Cellular Diagnostic of Allergic Rhinitis

Similar to asthma, AR is characterized by an inflammation produced by the T2 immune response, which involves: Th2 cells, IgE-producing B cells, basophils, mast cells, eosinophils, and a plethora of Th2 cytokines (IL-4, IL-5, IL-9, IL-13, IL-25, IL-31, IL-33, and TSLP) [134,135,136,137,138,139,140]. Patients with AR are characterized by a pattern with a cellular infiltrate that induces a nasal production of mediators such as tryptase, eosinophilic cationic protein (ECP) [141,142], and sIgE [143]. In this context, a study performed by Chen and colleagues concluded that the levels of activated and pathogenic eosinophils, which are associated with higher production of ECP, eosinophil peroxidase (EPX), and IL-4 in the peripheral blood were elevated in patients with moderate–severe AR in comparison with mild patients and healthy controls [144]. In addition, several chemokines, such as the eotaxin family, which recruit and activate Th2 lymphocytes, mast cells, and eosinophils, seem important in allergic diseases as well [145]. In this regard, for example, high levels of eotaxin-1 (CCL11) were obtained after NAC in patients with AR in comparison with controls [146].
On the other hand, ILC2 residing at mucosal and barrier surfaces can act as effector immune cells. ILC2 are associated with allergic disorders, including AR [147,148], and they are functionally like T cells (but lack antigen receptors). Allergic phenotypes of rhinitis are determined by measuring allergen-sIgE in serum and BAT [149]. BAT reproduces in vitro, after allergen exposure, the type I hypersensitivity reaction [150]; an immediate reaction that involves IgE-mediated release of antibodies against the soluble antigen [151]. For LAR diagnosis, BAT exhibits a high specificity (100%), in contrast to its sensibility (ranging from 50% to 66.6%) depending on the allergen evaluated [117,152,153]. In this regard, 37.5% of LAR individuals (all of them with NAC positive to Dermatophagoides pteronyssinus) and 60% of dual allergic rhinitis (DAR) patients showed positive BAT responses with perennial allergens, as opposed to NAR and healthy subjects (negative for all of them) [154].

Molecular Diagnostic Parameters in Allergic Rhinitis

In the last few decades, several studies have focused on molecular parameters associated with inflammation in AR, such as chemokines associated with Th2 function [8]. In this context, high levels of CCL22 (monocyte-derived chemokine (MDC)), which promote selective migration of Th2 cells, were found in the serum of patients with AR sensitized to birch pollen [155] and ragweed pollen [156], suggesting a possible role in the pathogenesis of AR. Another chemokine that has been associated with AR is CCL13, whose expression is stimulated by IL-4 and was found to be increased in the serum of AR patients after NAC [157].
On the other hand, miRNAs are thought to be involved in the pathogenesis of AR [40,158,159]. MiR-155 levels in serum of children with pollen-induced AR were higher in comparison with healthy controls, and miR-155 in the serum correlated significantly with nasal symptoms in children with AR (r = 0.494, p < 0.001) [160]. Moreover, Luo and colleagues found serum TSLP, expression of miR-375 from whole blood, and frequencies of ILC2 in peripheral blood levels significantly higher in AR children compared with controls [140]. Moreover, another study showed that the level expression of miR-487b was repressed in AR in comparison to control cases [161]. Finally, Teng et al. found that the expression of miR-143 was significantly decreased in nasal mucosal tissues from AR patients compared with tissues from NAR subjects [162].
Additionally, Th1/Th17 were also proposed to be involved in allergic diseases, such as AR [163]. In this context, Erkan et al. [143] showed that serum and nasal IL-17 were higher in AR in comparison with control individuals. Moreover, Lee and collaborators showed that serum levels of IL-8 were significantly higher in patients with allergic asthma in comparison with AR and controls, suggesting that IL-8 is associated with a more severe inflammatory response [164]. However, other studies showed that elevated levels of IL-8 can also be related to pollution and not only allergic sensitization, suggesting that air pollution might induce or aggravate AR through this cytokine [165]. Other studies, such as Yu et al., showed a decreased surface CXC motif chemokine receptor 3 (CXCR3) expression in CD4+ T cells of AR patients [166].
In the case of the innate immune response, a study performed by Kant (which excluded individuals with an active infection), with 205 AR patients and 49 healthy controls, found that the neutrophil/lymphocyte ratio was significantly lower in patients with AR than in healthy controls [167]. In relation to ILC2, several studies showed that these cells in peripheral blood and nasal samples are increased after NAC, and there was also a positive correlation between eosinophils and IL-5 concentrations in patients with AR [168,169]. In this context, several studies showed evidence of increased epithelial proinflammatory cytokines, such as IL-25, IL-33, and TSLP in the nasal lavage from patients with house dust mite (HDM) sensitivity [170,171]. Other studies have shown that patients with AR displayed high levels of IL-33 and TSLP mRNA in the nasal epithelium [172,173,174].

3.2.2. Treatment of Allergic Rhinitis

AR management includes patient education, allergen avoidance, pharmacotherapy, immunotherapy, and biologics. It is essential to explain to the patient about their disease and how to take their treatments. The first step is to avoid exposure to the allergen. However, if the symptoms persist, the first line of treatment is drug therapy. Immunotherapy is recommended when the disease is not controlled with the usual drugs. In addition, in recent years, biologics have emerged as a novel therapeutic option with promising results [175,176,177,178] (Figure 3).
Typical AR drugs include AHs, GCs, and leukotriene receptor antagonists. Second-generation oral antihistamines are the first line of pharmacological treatment, as well as intranasal corticosteroids (INCs), which have demonstrated even greater efficacy than AHs. [179]. Intranasal combination therapies with AHs and GCs, such as azelastine hydrochloride/fluticasone propionate (AZE/FP), are also recommended [180].
Antihistamines bind to the histamine H1 receptor and block its action. First-generation of oral antihistamines (OAHs), such as diphenhydramine or chlorpheniramine, have been widely used in the clinic. However, due to their adverse effects, they are no longer supported for AR [181]. Their main secondary effect is sedation as they cross the blood-brain barrier causing drowsiness and fatigue, among other symptoms [182]. Therefore, the use of new-generation AHs (such as desloratadine, loratadine, cetirizine, levocetirizine and rupatadine, fexofenadine, and bilastine) is strongly recommended. These have been shown to be safer than the previous ones, maintaining the same efficacy without the sedative effect [181,183].
Intranasal antihistamines (INAHs) improve the effect of oral antihistamines at the nasal mucosa [184,185]. Moreover, they are more effective in controlling local symptoms, such as nasal congestion [186,187]. In addition, they act faster than OAHs and reduce potentially systemic effects [187]. They are recommended as first-line treatment for seasonal AR (SAR) [122]. The two INAHs approved for the management of SAR are azelastine and olopatadine. Both have demonstrated similar efficacy [188]. They differ in that azelastine inhibits both H1 and H2 receptors, while olopatadine only inhibits H1.
INCs (beclomethasone, budesonide, ciclesonide, fluticasone propionate, fluticasone furoate, mometasone furoate, and triamcinolone acetonide) have a local anti-inflammatory effect by preventing the recruitment of immune cells into the nasal mucosa. They are indicated as first-line treatment in patients with moderate or persistent symptoms [180]. In addition, they have the advantage of having no systemic adverse effects [189,190]. INCs have demonstrated efficacy in controlling the main symptoms of AR: nasal congestion, itching, rhinorrhea, and sneezing [191,192]. INCs have been described to be more effective than AHs for nasal congestion [179].
OCs are not used as a routine treatment for rhinitis, because their adverse effects exceed the potential benefits [193]. A short course, only for a few days, with oral corticosteroids, can be indicated in patients with severe symptoms that do not respond to other drugs [194]
A novel formulation combines azelastine hydrochloride (AZE) and fluticasone propionate (FP) in a single nasal spray. This combined therapy has proved to be faster [195] and more effective than both drugs taken individually. [196,197]. One of the advantages is that it increases adherence to treatment by administering both drugs at the same time. In addition, the drugs are more homogeneously distributed in the nasal mucosa than if AZE and FP sprays were used sequentially [198]. It is recommended for the initial treatment of moderate to severe nasal symptoms of SAR [180], as well as for patients with both seasonal and perennial AR who do not respond to monotherapy [122].
LTRAs act by blocking the activity of cysteinyl leukotrienes, an inflammatory mediator associated with the main symptoms of AR, such as nasal congestion and mucus production [199]. The main LTRAs are montelukast and zafirlukast. They should only be used when the patient does not respond to any other drug. This is because they are equally or less effective than INAH, INCs, or OAH, [200,201] and have also been associated with serious neuropsychiatric adverse effects. In fact, in Europe, they are only approved for the treatment of patients with asthma and rhinitis comorbidity [202].

3.3. Biologicals in Asthma and Allergic Rhinitis

Due to the diversity of adverse effects induced by classical drugs, new approaches for the treatment of asthma have been sought. Among these new therapies for asthma and rhinitis are biologicals (Figure 4). Biologicals are humanized monoclonal antibodies that target several molecules responsible for the T2 response, inhibiting it and ameliorating asthma symptoms. The main targets of biologics for asthma treatment are IgE (omalizumab), IL-5 (mepolizumab and reslizumab), the IL-5 receptor (benralizumab), the IL-4/IL-13 receptor (dupilumab), and TSLP (tezepelumab) [203] (Figure 4), although novel biologicals against other targets are being developed. Although these drugs have been studied and approved for asthma treatment, only the use of omalizumab and dupilumab in rhinitis have also been extensively studied [204,205]; however, to date, none have been approved by drug agencies for the treatment of AR.

3.3.1. Omalizumab (Anti-IgE)

It was the first biological approved for the treatment of asthma by the Food and Drug Administration (FDA) in 2003 [206]. Nowadays, it is used in patients aged 6 years and older with moderate–severe asthma, and it is recommended its subcutaneous administration every 2 or 4 weeks depending on IgE serum levels and body weight [15].
Several studies have demonstrated the efficacy of omalizumab in the treatment of severe asthma [207]. The EXTRA study (NCT00314574) enrolled 850 patients with uncontrolled severe allergic asthma [208]. Asthma biomarkers: FeNO, blood eosinophils, and serum periostin showed a normalization in the patients that received omalizumab in comparison to the placebo group. The PROSPERO study (NCT01922037) demonstrated an improvement in asthma symptoms in 87% of the patients after receiving omalizumab for 48 weeks [209].
Recently, two meta-analyses have been performed, evaluating several trials testing the use of omalizumab in AR [176,177]. In one meta-analysis, Yu et al. included 16 randomized clinical trials, while Tsabouri et al. developed a systematic review of 12 trials. Both studies reached almost the same conclusions: treatment with omalizumab significantly improves both ocular and nasal symptoms, reduces the use of rescue medication, improves the quality of life, and, in addition, no increase in adverse effects was observed compared to the placebo group.

3.3.2. Mepolizumab and Reslizumab (Anti-IL-5)

In 2015 the FDA authorized mepolizumab [210] for patients aged 6 years and older with severe eosinophilic asthma. The recommended dose by subcutaneous injection every 4 weeks depends on the patient’s age [15].
Mepolizumab effectiveness was evaluated for the treatment of severe eosinophilic asthma in the MENSA study (NCT01691521) [211]. Mepolizumab reduced asthma exacerbations by 47% in patients who received intravenous doses and by 53% in those receiving subcutaneous doses as compared with the group receiving placebo. Recently, the COSMEX study (NCT02135692) showed the long-term efficacy of mepolizumab [212]. This Phase IIIb clinical trial involved 340 patients with eosinophilic asthma who had previously participated in the SIRIUS (NCT01691508), MENSA (NCT01691521), and COSMOS (NCT01842607) [211,213,214]. In the COSMEX study, patients received mepolizumab subcutaneously every 4 weeks for at least 188 weeks, showing a fivefold reduction in the rate of exacerbations per year [212].
On the other hand, reslizumab was approved by FDA one year after mepolizumab [215]. Its mechanism of action is the same as mepolizumab, but reslizumab has weight-based dosing, so it is useful for patients with high body mass index [216]. It is authorized for patients aged 18 years and older with severe eosinophilic asthma, recommended to administer 3 mg/kg by intravenous infusion every 4 weeks [15]. In 2016, Corren et al. showed the efficacy of reslizumab in a randomized, double-blind Phase III trial (NCT01508936) [217]. In this study, the administration of reslizumab once a month for 16 weeks in 492 patients with asthma exacerbations demonstrated an improvement in exacerbations, especially in those patients with baseline eosinophils ≥ 400 cells/µL.

3.3.3. Benralizumab (Anti-IL-5 Receptor)

At the end of 2017, benralizumab was approved [218]. It is administered to patients over 12 years with severe eosinophilic asthma by subcutaneous injection every 4 weeks for the first three doses and then every 8 weeks [15]. The CALIMA study (NCT01914757) showed the efficacy of benralizumab in the treatment of severe eosinophilic asthma [219]. This randomized Phase III clinical trial performed by FitzGerald et al. enrolled 1306 patients with asthma symptoms. In this study, the annual exacerbation rate in the patients who received benralizumab was diminished by 36% in those patients who received the treatment every 4 weeks and by 28% in those who did it every 8 weeks. Similar results were found in SIROCCO study [220], corroborating the efficacy of benralizumab in the treatment of severe eosinophilic asthma.

3.3.4. Dupilumab (Anti-IL-4/13 Receptor)

Dupilumab was approved by the FDA in 2017 [221]. It is recommended for patients aged 6 years and older who present severe eosinophilic asthma, but also for the treatment of moderate to severe atopic dermatitis, moderate to severe asthma, and chronic rhinosinusitis with nasal polyps (CRSwNP) [222]. It is administered by subcutaneous injection every 2 weeks in patients older than 12 years. For children between 6–11 years of age, the dose depends on weight [15].
Several studies have demonstrated the efficacy of dupilumab in the treatment of asthma and chronic rhinosinusitis [223]. In 2018, Castro et al. carried out a randomized, double-blind trial (NCT02414854) involving 1902 patients aged 12 years and older with uncontrolled asthma [224]. Participants received dupilumab 200 or 300 mg or placebo every 2 weeks for 52 weeks. Results showed that severe asthma exacerbations were reduced by 47.7% among patients receiving dupilumab as compared with individuals that received a placebo. Moreover, dupilumab has been shown to be more effective than OCs in the treatment of severe asthma [225]. Rabe and collaborators carried out a randomized phase III clinical trial (NCT02528214) in which 210 patients with OC-treated asthma were recruited [225]. They received dupilumab (300 mg) or a placebo every 2 weeks for 24 weeks. During the study, the glucocorticoid doses were reduced each week, and at the end of the study, the decrease in the dose was 70.1% for the dupilumab group and 41.9% for the placebo group. Despite dose reduction, dupilumab treatment showed a 59% lower exacerbation rate as compared with the placebo group. Nevertheless, this biological presents important adverse effects like blood eosinophilia, dry eyes, keratitis, conjunctivitis, head and neck dermatitis, and arthritis [226].
Regarding the use of dupilumab in AR, Weinstein et al. recently reviewed a randomized, double-blind Phase IIb clinical trial (NCT01854047) in patients with asthma and perennial AR (PAR) [227]. This trial evaluates the efficacy of dupilumab at different doses: 200 mg or 300 mg subcutaneously every 2 or 4 weeks; versus placebo. There was evidence that 300 mg of dupilumab every 2 weeks in combination with intranasal corticosteroids and β2 agonists, significantly reduced nasal symptoms in patients with asthma and comorbidity with PAR [178]. Although dupilumab has demonstrated several benefits in the treatment of AR, as well as omalizumab, further studies are needed to corroborate their cost-effectiveness against current therapies.

3.3.5. Novel Biologic Therapies

Nowadays, other biologicals are being studied with the aim of treating non-allergic asthma.
Tezepelumab is an IgG2 monoclonal antibody that targets TSLP, which is a protein produced by epithelial cells in response to both allergens, viruses and other toxins involved in the triggering of asthma. This biological has recently been approved to treat severe asthma in patients aged 12 years and older [228], supported by previous clinical trials that have shown promising results in the treatment of allergic and non-allergic asthma with this biological [229]. In this randomized, double-blind, placebo-controlled trial, 1061 patients were randomized to receive tezepelumab or placebo subcutaneously for 52 weeks, once a month. Results showed that patients that received tezepelumab had lower rates of asthma exacerbations, lower blood eosinophil count, and an improvement in lung function parameters.
On the other hand, another biological still under development is astegolimab, a monoclonal IgG2 antibody that binds to the IL-33 receptor (anti-ST2) [230]. The ZENYATTA study comprised 502 severe asthmatic individuals distributed into several groups: patients that received placebo or 70 mg, 210 mg, or 490 mg doses of astegolimab subcutaneously every 4 weeks [230]. Patients that received the highest and the lowest dose of astegolimab showed higher rates of reduction of asthma exacerbations in both types of patients with uncontrolled asthma: eosinophil—high (≥300 cells/μL) and eosinophil—low (<300 cells/μL).

3.4. Allergen-Specific Immunotherapy

Classical treatments for asthma have been used to relieve the symptoms caused by the inflammatory process. Nevertheless, these do not solve the underlying problem: the disproportionate response of the immune system. In addition, classical therapies have various long-term side effects, and it is difficult to find an optimal treatment for each patient, because of asthma heterogeneity. Therefore, immunotherapy has been postulated as an effective strategy offering long-term tolerance [231]; additionally, it is less expensive than other treatments commented on previously, like biologicals [96,232,233].
Immunotherapy involves the administration of increasing amounts of the allergen until an adequate dose to produce immunological tolerance is achieved [128]. Repeated exposure to the allergen causes the DCs to produce IL-10, IL-12, and IL-27, which are regulatory cytokines. They stimulate Th1 response, thus restoring the balance between the T1 and T2 response. In addition, IL-10, IL-12, and IL-27 also activate Treg and B cells, favoring the production of antibodies that will block future T2 responses, such as IgG4 [234]. There is also a modification of the activation threshold of mast cells and basophils [202].
It has been proven that AIT has long-term benefits for asthma and AR [235,236,237,238]. Its effect persists even years after ending the treatment, with a consequent reduction in medication use as well as a decrease in the risk of developing asthma [88]. Despite its more than proven efficacy, immunotherapy is mainly recommended when patients do not control their symptoms with conventional pharmacotherapy or if they do not tolerate these drugs [142,239]. Different types of allergens can be used in immunotherapy: crude allergen extracts, purified or recombinant allergens, modified allergens (allergoids), and purified peptides [240]. Allergen administration is usually subcutaneous, but sublingual immunotherapy (SLIT) is also available, in addition to a less common route of intralymphatic immunotherapy (ILIT). The main adverse effect of immunotherapy is the development of uncontrolled allergic responses [234].
On the one hand, SCIT is based on the periodical administration of increasing amounts of the allergen until immunological tolerance is achieved. SCIT is effective against several allergens, such as HDM, birch pollen, timothy grass, and ryegrass [241,242,243]. However, SCIT has some drawbacks, such as the need for frequent injections; moreover, these must be administered in a hospital [239], in addition to the potential risk of anaphylaxis [244]. In several studies, AIT has been demonstrated to be more effective than conventional drugs in controlling AR symptoms. A recent randomized clinical trial compared a group of patients receiving AIT plus pharmacotherapy versus a control group receiving only drug treatment. The results showed a significant decrease in symptoms in the immunotherapy-treated group, as well as an improvement in quality of life, compared to the control group [245]. Rondon and collaborators developed another randomized, double-blind placebo-controlled trial with Phleum pratense SCIT performed in 56 patients with moderate–severe LAR to grass pollen demonstrating that SCIT increased allergen tolerance [246]. Most patients treated with SCIT for more than 6 months showed an allergen tolerance over 50 times higher than baseline, and 56% showed a negative nasal allergen provocation test. Moreover, these authors demonstrated that SCIT was safe for patients not having serious adverse events related to the medication.
On the other hand, SLIT offers multiple benefits over the subcutaneous route as it does not require injections, can be administered at home, and has a lower risk of systemic adverse effects [247]. However, it must be administered daily, which may be a problem for adherence to treatment [248]. Previous studies have demonstrated the efficacy of SLIT in asthma treatment, although it has not been officially approved [15]. In a randomized trial, Virchow et al. showed that SLIT may reduce the dose of ICs needed in HDM allergic asthma [249]. The study enrolled 693 patients with no controlled asthma related to HDM who received placebo or HDM-SLIT tablets in addition to ICs and salbutamol. Results showed that HDM immunotherapy decreased asthma exacerbations during ICs reduction as compared with the placebo. Moreover, immunotherapy may also reduce the risk of suffering from asthma in patients with other allergic symptoms. In this context, Valovirta et al. carried out a randomized clinical trial involving 812 children with grass pollen allergies who had no signs of asthma [250]. Results showed that the treatment with grass pollen SLIT decreased the risk of suffering asthma symptoms at the end of the study (3 years) and in the following 2 years. Regarding AR treatment, both SLIT and SCIT have similar efficacy. A systematic review compared both immunotherapies against placebo, and the two were more effective, with a similar quality of life score [251]. A recent meta-analysis evaluated multiple clinical trials also comparing SCIT versus SLIT. It concluded that SCIT was slightly superior in improving AR symptoms, however, the differences between the two groups were not significant [252].
Finally, ILIT consists of injecting the allergen directly into the lymph nodes. The main advantage of this treatment is that it considerably reduces immunotherapy time, lasting only a few months, and reduces the amount of allergen used [87], but this AIT has been poorly studied in asthma [253]. A recent meta-analysis evaluated 17 clinical trials comparing ILIT against placebo. ILIT requires only three injections, spaced one month apart. This meta-analysis concluded that ILIT is safe and reduces both symptoms and medication use in patients with AR [254]. Another randomized, double-blind clinical trial evaluated the use of ILIT in patients with grass pollen rhinoconjunctivitis. Patients were also treated with 3 ILIT injections and the results show a significant reduction in allergy symptoms [255]. Although ILIT appears to be a promising new therapy for AR, further clinical trials are needed to demonstrate its efficacy against standard immunotherapies.

4. Conclusions

Asthma and AR are two common respiratory diseases that produce high socioeconomic and health problems; however, because of the heterogeneity of these pathologies, a search for precise and personalized medicine in these patients is mandatory. Moreover, the evolution of research on these pathologies has changed our understanding of cellular and molecular mechanisms; thus, treatments need to evolve. Although classic and emerging therapies for asthma and AR have improved the quality of life of patients, more efforts need to be performed to improve diagnosis and treatment. In this sense, a search of biomarkers for each phenotype and endotype is needed to choose the optimal treatment for each patient. However, currently, a lack of specific biomarkers of diagnosis often means that the administered treatment is not the right one. Moreover, a lack of biomarkers to predict immunotherapy efficacy courses with long and costly ineffective treatments for patients. In addition, the efficacy of immunotherapy is not clear yet, so the search for these biomarkers is crucial. However, promising drugs such as biologicals have been developed to improve the quality of treatments and the quality of life of patients. In conclusion, although nowadays there are several therapeutic and diagnostic tools for asthma and AR, further studies are needed to develop more precise and effective methods towards a more specific and personalized medicine.

Author Contributions

Conceptualization, C.J.A. and J.A.C.; investigation, M.E.-S., R.S.d.S.M., M.d.C.M.-A., C.L.-M. and M.J.D.; data curation, C.J.A. and J.A.C.; writing—original draft preparation, M.E.-S., R.S.d.S.M., M.d.C.M.-A., C.L.-M., M.J.D., I.E.-G., C.R., C.M., M.J.T., C.J.A. and J.A.C.; writing—review and editing, M.E-S., R.S.d.S.M., I.E.-G., C.R., C.M., M.J.T., C.J.A. and J.A.C.; supervision, C.M., M.J.T., C.J.A. and J.A.C.; funding acquisition, I.E.-G., C.R., C.M., M.J.T., C.J.A. and J.A.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by grants from the Institute of Health “Carlos III” (ISCIII) of the Ministry of Economy and Competitiveness: PI20/01715; P20-00405 (PAIDI Consejería de Transformación Económica, Industria, Conocimiento y Universidades); RETICS ARADyAL (RD16/0006/0001); Predoctoral Health Research Training Funding (FI21/00066); and Sara Borrell (CD20/00085, CD21/00034) Program. Andalusian Regional Ministry of Health, Nicolas Monardes Program (RC-0004-2021). Grants were co-funded by the European Regional Development Fund (ERDF). “Una manera de hacer Europa” “Andalucía se mueve con Europa”.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We thank Claudia Corazza for her help with the English version of the manuscript.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. GBD Chronic Respiratory Disease Collaborators. Prevalence and attributable health burden of chronic respiratory diseases, 1990–2017: A systematic analysis for the Global Burden of Disease Study 2017. Lancet Respir. Med. 2020, 8, 585–596. [Google Scholar] [CrossRef]
  2. Eguiluz-Gracia, I.; Mathioudakis, A.G.; Bartel, S.; Vijverberg, S.J.H.; Fuertes, E.; Comberiati, P.; Cai, Y.S.; Tomazic, P.V.; Diamant, Z.; Vestbo, J.; et al. The need for clean air: The way air pollution and climate change affect allergic rhinitis and asthma. Allergy 2020, 75, 2170–2184. [Google Scholar] [CrossRef] [PubMed]
  3. Labaki, W.W.; Han, M.K. Chronic respiratory diseases: A global view. Lancet Respir. Med. 2020, 8, 531–533. [Google Scholar] [CrossRef]
  4. He, Z.F.; Zhong, N.S.; Guan, W.J. Impact of Chronic Respiratory Diseases on the Outcomes of COVID-19. Arch. Bronconeumol. 2022, 58, 5–7. [Google Scholar] [CrossRef]
  5. Reddel, H.K.; Bacharier, L.B.; Bateman, E.D.; Brightling, C.E.; Brusselle, G.G.; Buhl, R.; Cruz, A.A.; Duijts, L.; Drazen, J.M.; FitzGerald, J.M.; et al. Global Initiative for Asthma Strategy 2021: Executive summary and rationale for key changes. Eur. Respir. J. 2022, 59, 2102730. [Google Scholar] [CrossRef] [PubMed]
  6. World Health Organization. Available online: https://www.who.int/news-room/fact-sheets/detail/asthma (accessed on 20 October 2022).
  7. Kuruvilla, M.E.; Lee, F.E.; Lee, G.B. Understanding Asthma Phenotypes, Endotypes, and Mechanisms of Disease. Clin. Rev. Allergy Immunol. 2019, 56, 219–233. [Google Scholar] [CrossRef] [PubMed]
  8. Agache, I.; Eguiluz-Gracia, I.; Cojanu, C.; Laculiceanu, A.; Del Giacco, S.; Zemelka-Wiacek, M.; Kosowska, A.; Akdis, C.A.; Jutel, M. Advances and highlights in asthma in 2021. Allergy 2021, 76, 3390–3407. [Google Scholar] [CrossRef]
  9. Oliveria, J.P.; Agayby, R.; Gauvreau, G.M. Regulatory and IgE(+) B Cells in Allergic Asthma. Methods Mol. Biol. 2021, 2270, 375–418. [Google Scholar] [CrossRef]
  10. Ricciardolo, F.L.M.; Sprio, A.E.; Baroso, A.; Gallo, F.; Riccardi, E.; Bertolini, F.; Carriero, V.; Arrigo, E.; Ciprandi, G. Characterization of T2-Low and T2-High Asthma Phenotypes in Real-Life. Biomedicines 2021, 9, 1684. [Google Scholar] [CrossRef] [PubMed]
  11. Sze, E.; Bhalla, A.; Nair, P. Mechanisms and therapeutic strategies for non-T2 asthma. Allergy 2020, 75, 311–325. [Google Scholar] [CrossRef]
  12. Hudey, S.N.; Ledford, D.K.; Cardet, J.C. Mechanisms of non-type 2 asthma. Curr. Opin. Immunol. 2020, 66, 123–128. [Google Scholar] [CrossRef]
  13. Wang, R.; Murray, C.S.; Fowler, S.J.; Simpson, A.; Durrington, H.J. Asthma diagnosis: Into the fourth dimension. Thorax 2021, 76, 624–631. [Google Scholar] [CrossRef] [PubMed]
  14. Guilleminault, L.; Ouksel, H.; Belleguic, C.; Le Guen, Y.; Germaud, P.; Desfleurs, E.; Leroyer, C.; Magnan, A. Personalised medicine in asthma: From curative to preventive medicine. Eur. Respir. Rev. 2017, 26, 160010. [Google Scholar] [CrossRef] [PubMed]
  15. Asthma, G.I.f. Global Strategy for Asthma Management and Prevention. Available online: www.ginasthma.org (accessed on 16 November 2022).
  16. Bernstein, I.L.; Li, J.T.; Bernstein, D.I.; Hamilton, R.; Spector, S.L.; Tan, R.; Sicherer, S.; Golden, D.B.; Khan, D.A.; Nicklas, R.A.; et al. Allergy diagnostic testing: An updated practice parameter. Ann. Allergy Asthma Immunol. 2008, 100, S1–S148. [Google Scholar] [CrossRef] [PubMed]
  17. Coates, A.L.; Wanger, J.; Cockcroft, D.W.; Culver, B.H.; Carlsen, K.-H.; Diamant, Z.; Gauvreau, G.; Hall, G.L.; Hallstrand, T.S.; Horvath, I.; et al. ERS technical standard on bronchial challenge testing: General considerations and performance of methacholine challenge tests. Eur. Respir. J. 2017, 49, 1601526. [Google Scholar] [CrossRef] [Green Version]
  18. Parsons, J.P.; Hallstrand, T.S.; Mastronarde, J.G.; Kaminsky, D.A.; Rundell, K.W.; Hull, J.H.; Storms, W.W.; Weiler, J.M.; Cheek, F.M.; Wilson, K.C.; et al. An official American Thoracic Society clinical practice guideline: Exercise-induced bronchoconstriction. Am. J. Respir. Crit. Care Med. 2013, 187, 1016–1027. [Google Scholar] [CrossRef]
  19. Saito, J.; Inoue, K.; Sugawara, A.; Yoshikawa, M.; Watanabe, K.; Ishida, T.; Ohtsuka, Y.; Munakata, M. Exhaled nitric oxide as a marker of airway inflammation for an epidemiologic study in schoolchildren. J. Allergy Clin. Immunol. 2004, 114, 512–516. [Google Scholar] [CrossRef]
  20. Reddel, H.K. Peak flow monitoring in clinical practice and clinical asthma trials. Curr. Opin. Pulm. Med. 2006, 12, 75–81. [Google Scholar] [CrossRef]
  21. James, B.N.; Oyeniran, C.; Sturgill, J.L.; Newton, J.; Martin, R.K.; Bieberich, E.; Weigel, C.; Maczis, M.A.; Palladino, E.N.D.; Lownik, J.C.; et al. Ceramide in apoptosis and oxidative stress in allergic inflammation and asthma. J. Allergy Clin. Immunol. 2021, 147, 1936–1948. [Google Scholar] [CrossRef]
  22. Spanish Asthma Management Guidelines (GEMA 5.0 2022). Available online: https://www.gemasma.com (accessed on 11 January 2023).
  23. Fahy, J.V. Type 2 inflammation in asthma--present in most, absent in many. Nat. Rev. Immunol. 2015, 15, 57–65. [Google Scholar] [CrossRef]
  24. Breiteneder, H.; Peng, Y.Q.; Agache, I.; Diamant, Z.; Eiwegger, T.; Fokkens, W.J.; Traidl-Hoffmann, C.; Nadeau, K.; O’Hehir, R.E.; O’Mahony, L.; et al. Biomarkers for diagnosis and prediction of therapy responses in allergic diseases and asthma. Allergy 2020, 75, 3039–3068. [Google Scholar] [CrossRef] [PubMed]
  25. Agache, I.; Akdis, C.A. Precision medicine and phenotypes, endotypes, genotypes, regiotypes, and theratypes of allergic diseases. J. Clin. Investig. 2019, 129, 1493–1503. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Ogulur, I.; Pat, Y.; Ardicli, O.; Barletta, E.; Cevhertas, L.; Fernandez-Santamaria, R.; Huang, M.; Bel Imam, M.; Koch, J.; Ma, S.; et al. Advances and highlights in biomarkers of allergic diseases. Allergy 2021, 76, 3659–3686. [Google Scholar] [CrossRef] [PubMed]
  27. Li, S.; Morita, H.; Sokolowska, M.; Tan, G.; Boonpiyathad, T.; Opitz, L.; Orimo, K.; Archer, S.K.; Jansen, K.; Tang, M.L.K.; et al. Gene expression signatures of circulating human type 1, 2, and 3 innate lymphoid cells. J. Allergy Clin. Immunol. 2019, 143, 2321–2325. [Google Scholar] [CrossRef] [Green Version]
  28. Matsumoto, H. Roles of Periostin in Asthma. Adv. Exp. Med. Biol. 2019, 1132, 145–159. [Google Scholar] [CrossRef] [PubMed]
  29. Izuhara, K.; Ohta, S.; Ono, J. Using Periostin as a Biomarker in the Treatment of Asthma. Allergy Asthma Immunol. Res. 2016, 8, 491–498. [Google Scholar] [CrossRef] [Green Version]
  30. Santos, A.F.; Alpan, O.; Hoffmann, H.J. Basophil activation test: Mechanisms and considerations for use in clinical trials and clinical practice. Allergy 2021, 76, 2420–2432. [Google Scholar] [CrossRef]
  31. Dahlen, B.; Nopp, A.; Johansson, S.G.; Eduards, M.; Skedinger, M.; Adedoyin, J. Basophil allergen threshold sensitivity, CD-sens, is a measure of allergen sensitivity in asthma. Clin. Exp. Allergy 2011, 41, 1091–1097. [Google Scholar] [CrossRef]
  32. Diamant, Z.; Vijverberg, S.; Alving, K.; Bakirtas, A.; Bjermer, L.; Custovic, A.; Dahlen, S.E.; Gaga, M.; Gerth van Wijk, R.; Giacco, S.D.; et al. Toward clinically applicable biomarkers for asthma: An EAACI position paper. Allergy 2019, 74, 1835–1851. [Google Scholar] [CrossRef] [Green Version]
  33. Perkins, T.N.; Donnell, M.L.; Oury, T.D. The axis of the receptor for advanced glycation endproducts in asthma and allergic airway disease. Allergy 2021, 76, 1350–1366. [Google Scholar] [CrossRef]
  34. McDowell, P.J.; Heaney, L.G. Different endotypes and phenotypes drive the heterogeneity in severe asthma. Allergy 2020, 75, 302–310. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Alving, K.; Diamant, Z.; Lucas, S.; Magnussen, H.; Pavord, I.D.; Piacentini, G.; Price, D.; Roche, N.; Sastre, J.; Thomas, M.; et al. Point-of-care biomarkers in asthma management: Time to move forward. Allergy 2020, 75, 995–997. [Google Scholar] [CrossRef] [PubMed]
  36. Wasti, B.; Liu, S.K.; Xiang, X.D. Role of Epigenetics in the Pathogenesis, Treatment, Prediction, and Cellular Transformation of Asthma. Mediat. Inflamm. 2021, 2021, 9412929. [Google Scholar] [CrossRef] [PubMed]
  37. Koenderman, L.; Hassani, M.; Mukherjee, M.; Nair, P. Monitoring eosinophils to guide therapy with biologics in asthma: Does the compartment matter? Allergy 2021, 76, 1294–1297. [Google Scholar] [CrossRef] [PubMed]
  38. Weidner, J.; Bartel, S.; Kilic, A.; Zissler, U.M.; Renz, H.; Schwarze, J.; Schmidt-Weber, C.B.; Maes, T.; Rebane, A.; Krauss-Etschmann, S.; et al. Spotlight on microRNAs in allergy and asthma. Allergy 2021, 76, 1661–1678. [Google Scholar] [CrossRef] [PubMed]
  39. Pua, H.H.; Ansel, K.M. MicroRNA regulation of allergic inflammation and asthma. Curr. Opin. Immunol. 2015, 36, 101–108. [Google Scholar] [CrossRef] [Green Version]
  40. Panganiban, R.P.; Wang, Y.; Howrylak, J.; Chinchilli, V.M.; Craig, T.J.; August, A.; Ishmael, F.T. Circulating microRNAs as biomarkers in patients with allergic rhinitis and asthma. J. Allergy Clin. Immunol. 2016, 137, 1423–1432. [Google Scholar] [CrossRef] [Green Version]
  41. Kho, A.T.; Sharma, S.; Davis, J.S.; Spina, J.; Howard, D.; McEnroy, K.; Moore, K.; Sylvia, J.; Qiu, W.; Weiss, S.T.; et al. Circulating MicroRNAs: Association with Lung Function in Asthma. PLoS ONE 2016, 11, e0157998. [Google Scholar] [CrossRef] [Green Version]
  42. Kho, A.T.; McGeachie, M.J.; Moore, K.G.; Sylvia, J.M.; Weiss, S.T.; Tantisira, K.G. Circulating microRNAs and prediction of asthma exacerbation in childhood asthma. Respir. Res. 2018, 19, 128. [Google Scholar] [CrossRef] [Green Version]
  43. Davis, J.S.; Sun, M.; Kho, A.T.; Moore, K.G.; Sylvia, J.M.; Weiss, S.T.; Lu, Q.; Tantisira, K.G. Circulating microRNAs and association with methacholine PC20 in the Childhood Asthma Management Program (CAMP) cohort. PLoS ONE 2017, 12, e0180329. [Google Scholar] [CrossRef]
  44. Lefaudeux, D.; De Meulder, B.; Loza, M.J.; Peffer, N.; Rowe, A.; Baribaud, F.; Bansal, A.T.; Lutter, R.; Sousa, A.R.; Corfield, J.; et al. U-BIOPRED clinical adult asthma clusters linked to a subset of sputum omics. J. Allergy Clin. Immunol. 2017, 139, 1797–1807. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Kuo, C.S.; Pavlidis, S.; Loza, M.; Baribaud, F.; Rowe, A.; Pandis, I.; Sousa, A.; Corfield, J.; Djukanovic, R.; Lutter, R.; et al. T-helper cell type 2 (Th2) and non-Th2 molecular phenotypes of asthma using sputum transcriptomics in U-BIOPRED. Eur. Respir. J. 2017, 49, 1602135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Guerra, E.N.S.; Acevedo, A.C.; de Toledo, I.P.; Combes, A.; Chardin, H. Do mucosal biomarkers reveal the immunological state associated with food allergy? Allergy 2018, 73, 2392–2394. [Google Scholar] [CrossRef]
  47. Walter, J.; O’Mahony, L. The importance of social networks-An ecological and evolutionary framework to explain the role of microbes in the aetiology of allergy and asthma. Allergy 2019, 74, 2248–2251. [Google Scholar] [CrossRef] [PubMed]
  48. Huang, Y.J.; Nariya, S.; Harris, J.M.; Lynch, S.V.; Choy, D.F.; Arron, J.R.; Boushey, H. The airway microbiome in patients with severe asthma: Associations with disease features and severity. J. Allergy Clin. Immunol. 2015, 136, 874–884. [Google Scholar] [CrossRef] [Green Version]
  49. Pitsios, C. Allergen Immunotherapy: Biomarkers and Clinical Outcome Measures. J. Asthma Allergy 2021, 14, 141–148. [Google Scholar] [CrossRef]
  50. Lunjani, N.; Satitsuksanoa, P.; Lukasik, Z.; Sokolowska, M.; Eiwegger, T.; O’Mahony, L. Recent developments and highlights in mechanisms of allergic diseases: Microbiome. Allergy 2018, 73, 2314–2327. [Google Scholar] [CrossRef] [Green Version]
  51. Ivanova, O.; Richards, L.B.; Vijverberg, S.J.; Neerincx, A.H.; Sinha, A.; Sterk, P.J.; Maitland-van der Zee, A.H. What did we learn from multiple omics studies in asthma? Allergy 2019, 74, 2129–2145. [Google Scholar] [CrossRef] [Green Version]
  52. Schofield, J.P.R.; Burg, D.; Nicholas, B.; Strazzeri, F.; Brandsma, J.; Staykova, D.; Folisi, C.; Bansal, A.T.; Xian, Y.; Guo, Y.; et al. Stratification of asthma phenotypes by airway proteomic signatures. J. Allergy Clin. Immunol. 2019, 144, 70–82. [Google Scholar] [CrossRef] [Green Version]
  53. Nieto-Fontarigo, J.J.; Gonzalez-Barcala, F.J.; Andrade-Bulos, L.J.; San-Jose, M.E.; Cruz, M.J.; Valdes-Cuadrado, L.; Crujeiras, R.M.; Arias, P.; Salgado, F.J. iTRAQ-based proteomic analysis reveals potential serum biomarkers of allergic and nonallergic asthma. Allergy 2020, 75, 3171–3183. [Google Scholar] [CrossRef]
  54. Abdel-Aziz, M.I.; de Vries, R.; Lammers, A.; Xu, B.; Neerincx, A.H.; Vijverberg, S.J.H.; Dagelet, Y.W.F.; Kraneveld, A.D.; Frey, U.; Lutter, R.; et al. Cross-sectional biomarker comparisons in asthma monitoring using a longitudinal design: The eNose premise. Allergy 2020, 75, 2690–2693. [Google Scholar] [CrossRef] [PubMed]
  55. Sokolowska, M.; Rovati, G.E.; Diamant, Z.; Untersmayr, E.; Schwarze, J.; Lukasik, Z.; Sava, F.; Angelina, A.; Palomares, O.; Akdis, C.A.; et al. Current perspective on eicosanoids in asthma and allergic diseases: EAACI Task Force consensus report, part I. Allergy 2021, 76, 114–130. [Google Scholar] [CrossRef] [PubMed]
  56. Yang, L.; Zhao, Q.; Wang, S. The role of serum periostin in the diagnosis of asthma: A meta-analysis. Allergy Asthma Proc. 2020, 41, 240–247. [Google Scholar] [CrossRef] [PubMed]
  57. Asano, T.; Ohbayashi, H.; Ariga, M.; Furuta, O.; Kudo, S.; Ono, J.; Izuhara, K. Serum periostin reflects dynamic hyperinflation in patients with asthma. ERJ Open Res. 2020, 6, 00347-2019. [Google Scholar] [CrossRef] [PubMed]
  58. Eguiluz-Gracia, I.; Tay, T.R.; Hew, M.; Escribese, M.M.; Barber, D.; O’Hehir, R.E.; Torres, M.J. Recent developments and highlights in biomarkers in allergic diseases and asthma. Allergy 2018, 73, 2290–2305. [Google Scholar] [CrossRef] [Green Version]
  59. Hur, G.Y.; Pham, A.; Miller, M.; Weng, N.; Hu, J.; Kurten, R.C.; Broide, D.H. ORMDL3 but not neighboring 17q21 gene LRRC3C is expressed in human lungs and lung cells of asthmatics. Allergy 2020, 75, 2061–2065. [Google Scholar] [CrossRef]
  60. Yang, I.V.; Pedersen, B.S.; Liu, A.H.; O’Connor, G.T.; Pillai, D.; Kattan, M.; Misiak, R.T.; Gruchalla, R.; Szefler, S.J.; Khurana Hershey, G.K.; et al. The nasal methylome and childhood atopic asthma. J. Allergy Clin. Immunol. 2017, 139, 1478–1488. [Google Scholar] [CrossRef] [Green Version]
  61. Li, C.Y.; Peng, J.; Ren, L.P.; Gan, L.X.; Lu, X.J.; Liu, Q.; Gu, W.; Guo, X.J. Roles of histone hypoacetylation in LAT expression on T cells and Th2 polarization in allergic asthma. J. Transl. Med. 2013, 11, 26. [Google Scholar] [CrossRef] [Green Version]
  62. Kidd, C.D.; Thompson, P.J.; Barrett, L.; Baltic, S. Histone Modifications and Asthma. The Interface of the Epigenetic and Genetic Landscapes. Am. J. Respir. Cell Mol. Biol. 2016, 54, 3–12. [Google Scholar] [CrossRef]
  63. Harb, H.; Raedler, D.; Ballenberger, N.; Bock, A.; Kesper, D.A.; Renz, H.; Schaub, B. Childhood allergic asthma is associated with increased IL-13 and FOXP3 histone acetylation. J. Allergy Clin. Immunol. 2015, 136, 200–202. [Google Scholar] [CrossRef]
  64. Hirai, K.; Shirai, T.; Shimoshikiryo, T.; Ueda, M.; Gon, Y.; Maruoka, S.; Itoh, K. Circulating microRNA-15b-5p as a biomarker for asthma-COPD overlap. Allergy 2021, 76, 766–774. [Google Scholar] [CrossRef] [PubMed]
  65. Specjalski, K.; Niedoszytko, M. MicroRNAs: Future biomarkers and targets of therapy in asthma? Curr. Opin. Pulm. Med. 2020, 26, 285–292. [Google Scholar] [CrossRef]
  66. Simpson, L.J.; Patel, S.; Bhakta, N.R.; Choy, D.F.; Brightbill, H.D.; Ren, X.; Wang, Y.; Pua, H.H.; Baumjohann, D.; Montoya, M.M.; et al. A microRNA upregulated in asthma airway T cells promotes TH2 cytokine production. Nat. Immunol. 2014, 15, 1162–1170. [Google Scholar] [CrossRef] [PubMed]
  67. Pinkerton, M.; Chinchilli, V.; Banta, E.; Craig, T.; August, A.; Bascom, R.; Cantorna, M.; Harvill, E.; Ishmael, F.T. Differential expression of microRNAs in exhaled breath condensates of patients with asthma, patients with chronic obstructive pulmonary disease, and healthy adults. J. Allergy Clin. Immunol. 2013, 132, 217–219. [Google Scholar] [CrossRef] [PubMed]
  68. Panganiban, R.P.; Pinkerton, M.H.; Maru, S.Y.; Jefferson, S.J.; Roff, A.N.; Ishmael, F.T. Differential microRNA epression in asthma and the role of miR-1248 in regulation of IL-5. Am. J. Clin. Exp. Immunol. 2012, 1, 154–165. [Google Scholar]
  69. Polikepahad, S.; Knight, J.M.; Naghavi, A.O.; Oplt, T.; Creighton, C.J.; Shaw, C.; Benham, A.L.; Kim, J.; Soibam, B.; Harris, R.A.; et al. Proinflammatory role for let-7 microRNAS in experimental asthma. J. Biol. Chem. 2010, 285, 30139–30149. [Google Scholar] [CrossRef] [Green Version]
  70. Nakano, T.; Inoue, Y.; Shimojo, N.; Yamaide, F.; Morita, Y.; Arima, T.; Tomiita, M.; Kohno, Y. Lower levels of hsa-mir-15a, which decreases VEGFA, in the CD4+ T cells of pediatric patients with asthma. J. Allergy Clin. Immunol. 2013, 132, 1224–1227. [Google Scholar] [CrossRef]
  71. Feketea, G.; Bocsan, C.I.; Popescu, C.; Gaman, M.; Stanciu, L.A.; Zdrenghea, M.T. A Review of Macrophage MicroRNAs’ Role in Human Asthma. Cells 2019, 8, 420. [Google Scholar] [CrossRef] [Green Version]
  72. Wardzynska, A.; Pawelczyk, M.; Rywaniak, J.; Makowska, J.; Jamroz-Brzeska, J.; Kowalski, M.L. Circulating miRNA expression in asthmatics is age-related and associated with clinical asthma parameters, respiratory function and systemic inflammation. Respir. Res. 2021, 22, 177. [Google Scholar] [CrossRef]
  73. Torregrosa Paredes, P.; Esser, J.; Admyre, C.; Nord, M.; Rahman, Q.K.; Lukic, A.; Radmark, O.; Gronneberg, R.; Grunewald, J.; Eklund, A.; et al. Bronchoalveolar lavage fluid exosomes contribute to cytokine and leukotriene production in allergic asthma. Allergy 2012, 67, 911–919. [Google Scholar] [CrossRef]
  74. Sastre, B.; Canas, J.A.; Rodrigo-Munoz, J.M.; Del Pozo, V. Novel Modulators of Asthma and Allergy: Exosomes and MicroRNAs. Front. Immunol. 2017, 8, 826. [Google Scholar] [CrossRef] [PubMed]
  75. Levanen, B.; Bhakta, N.R.; Torregrosa Paredes, P.; Barbeau, R.; Hiltbrunner, S.; Pollack, J.L.; Skold, C.M.; Svartengren, M.; Grunewald, J.; Gabrielsson, S.; et al. Altered microRNA profiles in bronchoalveolar lavage fluid exosomes in asthmatic patients. J. Allergy Clin. Immunol. 2013, 131, 894–903. [Google Scholar] [CrossRef] [Green Version]
  76. Kyyaly, M.A.; Vorobeva, E.V.; Kothalawala, D.M.; Fong, W.C.G.; He, P.; Sones, C.L.; Al-Zahrani, M.; Sanchez-Elsner, T.; Arshad, S.H.; Kurukulaaratchy, R.J. MicroRNAs-A Promising Tool for Asthma Diagnosis and Severity Assessment: A Systematic Review. J. Pers. Med. 2022, 12, 543. [Google Scholar] [CrossRef] [PubMed]
  77. Wu, X.B.; Wang, M.Y.; Zhu, H.Y.; Tang, S.Q.; You, Y.D.; Xie, Y.Q. Overexpression of microRNA-21 and microRNA-126 in the patients of bronchial asthma. Int. J. Clin. Exp. Med. 2014, 7, 1307–1312. [Google Scholar] [PubMed]
  78. Hammad Mahmoud Hammad, R.; Hamed, D.; Eldosoky, M.; Ahmad, A.; Osman, H.M.; Abd Elgalil, H.M.; Mahmoud Hassan, M.M. Plasma microRNA-21, microRNA-146a and IL-13 expression in asthmatic children. Innate Immun. 2018, 24, 171–179. [Google Scholar] [CrossRef] [Green Version]
  79. ElKashef, S.; Ahmad, S.E.; Soliman, Y.M.A.; Mostafa, M.S. Role of microRNA-21 and microRNA-155 as biomarkers for bronchial asthma. Innate Immun. 2021, 27, 61–69. [Google Scholar] [CrossRef] [Green Version]
  80. Maes, T.; Cobos, F.A.; Schleich, F.; Sorbello, V.; Henket, M.; De Preter, K.; Bracke, K.R.; Conickx, G.; Mesnil, C.; Vandesompele, J.; et al. Asthma inflammatory phenotypes show differential microRNA expression in sputum. J. Allergy Clin. Immunol. 2016, 137, 1433–1446. [Google Scholar] [CrossRef] [Green Version]
  81. Shi, J.; Chen, M.; Ouyang, L.; Wang, Q.; Guo, Y.; Huang, L.; Jiang, S. miR-142-5p and miR-130a-3p regulate pulmonary macrophage polarization and asthma airway remodeling. Immunol. Cell Biol. 2020, 98, 715–725. [Google Scholar] [CrossRef]
  82. Bartel, S.; Carraro, G.; Alessandrini, F.; Krauss-Etschmann, S.; Ricciardolo, F.L.M.; Bellusci, S. miR-142-3p is associated with aberrant WNT signaling during airway remodeling in asthma. Am. J. Physiol. Lung Cell Mol. Physiol. 2018, 315, 328–333. [Google Scholar] [CrossRef]
  83. Comer, B.S.; Camoretti-Mercado, B.; Kogut, P.C.; Halayko, A.J.; Solway, J.; Gerthoffer, W.T. MicroRNA-146a and microRNA-146b expression and anti-inflammatory function in human airway smooth muscle. Am. J. Physiol. Lung Cell Mol. Physiol. 2014, 307, 727–734. [Google Scholar] [CrossRef]
  84. Malmhall, C.; Johansson, K.; Winkler, C.; Alawieh, S.; Ekerljung, L.; Radinger, M. Altered miR-155 Expression in Allergic Asthmatic Airways. Scand. J. Immunol. 2017, 85, 300–307. [Google Scholar] [CrossRef]
  85. Rijavec, M.; Korosec, P.; Zavbi, M.; Kern, I.; Malovrh, M.M. Let-7a is differentially expressed in bronchial biopsies of patients with severe asthma. Sci. Rep. 2014, 4, 6103. [Google Scholar] [CrossRef] [Green Version]
  86. Shaik, N.A.; Nasser, K.; Mohammed, A.; Mujalli, A.; Obaid, A.A.; El-Harouni, A.A.; Elango, R.; Banaganapalli, B. Identification of miRNA-mRNA-TFs regulatory network and crucial pathways involved in asthma through advanced systems biology approaches. PLoS ONE 2022, 17, e0271262. [Google Scholar] [CrossRef]
  87. Wise, S.K.; Lin, S.Y.; Toskala, E.; Orlandi, R.R.; Akdis, C.A.; Alt, J.A.; Azar, A.; Baroody, F.M.; Bachert, C.; Canonica, G.W.; et al. International Consensus Statement on Allergy and Rhinology: Allergic Rhinitis. Int. Forum Allergy Rhinol. 2018, 8, 108–352. [Google Scholar] [CrossRef]
  88. Zielen, S.; Devillier, P.; Heinrich, J.; Richter, H.; Wahn, U. Sublingual immunotherapy provides long-term relief in allergic rhinitis and reduces the risk of asthma: A retrospective, real-world database analysis. Allergy 2018, 73, 165–177. [Google Scholar] [CrossRef] [Green Version]
  89. Dong, X.; Zhong, N.; Fang, Y.; Cai, Q.; Lu, M.; Lu, Q. MicroRNA 27b-3p Modulates SYK in Pediatric Asthma Induced by Dust Mites. Front. Pediatr. 2018, 6, 301. [Google Scholar] [CrossRef] [Green Version]
  90. Zhang, K.; Liang, Y.; Feng, Y.; Wu, W.; Zhang, H.; He, J.; Hu, Q.; Zhao, J.; Xu, Y.; Liu, Z.; et al. Decreased epithelial and sputum miR-221-3p associates with airway eosinophilic inflammation and CXCL17 expression in asthma. Am. J. Physiol. Lung Cell Mol. Physiol. 2018, 315, 253–264. [Google Scholar] [CrossRef] [PubMed]
  91. Weidner, J.; Ekerljung, L.; Malmhall, C.; Miron, N.; Radinger, M. Circulating microRNAs correlate to clinical parameters in individuals with allergic and non-allergic asthma. Respir Res 2020, 21, 107. [Google Scholar] [CrossRef] [PubMed]
  92. Sun, Q.; Liu, L.; Wang, H.; Mandal, J.; Khan, P.; Hostettler, K.E.; Stolz, D.; Tamm, M.; Molino, A.; Lardinois, D.; et al. Constitutive high expression of protein arginine methyltransferase 1 in asthmatic airway smooth muscle cells is caused by reduced microRNA-19a expression and leads to enhanced remodeling. J. Allergy Clin. Immunol. 2017, 140, 510–524. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Haj-Salem, I.; Fakhfakh, R.; Berube, J.C.; Jacques, E.; Plante, S.; Simard, M.J.; Bosse, Y.; Chakir, J. MicroRNA-19a enhances proliferation of bronchial epithelial cells by targeting TGFbetaR2 gene in severe asthma. Allergy 2015, 70, 212–219. [Google Scholar] [CrossRef] [PubMed]
  94. Rodrigo-Munoz, J.M.; Canas, J.A.; Sastre, B.; Rego, N.; Greif, G.; Rial, M.; Minguez, P.; Mahillo-Fernandez, I.; Fernandez-Nieto, M.; Mora, I.; et al. Asthma diagnosis using integrated analysis of eosinophil microRNAs. Allergy 2019, 74, 507–517. [Google Scholar] [CrossRef] [PubMed]
  95. Papi, A.; Brightling, C.; Pedersen, S.E.; Reddel, H.K. Asthma. Lancet 2018, 391, 783–800. [Google Scholar] [CrossRef]
  96. Tabar, A.I.; Delgado, J.; Gonzalez-Mancebo, E.; Arroabarren, E.; Soto Retes, L.; Dominguez-Ortega, J.; Spanish Allergy and Clinical Immunology Scientific Society (SEAIC). Recent Advances in Allergen-Specific Immunotherapy as Treatment for Allergic Asthma: A Practical Overview. Int. Arch. Allergy Immunol. 2021, 182, 496–514. [Google Scholar] [CrossRef] [PubMed]
  97. Schoettler, N.; Strek, M.E. Recent Advances in Severe Asthma: From Phenotypes to Personalized Medicine. Chest 2020, 157, 516–528. [Google Scholar] [CrossRef]
  98. Lommatzsch, M.; Buhl, R.; Korn, S. The Treatment of Mild and Moderate Asthma in Adults. Dtsch. Arztebl. Int. 2020, 117, 434–444. [Google Scholar] [CrossRef] [PubMed]
  99. Sobieraj, D.M.; Baker, W.L. Medications for Asthma. JAMA 2018, 319, 1520. [Google Scholar] [CrossRef]
  100. Kwah, J.H.; Peters, A.T. Asthma in adults: Principles of treatment. Allergy Asthma Proc. 2019, 40, 396–402. [Google Scholar] [CrossRef] [PubMed]
  101. Kume, H.; Fukunaga, K.; Oguma, T. Research and development of bronchodilators for asthma and COPD with a focus on G protein/KCa channel linkage and beta2-adrenergic intrinsic efficacy. Pharmacol. Ther. 2015, 156, 75–89. [Google Scholar] [CrossRef]
  102. Heffler, E.; Madeira, L.N.G.; Ferrando, M.; Puggioni, F.; Racca, F.; Malvezzi, L.; Passalacqua, G.; Canonica, G.W. Inhaled Corticosteroids Safety and Adverse Effects in Patients with Asthma. J. Allergy Clin. Immunol. Pract. 2018, 6, 776–781. [Google Scholar] [CrossRef]
  103. Philip, J. The effects of inhaled corticosteroids on growth in children. Open Respir. Med. J. 2014, 8, 66–73. [Google Scholar] [CrossRef] [Green Version]
  104. Sullivan, P.W.; Ghushchyan, V.H.; Globe, G.; Schatz, M. Oral corticosteroid exposure and adverse effects in asthmatic patients. J. Allergy Clin. Immunol. 2018, 141, 110–116. [Google Scholar] [CrossRef] [PubMed]
  105. Price, D.B.; Trudo, F.; Voorham, J.; Xu, X.; Kerkhof, M.; Jie, J.L.Z.; Tran, T.N. Adverse outcomes from initiation of systemic corticosteroids for asthma: Long-term observational study. J. Asthma Allergy 2018, 11, 193–204. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Figueroa, D.J.; Breyer, R.M.; Defoe, S.K.; Kargman, S.; Daugherty, B.L.; Waldburger, K.; Liu, Q.; Clements, M.; Zeng, Z.; O’Neill, G.P.; et al. Expression of the cysteinyl leukotriene 1 receptor in normal human lung and peripheral blood leukocytes. Am. J. Respir. Crit. Care Med. 2001, 163, 226–233. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Ban, G.Y.; Kim, S.H.; Park, H.S. Persistent Eosinophilic Inflammation in Adult Asthmatics with High Serum and Urine Levels of Leukotriene E4. J. Asthma Allergy 2021, 14, 1219–1230. [Google Scholar] [CrossRef]
  108. Yokomizo, T.; Nakamura, M.; Shimizu, T. Leukotriene receptors as potential therapeutic targets. J. Clin. Investig. 2018, 128, 2691–2701. [Google Scholar] [CrossRef]
  109. Chauhan, B.F.; Ducharme, F.M. Anti-leukotriene agents compared to inhaled corticosteroids in the management of recurrent and/or chronic asthma in adults and children. Cochrane Database Syst. Rev. 2012, 2012, CD002314. [Google Scholar] [CrossRef]
  110. Bleecker, E.R.; Welch, M.J.; Weinstein, S.F.; Kalberg, C.; Johnson, M.; Edwards, L.; Rickard, K.A. Low-dose inhaled fluticasone propionate versus oral zafirlukast in the treatment of persistent asthma. J. Allergy Clin. Immunol. 2000, 105, 1123–1129. [Google Scholar] [CrossRef]
  111. Gosens, R.; Gross, N. The mode of action of anticholinergics in asthma. Eur. Respir. J. 2018, 52, 1701247. [Google Scholar] [CrossRef]
  112. Buels, K.S.; Fryer, A.D. Muscarinic receptor antagonists: Effects on pulmonary function. Handb. Exp. Pharmacol. 2012, 208, 317–341. [Google Scholar] [CrossRef] [Green Version]
  113. Egan, M.; Bunyavanich, S. Allergic rhinitis: The “Ghost Diagnosis” in patients with asthma. Asthma Res. Pract. 2015, 1, 8. [Google Scholar] [CrossRef] [Green Version]
  114. Akhouri, S.; House, S.A. Allergic Rhinitis; StatPearls: Treasure Island, FL, USA, 2022. [Google Scholar]
  115. Skoner, D.P. Allergic rhinitis: Definition, epidemiology, pathophysiology, detection, and diagnosis. J. Allergy Clin. Immunol. 2001, 108, 2–8. [Google Scholar] [CrossRef]
  116. Eguiluz-Gracia, I.; Testera-Montes, A.; Gonzalez, M.; Perez-Sanchez, N.; Ariza, A.; Salas, M.; Moreno-Aguilar, C.; Campo, P.; Torres, M.J.; Rondon, C. Safety and reproducibility of nasal allergen challenge. Allergy 2019, 74, 1125–1134. [Google Scholar] [CrossRef] [PubMed]
  117. Gomez, E.; Campo, P.; Rondon, C.; Barrionuevo, E.; Blanca-Lopez, N.; Torres, M.J.; Herrera, R.; Galindo, L.; Mayorga, C.; Blanca, M. Role of the basophil activation test in the diagnosis of local allergic rhinitis. J. Allergy Clin. Immunol. 2013, 132, 975–976. [Google Scholar] [CrossRef]
  118. Gevaert, P.; Omachi, T.A.; Corren, J.; Mullol, J.; Han, J.; Lee, S.E.; Kaufman, D.; Ligueros-Saylan, M.; Howard, M.; Zhu, R.; et al. Efficacy and safety of omalizumab in nasal polyposis: 2 randomized phase 3 trials. J. Allergy Clin. Immunol. 2020, 146, 595–605. [Google Scholar] [CrossRef]
  119. Emson, C.; Corren, J.; Salapa, K.; Hellqvist, A.; Parnes, J.R.; Colice, G. Efficacy of Tezepelumab in Patients with Severe, Uncontrolled Asthma with and without Nasal Polyposis: A Post Hoc Analysis of the Phase 2b PATHWAY Study. J. Asthma Allergy 2021, 14, 91–99. [Google Scholar] [CrossRef] [PubMed]
  120. Bachert, C.; Sousa, A.R.; Lund, V.J.; Scadding, G.K.; Gevaert, P.; Nasser, S.; Durham, S.R.; Cornet, M.E.; Kariyawasam, H.H.; Gilbert, J.; et al. Reduced need for surgery in severe nasal polyposis with mepolizumab: Randomized trial. J. Allergy Clin. Immunol. 2017, 140, 1024–1031. [Google Scholar] [CrossRef] [Green Version]
  121. Bachert, C.; Han, J.K.; Desrosiers, M.; Hellings, P.W.; Amin, N.; Lee, S.E.; Mullol, J.; Greos, L.S.; Bosso, J.V.; Laidlaw, T.M.; et al. Efficacy and safety of dupilumab in patients with severe chronic rhinosinusitis with nasal polyps (LIBERTY NP SINUS-24 and LIBERTY NP SINUS-52): Results from two multicentre, randomised, double-blind, placebo-controlled, parallel-group phase 3 trials. Lancet 2019, 394, 1638–1650. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Dykewicz, M.S.; Wallace, D.V.; Amrol, D.J.; Baroody, F.M.; Bernstein, J.A.; Craig, T.J.; Dinakar, C.; Ellis, A.K.; Finegold, I.; Golden, D.B.K.; et al. Rhinitis 2020: A practice parameter update. J. Allergy Clin. Immunol. 2020, 146, 721–767. [Google Scholar] [CrossRef] [PubMed]
  123. Alobid, I.; Anton, E.; Armengot, M.; Chao, J.; Colas, C.; del Cuvillo, A.; Davila, I.; Dordal, M.T.; Escobar, C.; Fernandez-Parra, B.; et al. SEAIC-SEORL. Consensus Document on Nasal Polyposis. POLINA Project. J. Investig. Allergol. Clin. Immunol. 2011, 21 (Suppl. 1), 1–58. [Google Scholar]
  124. Bousquet, P.J.; Combescure, C.; Neukirch, F.; Klossek, J.M.; Mechin, H.; Daures, J.P.; Bousquet, J. Visual analog scales can assess the severity of rhinitis graded according to ARIA guidelines. Allergy 2007, 62, 367–372. [Google Scholar] [CrossRef]
  125. Seidman, M.D.; Gurgel, R.K.; Lin, S.Y.; Schwartz, S.R.; Baroody, F.M.; Bonner, J.R.; Dawson, D.E.; Dykewicz, M.S.; Hackell, J.M.; Han, J.K.; et al. Clinical practice guideline: Allergic rhinitis executive summary. Otolaryngol. Head Neck Surg. 2015, 152, 197–206. [Google Scholar] [CrossRef]
  126. Dordal, M.T.; Lluch-Bernal, M.; Sanchez, M.C.; Rondon, C.; Navarro, A.; Montoro, J.; Matheu, V.; Ibanez, M.D.; Fernandez-Parra, B.; Davila, I.; et al. Allergen-specific nasal provocation testing: Review by the rhinoconjunctivitis committee of the Spanish Society of Allergy and Clinical Immunology. J. Investig. Allergol. Clin. Immunol. 2011, 21, 1–12. [Google Scholar]
  127. Zhang, Y.; Lan, F.; Zhang, L. Advances and highlights in allergic rhinitis. Allergy 2021, 76, 3383–3389. [Google Scholar] [CrossRef] [PubMed]
  128. Small, P.; Keith, P.K.; Kim, H. Allergic rhinitis. Allergy Asthma Clin. Immunol. 2018, 14, 51. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Canonica, G.W.; Ansotegui, I.J.; Pawankar, R.; Schmid-Grendelmeier, P.; van Hage, M.; Baena-Cagnani, C.E.; Melioli, G.; Nunes, C.; Passalacqua, G.; Rosenwasser, L.; et al. A WAO—ARIA—GA(2)LEN consensus document on molecular-based allergy diagnostics. World Allergy Organ. J. 2013, 6, 17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Luengo, O.; Cardona, V. Component resolved diagnosis: When should it be used? Clin. Transl. Allergy 2014, 4, 28. [Google Scholar] [CrossRef] [Green Version]
  131. Papadopoulos, N.G.; Bernstein, J.A.; Demoly, P.; Dykewicz, M.; Fokkens, W.; Hellings, P.W.; Peters, A.T.; Rondon, C.; Togias, A.; Cox, L.S. Phenotypes and endotypes of rhinitis and their impact on management: A PRACTALL report. Allergy 2015, 70, 474–494. [Google Scholar] [CrossRef] [PubMed]
  132. Hamilton, R.G. Allergic sensitization is a key risk factor for but not synonymous with allergic disease. J. Allergy Clin. Immunol. 2014, 134, 360–361. [Google Scholar] [CrossRef] [PubMed]
  133. Campo, P.; Eguiluz-Gracia, I.; Bogas, G.; Salas, M.; Plaza Seron, C.; Perez, N.; Mayorga, C.; Torres, M.J.; Shamji, M.H.; Rondon, C. Local allergic rhinitis: Implications for management. Clin. Exp. Allergy 2019, 49, 6–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Vlaykov, A.N.; Tacheva, T.T.; Vlaykova, T.I.; Stoyanov, V.K. Serum and local IL-4, IL-5, IL-13 and immunoglobulin E in allergic rhinitis. Postepy Dermatol. Alergol. 2020, 37, 719–724. [Google Scholar] [CrossRef]
  135. Nur Husna, S.M.; Md Shukri, N.; Tuan Sharif, S.E.; Tan, H.T.T.; Mohd Ashari, N.S.; Wong, K.K. IL-4/IL-13 Axis in Allergic Rhinitis: Elevated Serum Cytokines Levels and Inverse Association With Tight Junction Molecules Expression. Front. Mol. Biosci. 2022, 9, 819772. [Google Scholar] [CrossRef]
  136. Li, C.; Xu, Y.; Luo, X.; Chen, F. The Effect of IL-18 on Group 2 Innate Lymphoid Cells in Allergic Rhinitis. Iran J. Immunol. 2021, 18, 188–194. [Google Scholar] [CrossRef]
  137. Kim, D.W.; Kim, D.K.; Eun, K.M.; Bae, J.S.; Chung, Y.J.; Xu, J.; Kim, Y.M.; Mo, J.H. IL-25 Could Be Involved in the Development of Allergic Rhinitis Sensitized to House Dust Mite. Mediat. Inflamm. 2017, 2017, 3908049. [Google Scholar] [CrossRef]
  138. Qiao, Y.; Chen, J. Serum levels of IL-31, IL-33 and ST2 in allergic rhinitis of children in China. Cell. Mol. Biol. 2018, 64, 52–55. [Google Scholar] [CrossRef]
  139. Tyurin, Y.A.; Lissovskaya, S.A.; Fassahov, R.S.; Mustafin, I.G.; Shamsutdinov, A.F.; Shilova, M.A.; Rizvanov, A.A. Cytokine Profile of Patients with Allergic Rhinitis Caused by Pollen, Mite, and Microbial Allergen Sensitization. J. Immunol. Res. 2017, 2017, 3054217. [Google Scholar] [CrossRef] [PubMed]
  140. Luo, X.; Zeng, Q.; Yan, S.; Liu, W.; Luo, R. MicroRNA-375-mediated regulation of ILC2 cells through TSLP in allergic rhinitis. World Allergy Organ. J. 2020, 13, 100451. [Google Scholar] [CrossRef] [PubMed]
  141. Rondon, C.; Eguiluz-Gracia, I.; Shamji, M.H.; Layhadi, J.A.; Salas, M.; Torres, M.J.; Campo, P. IgE Test in Secretions of Patients with Respiratory Allergy. Curr. Allergy Asthma Rep. 2018, 18, 67. [Google Scholar] [CrossRef]
  142. Eguiluz-Gracia, I.; Perez-Sanchez, N.; Bogas, G.; Campo, P.; Rondon, C. How to Diagnose and Treat Local Allergic Rhinitis: A Challenge for Clinicians. J. Clin. Med. 2019, 8, 1062. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Erkan, K.; Bozkurt, M.K.; Artac, H.; Ozdemir, H.; Unlu, A.; Korucu, E.N.; Elsurer, C. The role of regulatory T cells in allergic rhinitis and their correlation with IL-10, IL-17 and neopterin levels in serum and nasal lavage fluid. Eur. Arch. Otorhinolaryngol. 2020, 277, 1109–1114. [Google Scholar] [CrossRef] [PubMed]
  144. Chen, Y.; Yang, M.; Deng, J.; Wang, K.; Shi, J.; Sun, Y. Elevated Levels of Activated and Pathogenic Eosinophils Characterize Moderate-Severe House Dust Mite Allergic Rhinitis. J. Immunol. Res. 2020, 2020, 8085615. [Google Scholar] [CrossRef]
  145. Holgate, S.T.; Davies, D.E. Rethinking the pathogenesis of asthma. Immunity 2009, 31, 362–367. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Semik-Orzech, A.; Barczyk, A.; Wiaderkiewicz, R.; Pierzchala, W. Eotaxin, but not IL-8, is increased in upper and lower airways of allergic rhinitis subjects after nasal allergen challenge. Allergy Asthma Proc. 2011, 32, 230–238. [Google Scholar] [CrossRef] [PubMed]
  147. Bartemes, K.R.; Kephart, G.M.; Fox, S.J.; Kita, H. Enhanced innate type 2 immune response in peripheral blood from patients with asthma. J. Allergy Clin. Immunol. 2014, 134, 671–678. [Google Scholar] [CrossRef]
  148. Sun, R.; Yang, Y.; Huo, Q.; Gu, Z.; Wei, P.; Tang, X. Increased expression of type 2 innate lymphoid cells in pediatric patients with allergic rhinitis. Exp. Ther. Med. 2020, 19, 735–740. [Google Scholar] [CrossRef] [Green Version]
  149. Testera-Montes, A.; Jurado, R.; Salas, M.; Eguiluz-Gracia, I.; Mayorga, C. Diagnostic Tools in Allergic Rhinitis. Front. Allergy 2021, 2, 721851. [Google Scholar] [CrossRef]
  150. Hemmings, O.; Kwok, M.; McKendry, R.; Santos, A.F. Basophil Activation Test: Old and New Applications in Allergy. Curr. Allergy Asthma Rep. 2018, 18, 77. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  151. Abbas, M.; Moussa, M.; Akel, H. Type I Hypersensitivity Reaction; StatPearls: Treasure Island, FL, USA, 2022. [Google Scholar]
  152. Duarte Ferreira, R.; Ornelas, C.; Silva, S.; Morgado, R.; Pereira, D.; Escaleira, D.; Moreira, S.; Valenca, J.; Pedro, E.; Branco Ferreira, M.; et al. Contribution of In Vivo and In Vitro Testing for The Diagnosis of Local Allergic Rhinitis. J. Investig. Allergol. Clin. Immunol. 2019, 29, 46–48. [Google Scholar] [CrossRef] [Green Version]
  153. Campo, P.; Villalba, M.; Barrionuevo, E.; Rondon, C.; Salas, M.; Galindo, L.; Rodriguez, M.J.; Lopez-Rodriguez, J.C.; Prieto-Del Prado, M.A.; Torres, M.J.; et al. Immunologic responses to the major allergen of Olea europaea in local and systemic allergic rhinitis subjects. Clin. Exp. Allergy 2015, 45, 1703–1712. [Google Scholar] [CrossRef] [Green Version]
  154. Eguiluz-Gracia, I.; Fernandez-Santamaria, R.; Testera-Montes, A.; Ariza, A.; Campo, P.; Prieto, A.; Perez-Sanchez, N.; Salas, M.; Mayorga, C.; Torres, M.J.; et al. Coexistence of nasal reactivity to allergens with and without IgE sensitization in patients with allergic rhinitis. Allergy 2020, 75, 1689–1698. [Google Scholar] [CrossRef]
  155. Yanai, M.; Sato, K.; Aoki, N.; Takiyama, Y.; Oikawa, K.; Kobayashi, H.; Kimura, S.; Harabuchi, Y.; Tateno, M. The role of CCL22/macrophage-derived chemokine in allergic rhinitis. Clin. Immunol. 2007, 125, 291–298. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Sun, J.; Wong, B.; Cundall, M.; Goncharova, S.; Conway, M.; Dalrymple, A.; Coyle, A.J.; Waserman, S.; Jordana, M. Immunoreactivity profile of peripheral blood mononuclear cells from patients with ragweed-induced allergic rhinitis. Clin. Exp. Allergy 2007, 37, 901–908. [Google Scholar] [CrossRef]
  157. Baumann, R.; Rabaszowski, M.; Stenin, I.; Tilgner, L.; Scheckenbach, K.; Wiltfang, J.; Schipper, J.; Chaker, A.; Wagenmann, M. Comparison of the nasal release of IL-4, IL-10, IL-17, CCL13/MCP-4, and CCL26/eotaxin-3 in allergic rhinitis during season and after allergen challenge. Am. J. Rhinol. Allergy 2013, 27, 266–272. [Google Scholar] [CrossRef]
  158. Wu, G.; Yang, G.; Zhang, R.; Xu, G.; Zhang, L.; Wen, W.; Lu, J.; Liu, J.; Yu, Y. Altered microRNA Expression Profiles of Extracellular Vesicles in Nasal Mucus From Patients With Allergic Rhinitis. Allergy Asthma Immunol. Res. 2015, 7, 449–457. [Google Scholar] [CrossRef] [PubMed]
  159. Zhang, X.H.; Zhang, Y.N.; Liu, Z. MicroRNA in chronic rhinosinusitis and allergic rhinitis. Curr. Allergy Asthma Rep. 2014, 14, 415. [Google Scholar] [CrossRef] [PubMed]
  160. Hammad, N.M.; Nabil, F.; Elbehedy, E.M.; Sedeek, R.; Gouda, M.I.; Arafa, M.A.; Elalawi, S.M.; El Shahawy, A.A. Role of MicroRNA-155 as a Potential Biomarker for Allergic Rhinitis in Children. Can. Respir. J. 2021, 2021, 5554461. [Google Scholar] [CrossRef]
  161. Suojalehto, H.; Lindstrom, I.; Majuri, M.L.; Mitts, C.; Karjalainen, J.; Wolff, H.; Alenius, H. Altered microRNA expression of nasal mucosa in long-term asthma and allergic rhinitis. Int. Arch. Allergy Immunol. 2014, 163, 168–178. [Google Scholar] [CrossRef] [PubMed]
  162. Teng, Y.; Zhang, R.; Liu, C.; Zhou, L.; Wang, H.; Zhuang, W.; Huang, Y.; Hong, Z. miR-143 inhibits interleukin-13-induced inflammatory cytokine and mucus production in nasal epithelial cells from allergic rhinitis patients by targeting IL13Ralpha1. Biochem. Biophys. Res. Commun. 2015, 457, 58–64. [Google Scholar] [CrossRef]
  163. Stelmaszczyk-Emmel, A.; Zawadzka-Krajewska, A.; Kopatys, A.; Demkow, U. Th1, Th2, Th17, and regulatory cytokines in children with different clinical forms of allergy. Adv. Exp. Med. Biol. 2013, 788, 321–328. [Google Scholar] [CrossRef]
  164. Lee, M.F.; Song, P.P.; Hwang, G.Y.; Lin, S.J.; Chen, Y.H. Sensitization to Per a 2 of the American cockroach correlates with more clinical severity among airway allergic patients in Taiwan. Ann. Allergy Asthma Immunol. 2012, 108, 243–248. [Google Scholar] [CrossRef]
  165. Kim, J.A.; Cho, J.H.; Park, I.H.; Shin, J.M.; Lee, S.A.; Lee, H.M. Diesel Exhaust Particles Upregulate Interleukins IL-6 and IL-8 in Nasal Fibroblasts. PLoS ONE 2016, 11, e0157058. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Yu, X.; Wang, M.; Cao, Z. Reduced CD4(+)T Cell CXCR3 Expression in Patients With Allergic Rhinitis. Front. Immunol. 2020, 11, 581180. [Google Scholar] [CrossRef] [PubMed]
  167. Kant, A.; Terzioglu, K. Association of severity of allergic rhinitis with neutrophil-to-lymphocyte, eosinophil-to-neutrophil, and eosinophil-to-lymphocyte ratios in adults. Allergol. Immunopathol. 2021, 49, 94–99. [Google Scholar] [CrossRef] [PubMed]
  168. Doherty, T.A.; Scott, D.; Walford, H.H.; Khorram, N.; Lund, S.; Baum, R.; Chang, J.; Rosenthal, P.; Beppu, A.; Miller, M.; et al. Allergen challenge in allergic rhinitis rapidly induces increased peripheral blood type 2 innate lymphoid cells that express CD84. J. Allergy Clin. Immunol. 2014, 133, 1203–1205. [Google Scholar] [CrossRef] [PubMed]
  169. Dhariwal, J.; Cameron, A.; Trujillo-Torralbo, M.B.; Del Rosario, A.; Bakhsoliani, E.; Paulsen, M.; Jackson, D.J.; Edwards, M.R.; Rana, B.M.J.; Cousins, D.J.; et al. Mucosal Type 2 Innate Lymphoid Cells Are a Key Component of the Allergic Response to Aeroallergens. Am. J. Respir. Crit. Care Med. 2017, 195, 1586–1596. [Google Scholar] [CrossRef] [Green Version]
  170. Xu, G.; Zhang, L.; Wang, D.Y.; Xu, R.; Liu, Z.; Han, D.M.; Wang, X.D.; Zuo, K.J.; Li, H.B. Opposing roles of IL-17A and IL-25 in the regulation of TSLP production in human nasal epithelial cells. Allergy 2010, 65, 581–589. [Google Scholar] [CrossRef]
  171. Asaka, D.; Yoshikawa, M.; Nakayama, T.; Yoshimura, T.; Moriyama, H.; Otori, N. Elevated levels of interleukin-33 in the nasal secretions of patients with allergic rhinitis. Int. Arch. Allergy Immunol. 2012, 158, 47–50. [Google Scholar] [CrossRef]
  172. Zhu, D.D.; Zhu, X.W.; Jiang, X.D.; Dong, Z. Thymic stromal lymphopoietin expression is increased in nasal epithelial cells of patients with mugwort pollen sensitive-seasonal allergic rhinitis. Chin. Med. J. 2009, 122, 2303–2307. [Google Scholar] [CrossRef]
  173. Kamekura, R.; Kojima, T.; Takano, K.; Go, M.; Sawada, N.; Himi, T. The role of IL-33 and its receptor ST2 in human nasal epithelium with allergic rhinitis. Clin. Exp. Allergy 2012, 42, 218–228. [Google Scholar] [CrossRef]
  174. Kamekura, R.; Kojima, T.; Koizumi, J.; Ogasawara, N.; Kurose, M.; Go, M.; Harimaya, A.; Murata, M.; Tanaka, S.; Chiba, H.; et al. Thymic stromal lymphopoietin enhances tight-junction barrier function of human nasal epithelial cells. Cell Tissue Res. 2009, 338, 283–293. [Google Scholar] [CrossRef]
  175. Busse, W.W.; Maspero, J.F.; Lu, Y.; Corren, J.; Hanania, N.A.; Chipps, B.E.; Katelaris, C.H.; FitzGerald, J.M.; Quirce, S.; Ford, L.B.; et al. Efficacy of dupilumab on clinical outcomes in patients with asthma and perennial allergic rhinitis. Ann. Allergy Asthma Immunol. 2020, 125, 565–576. [Google Scholar] [CrossRef]
  176. Yu, C.; Wang, K.; Cui, X.; Lu, L.; Dong, J.; Wang, M.; Gao, X. Clinical Efficacy and Safety of Omalizumab in the Treatment of Allergic Rhinitis: A Systematic Review and Meta-analysis of Randomized Clinical Trials. Am. J. Rhinol. Allergy 2020, 34, 196–208. [Google Scholar] [CrossRef] [PubMed]
  177. Tsabouri, S.; Ntritsos, G.; Koskeridis, F.; Evangelou, E.; Olsson, P.; Kostikas, K. Omalizumab for the treatment of allergic rhinitis: A systematic review and meta-analysis. Rhinology 2021, 59, 501–510. [Google Scholar] [CrossRef] [PubMed]
  178. Weinstein, S.F.; Katial, R.; Jayawardena, S.; Pirozzi, G.; Staudinger, H.; Eckert, L.; Joish, V.N.; Amin, N.; Maroni, J.; Rowe, P.; et al. Efficacy and safety of dupilumab in perennial allergic rhinitis and comorbid asthma. J. Allergy Clin. Immunol. 2018, 142, 171–177. [Google Scholar] [CrossRef]
  179. Juel-Berg, N.; Darling, P.; Bolvig, J.; Foss-Skiftesvik, M.H.; Halken, S.; Winther, L.; Hansen, K.S.; Askjaer, N.; Heegaard, S.; Madsen, A.R.; et al. Intranasal corticosteroids compared with oral antihistamines in allergic rhinitis: A systematic review and meta-analysis. Am. J. Rhinol. Allergy 2017, 31, 19–28. [Google Scholar] [CrossRef] [PubMed]
  180. Bousquet, J.; Schunemann, H.J.; Togias, A.; Bachert, C.; Erhola, M.; Hellings, P.W.; Klimek, L.; Pfaar, O.; Wallace, D.; Ansotegui, I.; et al. Next-generation Allergic Rhinitis and Its Impact on Asthma (ARIA) guidelines for allergic rhinitis based on Grading of Recommendations Assessment, Development and Evaluation (GRADE) and real-world evidence. J. Allergy Clin. Immunol. 2020, 145, 70–80. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  181. Fein, M.N.; Fischer, D.A.; O’Keefe, A.W.; Sussman, G.L. CSACI position statement: Newer generation H1-antihistamines are safer than first-generation H1-antihistamines and should be the first-line antihistamines for the treatment of allergic rhinitis and urticaria. Allergy Asthma Clin. Immunol. 2019, 15, 61. [Google Scholar] [CrossRef] [Green Version]
  182. Church, M.K.; Maurer, M.; Simons, F.E.; Bindslev-Jensen, C.; van Cauwenberge, P.; Bousquet, J.; Holgate, S.T.; Zuberbier, T.; Global, A.; Asthma European, N. Risk of first-generation H(1)-antihistamines: A GA(2)LEN position paper. Allergy 2010, 65, 459–466. [Google Scholar] [CrossRef]
  183. Sanchez-Borges, M.; Ansotegui, I.J. Second generation antihistamines: An update. Curr. Opin. Allergy Clin. Immunol. 2019, 19, 358–364. [Google Scholar] [CrossRef]
  184. Feng, S.; Deng, C.; Li, L.; Liao, W.; Fan, Y.; Xu, G.; Li, H. Efficacy of intranasal antihistamine in the treatment of allergic rhinitis: A meta-analysis. Zhonghua Er Bi Yan Hou Tou Jing Wai Ke Za Zhi 2014, 49, 832–838. [Google Scholar]
  185. LaForce, C.F.; Corren, J.; Wheeler, W.J.; Berger, W.E.; Rhinitis Study, G. Efficacy of azelastine nasal spray in seasonal allergic rhinitis patients who remain symptomatic after treatment with fexofenadine. Ann. Allergy Asthma Immunol. 2004, 93, 154–159. [Google Scholar] [CrossRef]
  186. Kaliner, M.A.; Berger, W.E.; Ratner, P.H.; Siegel, C.J. The efficacy of intranasal antihistamines in the treatment of allergic rhinitis. Ann. Allergy Asthma Immunol. 2011, 106, 6–11. [Google Scholar] [CrossRef]
  187. Horak, F.; Zieglmayer, U.P.; Zieglmayer, R.; Kavina, A.; Marschall, K.; Munzel, U.; Petzold, U. Azelastine nasal spray and desloratadine tablets in pollen-induced seasonal allergic rhinitis: A pharmacodynamic study of onset of action and efficacy. Curr. Med. Res. Opin. 2006, 22, 151–157. [Google Scholar] [CrossRef]
  188. Shah, S.R.; Nayak, A.; Ratner, P.; Roland, P.; Michael Wall, G. Effects of olopatadine hydrochloride nasal spray 0.6% in the treatment of seasonal allergic rhinitis: A phase III, multicenter, randomized, double-blind, active- and placebo-controlled study in adolescents and adults. Clin. Ther. 2009, 31, 99–107. [Google Scholar] [CrossRef] [PubMed]
  189. Sastre, J.; Mosges, R. Local and systemic safety of intranasal corticosteroids. J. Investig. Allergol. Clin. Immunol. 2012, 22, 1–12. [Google Scholar] [PubMed]
  190. Bensch, G.W. Safety of intranasal corticosteroids. Ann. Allergy Asthma Immunol. 2016, 117, 601–605. [Google Scholar] [CrossRef] [PubMed]
  191. Kirtsreesakul, V.; Chansaksung, P.; Ruttanaphol, S. Dose-related effect of intranasal corticosteroids on treatment outcome of persistent allergic rhinitis. Otolaryngol. Head Neck Surg. 2008, 139, 565–569. [Google Scholar] [CrossRef] [PubMed]
  192. Zhang, K.; Li, A.R.; Miglani, A.; Nguyen, S.A.; Schlosser, R.J. Effect of Medical Therapy in Allergic Rhinitis: A Systematic Review and Meta-Analysis. Am. J. Rhinol. Allergy 2022, 36, 269–280. [Google Scholar] [CrossRef]
  193. Joseph, R.M.; Hunter, A.L.; Ray, D.W.; Dixon, W.G. Systemic glucocorticoid therapy and adrenal insufficiency in adults: A systematic review. Semin. Arthritis Rheum. 2016, 46, 133–141. [Google Scholar] [CrossRef]
  194. Hox, V.; Lourijsen, E.; Jordens, A.; Aasbjerg, K.; Agache, I.; Alobid, I.; Bachert, C.; Boussery, K.; Campo, P.; Fokkens, W.; et al. Benefits and harm of systemic steroids for short- and long-term use in rhinitis and rhinosinusitis: An EAACI position paper. Clin. Transl. Allergy 2020, 10, 1–27. [Google Scholar] [CrossRef]
  195. Bousquet, J.; Meltzer, E.O.; Couroux, P.; Koltun, A.; Kopietz, F.; Munzel, U.; Kuhl, H.C.; Nguyen, D.T.; Salapatek, A.M.; Price, D. Onset of Action of the Fixed Combination Intranasal Azelastine-Fluticasone Propionate in an Allergen Exposure Chamber. J. Allergy Clin. Immunol. Pract. 2018, 6, 1726–1732. [Google Scholar] [CrossRef]
  196. Price, D.; Shah, S.; Bhatia, S.; Bachert, C.; Berger, W.; Bousquet, J.; Carr, W.; Hellings, P.; Munzel, U.; Scadding, G.; et al. A new therapy (MP29-02) is effective for the long-term treatment of chronic rhinitis. J. Investig. Allergol. Clin. Immunol. 2013, 23, 495–503. [Google Scholar]
  197. Carr, W.; Bernstein, J.; Lieberman, P.; Meltzer, E.; Bachert, C.; Price, D.; Munzel, U.; Bousquet, J. A novel intranasal therapy of azelastine with fluticasone for the treatment of allergic rhinitis. J. Allergy Clin. Immunol. 2012, 129, 1282–1289. [Google Scholar] [CrossRef] [PubMed]
  198. Meltzer, E.O.; Bachert, C.; Mayer, M.J.; Kopietz, F.; Koltun, A.; Maus, J.; D’Addio, A.D. Deposition characteristics of a novel intranasal formulation of azelastine hydrochloride plus fluticasone propionate in an anatomic model of the human nasal cavity. Allergy Asthma Proc. 2020, 41, 265–270. [Google Scholar] [CrossRef] [PubMed]
  199. Hossenbaccus, L.; Linton, S.; Garvey, S.; Ellis, A.K. Towards definitive management of allergic rhinitis: Best use of new and established therapies. Allergy Asthma Clin. Immunol. 2020, 16, 39. [Google Scholar] [CrossRef]
  200. Wilson, A.M.; O’Byrne, P.M.; Parameswaran, K. Leukotriene receptor antagonists for allergic rhinitis: A systematic review and meta-analysis. Am. J. Med. 2004, 116, 338–344. [Google Scholar] [CrossRef] [PubMed]
  201. Rodrigo, G.J.; Yanez, A. The role of antileukotriene therapy in seasonal allergic rhinitis: A systematic review of randomized trials. Ann. Allergy Asthma Immunol. 2006, 96, 779–786. [Google Scholar] [CrossRef] [PubMed]
  202. Bousquet, J.; Anto, J.M.; Bachert, C.; Baiardini, I.; Bosnic-Anticevich, S.; Walter Canonica, G.; Melen, E.; Palomares, O.; Scadding, G.K.; Togias, A.; et al. Allergic rhinitis. Nat. Rev. Dis. Prim. 2020, 6, 95. [Google Scholar] [CrossRef]
  203. Saco, T.; Ugalde, I.C.; Cardet, J.C.; Casale, T.B. Strategies for choosing a biologic for your patient with allergy or asthma. Ann. Allergy Asthma Immunol. 2021, 127, 627–637. [Google Scholar] [CrossRef]
  204. Geng, B.; Dilley, M.; Anterasian, C. Biologic Therapies for Allergic Rhinitis and Nasal Polyposis. Curr. Allergy Asthma Rep. 2021, 21, 36. [Google Scholar] [CrossRef]
  205. Casale, T.B.; Condemi, J.; LaForce, C.; Nayak, A.; Rowe, M.; Watrous, M.; McAlary, M.; Fowler-Taylor, A.; Racine, A.; Gupta, N.; et al. Effect of omalizumab on symptoms of seasonal allergic rhinitis: A randomized controlled trial. JAMA 2001, 286, 2956–2967. [Google Scholar] [CrossRef] [Green Version]
  206. Pelaia, C.; Calabrese, C.; Terracciano, R.; de Blasio, F.; Vatrella, A.; Pelaia, G. Omalizumab, the first available antibody for biological treatment of severe asthma: More than a decade of real-life effectiveness. Ther. Adv. Respir. Dis. 2018, 12, 1753466618810192. [Google Scholar] [CrossRef] [Green Version]
  207. Bousquet, J.; Humbert, M.; Gibson, P.G.; Kostikas, K.; Jaumont, X.; Pfister, P.; Nissen, F. Real-World Effectiveness of Omalizumab in Severe Allergic Asthma: A Meta-Analysis of Observational Studies. J. Allergy Clin. Immunol. Pract. 2021, 9, 2702–2714. [Google Scholar] [CrossRef] [PubMed]
  208. Zhu, R.; Zheng, Y.; Putnam, W.S.; Visich, J.; Eisner, M.D.; Matthews, J.G.; Rosen, K.E.; D’Argenio, D.Z. Population-based efficacy modeling of omalizumab in patients with severe allergic asthma inadequately controlled with standard therapy. AAPS J. 2013, 15, 559–570. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  209. Casale, T.B.; Luskin, A.T.; Busse, W.; Zeiger, R.S.; Trzaskoma, B.; Yang, M.; Griffin, N.M.; Chipps, B.E. Omalizumab Effectiveness by Biomarker Status in Patients with Asthma: Evidence From PROSPERO, A Prospective Real-World Study. J. Allergy Clin. Immunol. Pract. 2019, 7, 156–164. [Google Scholar] [CrossRef]
  210. Fala, L. Nucala (Mepolizumab): First IL-5 Antagonist Monoclonal Antibody FDA Approved for Maintenance Treatment of Patients with Severe Asthma. Am. Health Drug Benefits 2016, 9, 106–110. [Google Scholar] [PubMed]
  211. Ortega, H.G.; Liu, M.C.; Pavord, I.D.; Brusselle, G.G.; FitzGerald, J.M.; Chetta, A.; Humbert, M.; Katz, L.E.; Keene, O.N.; Yancey, S.W.; et al. Mepolizumab treatment in patients with severe eosinophilic asthma. N. Engl. J. Med. 2014, 371, 1198–1207. [Google Scholar] [CrossRef] [Green Version]
  212. Khurana, S.; Brusselle, G.G.; Bel, E.H.; FitzGerald, J.M.; Masoli, M.; Korn, S.; Kato, M.; Albers, F.C.; Bradford, E.S.; Gilson, M.J.; et al. Long-term Safety and Clinical Benefit of Mepolizumab in Patients With the Most Severe Eosinophilic Asthma: The COSMEX Study. Clin. Ther. 2019, 41, 2041–2056. [Google Scholar] [CrossRef] [Green Version]
  213. Lugogo, N.; Domingo, C.; Chanez, P.; Leigh, R.; Gilson, M.J.; Price, R.G.; Yancey, S.W.; Ortega, H.G. Long-term Efficacy and Safety of Mepolizumab in Patients With Severe Eosinophilic Asthma: A Multi-center, Open-label, Phase IIIb Study. Clin. Ther. 2016, 38, 2058–2070. [Google Scholar] [CrossRef] [Green Version]
  214. Bel, E.H.; Wenzel, S.E.; Thompson, P.J.; Prazma, C.M.; Keene, O.N.; Yancey, S.W.; Ortega, H.G.; Pavord, I.D.; Investigators, S. Oral glucocorticoid-sparing effect of mepolizumab in eosinophilic asthma. N. Engl. J. Med. 2014, 371, 1189–1197. [Google Scholar] [CrossRef] [PubMed]
  215. Markham, A. Reslizumab: First Global Approval. Drugs 2016, 76, 907–911. [Google Scholar] [CrossRef]
  216. Mukherjee, M.; Aleman Paramo, F.; Kjarsgaard, M.; Salter, B.; Nair, G.; LaVigne, N.; Radford, K.; Sehmi, R.; Nair, P. Weight-adjusted Intravenous Reslizumab in Severe Asthma with Inadequate Response to Fixed-Dose Subcutaneous Mepolizumab. Am. J. Respir. Crit. Care Med. 2018, 197, 38–46. [Google Scholar] [CrossRef]
  217. Corren, J.; Weinstein, S.; Janka, L.; Zangrilli, J.; Garin, M. Phase 3 Study of Reslizumab in Patients With Poorly Controlled Asthma: Effects Across a Broad Range of Eosinophil Counts. Chest 2016, 150, 799–810. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  218. Markham, A. Benralizumab: First Global Approval. Drugs 2018, 78, 505–511. [Google Scholar] [CrossRef]
  219. FitzGerald, J.M.; Bleecker, E.R.; Nair, P.; Korn, S.; Ohta, K.; Lommatzsch, M.; Ferguson, G.T.; Busse, W.W.; Barker, P.; Sproule, S.; et al. Benralizumab, an anti-interleukin-5 receptor alpha monoclonal antibody, as add-on treatment for patients with severe, uncontrolled, eosinophilic asthma (CALIMA): A randomised, double-blind, placebo-controlled phase 3 trial. Lancet 2016, 388, 2128–2141. [Google Scholar] [CrossRef] [PubMed]
  220. Bleecker, E.R.; FitzGerald, J.M.; Chanez, P.; Papi, A.; Weinstein, S.F.; Barker, P.; Sproule, S.; Gilmartin, G.; Aurivillius, M.; Werkstrom, V.; et al. Efficacy and safety of benralizumab for patients with severe asthma uncontrolled with high-dosage inhaled corticosteroids and long-acting beta2-agonists (SIROCCO): A randomised, multicentre, placebo-controlled phase 3 trial. Lancet 2016, 388, 2115–2127. [Google Scholar] [CrossRef] [PubMed]
  221. Cheraghlou, S.; Cohen, J.M. Early Adoption of Dupilumab in the Medicare Population in 2017. Yale J. Biol. Med. 2020, 93, 675–677. [Google Scholar] [PubMed]
  222. Eschenbacher, W.; Straesser, M.; Knoeddler, A.; Li, R.C.; Borish, L. Biologics for the Treatment of Allergic Rhinitis, Chronic Rhinosinusitis, and Nasal Polyposis. Immunol. Allergy Clin. N. Am. 2020, 40, 539–547. [Google Scholar] [CrossRef]
  223. Berger, P.; Menzies-Gow, A.; Peters, A.T.; Kuna, P.; Rabe, K.F.; Altincatal, A.; Soler, X.; Pandit-Abid, N.; Siddiqui, S.; Jacob-Nara, J.A.; et al. Long-term efficacy of dupilumab in asthma with and without chronic rhinosinusitis and/or nasal polyps. Ann. Allergy Asthma Immunol. 2022, S1081-1206(22)01912-3. [Google Scholar] [CrossRef]
  224. Castro, M.; Corren, J.; Pavord, I.D.; Maspero, J.; Wenzel, S.; Rabe, K.F.; Busse, W.W.; Ford, L.; Sher, L.; FitzGerald, J.M.; et al. Dupilumab Efficacy and Safety in Moderate-to-Severe Uncontrolled Asthma. N. Engl. J. Med. 2018, 378, 2486–2496. [Google Scholar] [CrossRef]
  225. Rabe, K.F.; Nair, P.; Brusselle, G.; Maspero, J.F.; Castro, M.; Sher, L.; Zhu, H.; Hamilton, J.D.; Swanson, B.N.; Khan, A.; et al. Efficacy and Safety of Dupilumab in Glucocorticoid-Dependent Severe Asthma. N. Engl. J. Med. 2018, 378, 2475–2485. [Google Scholar] [CrossRef]
  226. Kychygina, A.; Cassagne, M.; Tauber, M.; Galiacy, S.; Paul, C.; Fournie, P.; Simon, M. Dupilumab-Associated Adverse Events During Treatment of Allergic Diseases. Clin. Rev. Allergy Immunol. 2022, 62, 519–533. [Google Scholar] [CrossRef] [PubMed]
  227. Wenzel, S.; Castro, M.; Corren, J.; Maspero, J.; Wang, L.; Zhang, B.; Pirozzi, G.; Sutherland, E.R.; Evans, R.R.; Joish, V.N.; et al. Dupilumab efficacy and safety in adults with uncontrolled persistent asthma despite use of medium-to-high-dose inhaled corticosteroids plus a long-acting beta2 agonist: A randomised double-blind placebo-controlled pivotal phase 2b dose-ranging trial. Lancet 2016, 388, 31–44. [Google Scholar] [CrossRef]
  228. Hoy, S.M. Tezepelumab: First Approval. Drugs 2022, 82, 461–468. [Google Scholar] [CrossRef] [PubMed]
  229. Menzies-Gow, A.; Corren, J.; Bourdin, A.; Chupp, G.; Israel, E.; Wechsler, M.E.; Brightling, C.E.; Griffiths, J.M.; Hellqvist, A.; Bowen, K.; et al. Tezepelumab in Adults and Adolescents with Severe, Uncontrolled Asthma. N. Engl. J. Med. 2021, 384, 1800–1809. [Google Scholar] [CrossRef] [PubMed]
  230. Kelsen, S.G.; Agache, I.O.; Soong, W.; Israel, E.; Chupp, G.L.; Cheung, D.S.; Theess, W.; Yang, X.; Staton, T.L.; Choy, D.F.; et al. Astegolimab (anti-ST2) efficacy and safety in adults with severe asthma: A randomized clinical trial. J. Allergy Clin. Immunol. 2021, 148, 790–798. [Google Scholar] [CrossRef]
  231. Penagos, M.; Durham, S.R. Allergen immunotherapy for long-term tolerance and prevention. J. Allergy Clin. Immunol. 2022, 149, 802–811. [Google Scholar] [CrossRef] [PubMed]
  232. Cox, L.S.; Murphey, A.; Hankin, C. The Cost-Effectiveness of Allergen Immunotherapy Compared with Pharmacotherapy for Treatment of Allergic Rhinitis and Asthma. Immunol. Allergy Clin. N. Am. 2020, 40, 69–85. [Google Scholar] [CrossRef]
  233. Akkoc, T.; Genc, D. Asthma immunotherapy and treatment approaches with mesenchymal stem cells. Immunotherapy 2020, 12, 665–674. [Google Scholar] [CrossRef] [PubMed]
  234. Gans, M.D.; Gavrilova, T. Understanding the immunology of asthma: Pathophysiology, biomarkers, and treatments for asthma endotypes. Paediatr. Respir. Rev. 2020, 36, 118–127. [Google Scholar] [CrossRef]
  235. Rondón, C.; Campo, P.; López-Blanca, N.; Torres, M.J.; Blanca, M. Local Allergic Rhinitis: Is There a Role for Systemic Allergy Immunotherapy? Curr. Treat. Options Allergy 2015, 2, 54–63. [Google Scholar] [CrossRef] [Green Version]
  236. Kouzegaran, S.; Zamani, M.A.; Faridhosseini, R.; Rafatpanah, H.; Rezaee, A.; Yousefzadeh, H.; Movahed, R.; Azad, F.J.; Tehrani, H. Immunotherapy in Allergic Rhinitis: It’s Effect on the Immune System and Clinical Symptoms. Open Access Maced. J. Med. Sci. 2018, 6, 1248–1252. [Google Scholar] [CrossRef] [Green Version]
  237. Hoshino, M.; Akitsu, K.; Kubota, K.; Ohtawa, J. Efficacy of a house dust mite sublingual immunotherapy tablet as add-on dupilumab in asthma with rhinitis. Allergol. Int. 2022, 71, 490–497. [Google Scholar] [CrossRef] [PubMed]
  238. Fritzsching, B.; Contoli, M.; Porsbjerg, C.; Buchs, S.; Larsen, J.R.; Elliott, L.; Rodriguez, M.R.; Freemantle, N. Long-term real-world effectiveness of allergy immunotherapy in patients with allergic rhinitis and asthma: Results from the REACT study, a retrospective cohort study. Lancet Reg. Health Eur. 2022, 13, 100275. [Google Scholar] [CrossRef]
  239. Roberts, G.; Pfaar, O.; Akdis, C.A.; Ansotegui, I.J.; Durham, S.R.; Gerth van Wijk, R.; Halken, S.; Larenas-Linnemann, D.; Pawankar, R.; Pitsios, C.; et al. EAACI Guidelines on Allergen Immunotherapy: Allergic rhinoconjunctivitis. Allergy 2018, 73, 765–798. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  240. Valenta, R.; Campana, R.; Focke-Tejkl, M.; Niederberger, V. Vaccine development for allergen-specific immunotherapy based on recombinant allergens and synthetic allergen peptides: Lessons from the past and novel mechanisms of action for the future. J. Allergy Clin. Immunol. 2016, 137, 351–357. [Google Scholar] [CrossRef]
  241. Rondon, C.; Blanca-Lopez, N.; Aranda, A.; Herrera, R.; Rodriguez-Bada, J.L.; Canto, G.; Mayorga, C.; Torres, M.J.; Campo, P.; Blanca, M. Local allergic rhinitis: Allergen tolerance and immunologic changes after preseasonal immunotherapy with grass pollen. J. Allergy Clin. Immunol. 2011, 127, 1069–1071. [Google Scholar] [CrossRef] [PubMed]
  242. Klimek, L.; Fox, G.C.; Thum-Oltmer, S. SCIT with a high-dose house dust mite allergoid is well tolerated: Safety data from pooled clinical trials and more than 10 years of daily practice analyzed in different subgroups. Allergo J. Int. 2018, 27, 131–139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  243. Shamji, M.H.; Ceuppens, J.; Bachert, C.; Hellings, P.; Placier, G.; Thirion, G.; Bovy, N.; Durham, S.R.; Duchateau, J.; Legon, T.; et al. Lolium perenne peptides for treatment of grass pollen allergy: A randomized, double-blind, placebo-controlled clinical trial. J. Allergy Clin. Immunol. 2018, 141, 448–451. [Google Scholar] [CrossRef] [Green Version]
  244. Epstein, T.G.; Liss, G.M.; Murphy-Berendts, K.; Bernstein, D.I. Risk factors for fatal and nonfatal reactions to subcutaneous immunotherapy: National surveillance study on allergen immunotherapy (2008-2013). Ann. Allergy Asthma Immunol. 2016, 116, 354–359. [Google Scholar] [CrossRef]
  245. Unal, D. Effects of Perennial Allergen Immunotherapy in Allergic Rhinitis in Patients with/without Asthma: A-Randomized Controlled Real-Life Study. Int. Arch. Allergy Immunol. 2020, 181, 141–148. [Google Scholar] [CrossRef]
  246. Rondon, C.; Blanca-Lopez, N.; Campo, P.; Mayorga, C.; Jurado-Escobar, R.; Torres, M.J.; Canto, G.; Blanca, M. Specific immunotherapy in local allergic rhinitis: A randomized, double-blind placebo-controlled trial with Phleum pratense subcutaneous allergen immunotherapy. Allergy 2018, 73, 905–915. [Google Scholar] [CrossRef]
  247. Calderon, M.A.; Simons, F.E.; Malling, H.J.; Lockey, R.F.; Moingeon, P.; Demoly, P. Sublingual allergen immunotherapy: Mode of action and its relationship with the safety profile. Allergy 2012, 67, 302–311. [Google Scholar] [CrossRef]
  248. Pfaar, O.; Lou, H.; Zhang, Y.; Klimek, L.; Zhang, L. Recent developments and highlights in allergen immunotherapy. Allergy 2018, 73, 2274–2289. [Google Scholar] [CrossRef] [PubMed]
  249. Virchow, J.C.; Backer, V.; Kuna, P.; Prieto, L.; Nolte, H.; Villesen, H.H.; Ljorring, C.; Riis, B.; de Blay, F. Efficacy of a House Dust Mite Sublingual Allergen Immunotherapy Tablet in Adults With Allergic Asthma: A Randomized Clinical Trial. JAMA 2016, 315, 1715–1725. [Google Scholar] [CrossRef]
  250. Valovirta, E.; Petersen, T.H.; Piotrowska, T.; Laursen, M.K.; Andersen, J.S.; Sorensen, H.F.; Klink, R.; GAP Investigators. Results from the 5-year SQ grass sublingual immunotherapy tablet asthma prevention (GAP) trial in children with grass pollen allergy. J. Allergy Clin. Immunol. 2018, 141, 529–538. [Google Scholar] [CrossRef]
  251. Elliott, J.; Kelly, S.E.; Johnston, A.; Skidmore, B.; Gomes, T.; Wells, G.A. Allergen immunotherapy for the treatment of allergic rhinitis and/or asthma: An umbrella review. CMAJ Open 2017, 5, 373–385. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  252. Kim, J.Y.; Jang, M.J.; Kim, D.Y.; Park, S.W.; Han, D.H. Efficacy of Subcutaneous and Sublingual Immunotherapy for House Dust Mite Allergy: A Network Meta-Analysis-Based Comparison. J. Allergy Clin. Immunol. Pract. 2021, 9, 4450–4458. [Google Scholar] [CrossRef] [PubMed]
  253. Konradsen, J.R.; Grundstrom, J.; Hellkvist, L.; Tran, T.A.T.; Andersson, N.; Gafvelin, G.; Kiewiet, M.B.G.; Hamsten, C.; Tang, J.; Parkin, R.V.; et al. Intralymphatic immunotherapy in pollen-allergic young adults with rhinoconjunctivitis and mild asthma: A randomized trial. J. Allergy Clin. Immunol. 2020, 145, 1005–1007. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  254. Werner, M.T.; Bosso, J.V. Intralymphatic immunotherapy for allergic rhinitis: A systematic review and meta-analysis. Allergy Asthma Proc. 2021, 42, 283–292. [Google Scholar] [CrossRef]
  255. Skaarup, S.H.; Schmid, J.M.; Skjold, T.; Graumann, O.; Hoffmann, H.J. Intralymphatic immunotherapy improves grass pollen allergic rhinoconjunctivitis: A 3-year randomized placebo-controlled trial. J. Allergy Clin. Immunol. 2021, 147, 1011–1019. [Google Scholar] [CrossRef]
Figure 1. Overview of literature search and the steps performed.
Figure 1. Overview of literature search and the steps performed.
Applsci 13 01273 g001
Figure 2. Diagnostic tools in asthma and AR. Different diagnostic methods can be used in asthma diagnosis, from classical methods (lung function, medical history, skin prick test) to cellular (eosinophil counts, basophil activation test) and molecular (measurements of cytokines and chemokines, and other biomarkers such as miRNAs) novel methods.
Figure 2. Diagnostic tools in asthma and AR. Different diagnostic methods can be used in asthma diagnosis, from classical methods (lung function, medical history, skin prick test) to cellular (eosinophil counts, basophil activation test) and molecular (measurements of cytokines and chemokines, and other biomarkers such as miRNAs) novel methods.
Applsci 13 01273 g002
Figure 3. Therapeutic tools in asthma and AR. Traditionally, asthma and AR have been treated with classical drugs, such as bronchodilators, antihistamines, and corticosteroids. The form of administration of glucocorticoids is inhaled in asthmatic patients, while in patients with AR it is intranasal. However, in the last decades, the development of novel and more specific therapeutic tools such as biologicals and immunotherapy have permitted a more personalized medicine for asthma and AR.
Figure 3. Therapeutic tools in asthma and AR. Traditionally, asthma and AR have been treated with classical drugs, such as bronchodilators, antihistamines, and corticosteroids. The form of administration of glucocorticoids is inhaled in asthmatic patients, while in patients with AR it is intranasal. However, in the last decades, the development of novel and more specific therapeutic tools such as biologicals and immunotherapy have permitted a more personalized medicine for asthma and AR.
Applsci 13 01273 g003
Figure 4. Biologicals used in asthma and AR. Mechanisms of action of different biologics are schematized (omalizumab, mepolizumab, reslizumab, benralizumab, tezepelumab, and dupilumab). Biologicals usually target inflammatory molecules, cytokines, or their receptors, blocking downstream inflammatory pathways or triggering-induced cell death. ADCC: antibody-dependent cell-mediated cytotoxicity; mAb: monoclonal antibody.
Figure 4. Biologicals used in asthma and AR. Mechanisms of action of different biologics are schematized (omalizumab, mepolizumab, reslizumab, benralizumab, tezepelumab, and dupilumab). Biologicals usually target inflammatory molecules, cytokines, or their receptors, blocking downstream inflammatory pathways or triggering-induced cell death. ADCC: antibody-dependent cell-mediated cytotoxicity; mAb: monoclonal antibody.
Applsci 13 01273 g004
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Espada-Sánchez, M.; Sáenz de Santa María, R.; Martín-Astorga, M.d.C.; Lebrón-Martín, C.; Delgado, M.J.; Eguiluz-Gracia, I.; Rondón, C.; Mayorga, C.; Torres, M.J.; Aranda, C.J.; et al. Diagnosis and Treatment in Asthma and Allergic Rhinitis: Past, Present, and Future. Appl. Sci. 2023, 13, 1273. https://doi.org/10.3390/app13031273

AMA Style

Espada-Sánchez M, Sáenz de Santa María R, Martín-Astorga MdC, Lebrón-Martín C, Delgado MJ, Eguiluz-Gracia I, Rondón C, Mayorga C, Torres MJ, Aranda CJ, et al. Diagnosis and Treatment in Asthma and Allergic Rhinitis: Past, Present, and Future. Applied Sciences. 2023; 13(3):1273. https://doi.org/10.3390/app13031273

Chicago/Turabian Style

Espada-Sánchez, Marta, Rocío Sáenz de Santa María, María del Carmen Martín-Astorga, Clara Lebrón-Martín, María Jesús Delgado, Ibón Eguiluz-Gracia, Carmen Rondón, Cristobalina Mayorga, María José Torres, Carlos José Aranda, and et al. 2023. "Diagnosis and Treatment in Asthma and Allergic Rhinitis: Past, Present, and Future" Applied Sciences 13, no. 3: 1273. https://doi.org/10.3390/app13031273

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

Article Metrics

Back to TopTop