Abstract
Allergic diseases are highly complex with respect to pathogenesis, inflammation, and response to treatment. Current efforts for allergic disease diagnosis have focused on clinical evidence as a binary outcome. Although outcome status based on clinical phenotypes (observable characteristics) is convenient and inexpensive to measure in large studies, it does not adequately provide insight into the complex molecular determinants of allergic disease. Individuals with similar clinical diagnoses do not necessarily have similar disease etiologies, natural histories, or responses to treatment. This heterogeneity contributes to the ineffective response to treatment leading to an annual estimated cost of $350 billion in the USA alone. There has been a recent focus to deconvolute the clinical heterogeneity of allergic diseases into specific endotypes using molecular and omics approaches. Endotypes are a means to classify patients based on the underlying pathophysiological mechanisms involving distinct functions or treatment response. The advent of high-throughput molecular omics, immunophenotyping, and bioinformatics methods including machine learning algorithms is facilitating the development of endotype-based diagnosis. As we move to the next decade, we should truly start treating clinical endotypes not clinical phenotype. This review highlights current efforts taking place to improve allergic disease endotyping via molecular omics profiling, immunophenotyping, and machine learning approaches in the context of precision diagnostics in allergic diseases.
Keywords: Allergic diseases, Endotype, Molecular profiling, Big data, Multi-omics, Machine learning
Introduction
Allergic diseases (asthma, atopic dermatitis (AD), allergic rhinitis (AR), food allergy (FA), and eosinophilic esophagitis (EoE)) are conditions caused by inappropriate immunological responses to normally harmless antigens. Collectively, they affect more than 50 million Americans each year making them the sixth leading cause of chronic illnesses in the USA [1, 2]. Although we may observe organ-specific symptoms, allergic diseases are also systemic and are caused by partially shared etiologies. Individuals who have one allergic condition are more likely to develop other allergic conditions and the triggers for each may be shared. Such co-occurrence of allergic conditions in children has been estimated to be ~10-fold higher than one would expect by chance [3–5], suggesting a shared etiology or comorbidity caused in part by a process known as the allergic march (Fig. 1). While specific individuals may not have every disorder in the allergic march, the sequence starts with AD often in infancy, followed by FA and ending with AR or asthma [6, 7]. Furthermore, patients with AD, FA, asthma, or AR are at increased risk of being diagnosed with EoE [8]. The relationship between FA and EoE seems to be particularly strong, with FA patients developing EoE almost nine times more often than their healthy peers [9, 10]. Further, the risk relationship between EoE and AR seems to be bidirectional, with each condition imparting risk for the subsequent diagnosis of the other [8]. As a result of these various comorbidities, although a rare occurrence compared with other atopic diseases, EoE has been proposed as a fifth member of the allergic march [8, 11].
Fig. 1.

Schematic diagram illustrating progression, coexistence, and comorbidity of allergic diseases commonly known as “allergic march” in a single individual. FA, AD peak in the early years of life and decrease after a while. Asthma and AR increase over time as sensitization develops further
Traditional classification of allergic diseases is typically based on severity, which relies on some form of “symptom score,” which is used as an indirect measure of risk and/or impairment. While this may be convenient and inexpensive to measure, it is a poor surrogate into the complex molecular determinants underlying allergic disease. Other prior attempts at classification of allergic disease used binary clinical outcomes such as atopy versus non-atopy implying suspected differences in underlying mechanisms. However, atopy is classically used to represent either genetic susceptibility or excessive immunoglobulin E (IgE) production regardless of whether this sensitization is contributory [12]. The problem is that atopy is likely too broad a category, and sensitization alone is unlikely to represent a true allergic endotype [12–15]. Notably, allergen-specific serum IgE and skin prick test (SPT) can assist in allergic diagnoses, but their diagnostic specificity and sensitivity remain limited. Indeed, for multiple allergic conditions, a detailed clinical history, additional provocation testing/challenges, or response to standard therapy together must guide diagnosis and treatment [16, 17]. Such heterogeneity, lack of robust biomarkers, and inadequate treatments warrant further research into the mechanisms underlying allergic diseases [17, 18].
Mechanism-based endotype classification can be different from patient to patient. Allergy endotypes are constantly evolving, and it is currently defined by the presence of a biologically tractable distinct pathophysiological mechanism, whereas phenotype is an observable physical characteristic or binary outcome of disease without any implication of a mechanism [19–22]. Endotypes found in a specific biological pathway that explains the observable properties of a phenotype could provide an improved understanding of the distinct mechanisms driving the disease and advance precision medicine [23]. A natural result of these traditional classifications of allergic disease is that multiple molecular endotypes have been clustered together into a single phenotype (Fig. 2). Such oversimplification of phenotypes creates a confounding effect which hampers the efficacy of targeted treatment [24, 25]. As the development of targeted therapies has advanced, we are learning more about how molecular mechanisms play a role in phenotype definitions. Nowhere was this more apparent than with the initial trials of anti-IL-5 biologics in the treatment of asthma. The initial trials utilizing mild or moderate asthmatics (defined by severity) were a failure [26, 27], but when the target population was refined to the endotype of glucocorticoid-dependent eosinophilic asthma, the clinical studies were successful, leading to the approval of the first of a new class of asthma medicines in 12 years [28, 29].
Fig. 2.

Traditional methods of allergic disease diagnosis followed by cluster analysis leading patient classification with different endotypes and response to treatment
This trend has continued with further development of cytokine-specific therapies, though there remains a large degree of variability in patient treatment response. This not only highlights the importance of molecular pathways in defining endotypes, but contributes to the rising cost of drug development with a current estimate of $2.6 billion per drug [30, 31]. Thus, clinical phenotyping has been lagging behind other areas of medicine, mostly relying on data collection using self-reported questionnaires and interviews. Biologic data is only as good as the associated clinical data collected. Given that there is a significant degree of heterogeneity between phenotypes and endotypes, it becomes very difficult to collect accurate clinical information in large populations that allows for selection of the most suitable treatment approaches for patients presenting with a spectrum of allergic disorders [20]. Such heterogeneity hinders the successful management of allergic diseases in the clinic as well.
The increased recognition among the scientific community that allergic diseases are comprised of a spectrum of different diseases with distinct clinical presentations and pathogenic mechanisms, along with the differences in prevalence and morbidity among different ethnic groups in the USA, is improving the way we study allergic conditions [32–37]. It is now clear that the traditional concept of phenotypes are a poor indicator of allergic disease severity and progression. Accurate reclassification of allergic diseases into clinically and biologically homogeneous subtypes could facilitate the understanding of disease pathophysiology and development of more targeted interventions [38]. A single phenotype may encompass multiple endotypes and such distinct endotypes likely have different genetic underpinnings [19]. The emergence of new biologic agents leads the way into the identification of clinically meaningful biomarkers that might help to stratify patients and identify those most likely to experience benefit [21, 39]. Machine learning (ML) provides systemlevel opportunities, giving an improvement over conventional regression models by capturing complex, nonlinear relationships in multi-omics and clinical allergic data [40].
This review will highlight the current methods used to improve clinical phenotyping and molecular profiling of allergic diseases and outline how omics-based endotyping is transforming the traditional concepts of allergy through the discovery of novel endotypes. In addition, we will list the most common examples of ML approaches and new bioinformatics tools being utilized and integrated in clinical applications to study allergic disease endotypes and biomarkers. Finally, we will describe how this new knowledge is being utilized for the discovery and development of new treatment drugs.
Biomarkers for Allergic Diseases
Discovering and studying endotypes require the discovery and development of biological markers (biomarkers) in order to characterize the mechanisms and processes underlying different endotypes. Biomarkers are objectively measurable indicators that can be used to predict or monitor different aspects of a disease, such as risk, severity, clinical profile, and response to treatment. Types of biomarkers include genetic variation, epigenetic modifications, gene expression, protein levels, metabolites, and microbiomes [41–44]. An ideal biomarker will (a) link a disease endotype with its phenotype, (b) be stable or predictable over time, and (c) be detectable among populations with genetically distinct backgrounds [41]. The emergence of new biologic agents brings with it the challenge of identifying clinically meaningful biomarkers that might help stratify patients and identify those most likely to experience benefit [21]. Biomarkers also need to be available from relevant tissues with the least invasive methods possible. Systemic biomarkers relevant to allergy can be obtained from blood, serum, or plasma. Local biomarkers for AD may be available in the skin using tape stripping or skin biopsies. Nasal swabs or lavage, induced sputum, bron-choalveolar lavage (BAL), bronchial biopsies, or exhaled breath condensate can be used to obtain local asthma biomarkers, although bronchial biopsies, BAL, and sputum induction are fairly invasive, can only be performed in specialized medical settings by trained personnel, and may be unsuitable for young children. Biomarkers for allergic diseases have been reviewed extensively elsewhere [45–51].
Two broad asthma endotypes have been characterized based on the presence of type 2 (T2) inflammation. T2 inflammation is associated with eosinophilia and involves type 2 helper T cells (Th2 cells), which secrete the inflammatory cytokines IL-4, IL-5, and IL-13. Most established asthma biomarkers are directed toward the most common asthma endotype which is characterized by T2-high inflammation [52]. Eosinophil counts in peripheral blood have been shown to correlate with asthma severity in children with T2-high asthma, and may predict response to treatment with biologics. Eosinophil numbers in sputum, when available, can act as a predictor of asthma exacerbations [53] and may also predict response to treatment with glucocorticoids [54]. Total and allergen-specific IgE are markers for allergic asthma which assess atopy. The extracellular matrix protein periostin has been associated with T2-high asthma and has been suggested as a biomarker of T2-high inflammation in adult asthma patients [55]. Very few biomarkers for the less studied T2-low (or T1) asthma phenotypes are currently in use. T2-low asthma is often associated with neutrophilic inflammation, and high levels of IL-17 have been detected in neutrophilic asthma [52]. Several biomarkers have been suggested for the monitoring of AD severity. Serum levels of thymus- and activation-regulated chemokine (TARC/CCL17) have been reported to strongly correlate with disease activity [56], and among other potential monitoring biomarkers are IL-18 [57, 58], lactate dehydrogenase activity [59], and the chemokine +CTACK/ CCL27 [60]. Recently, genetic variants have been identified that are specifically associated with the atopic march-like phenotype of AD + asthma comorbidity [61, 62], suggesting that such comorbidity constitutes an allergic endotype that may be predicted using biomarkers.
Skin prick testing to individual allergens has long been used to diagnose allergic rhinitis [63]. In addition to skin prick testing, TARC/CCL17 in nasal secretions may be used to distinguish allergic and nonallergic rhinitis [64]. Skin prick testing together with oral food challenge is used to diagnose food allergy. The basophil activation test and the more recently developed mast cell activation test have shown potential for distinguishing between food allergy and mere sensitization without the need for oral food challenge. Table 1 provides examples of established and emerging biomarkers for major allergic disorders, many of which may be of use for phenoand endotyping. The table is not exhaustive, and the extensive literature on allergy biomarkers contains many reports of suggested biomarkers that may become useful in the future.
Table 1.
Established and emerging biomarkers for major allergic disorders
| Disease | Source | Marker | Phenotype/outcome | Reference(s) |
|---|---|---|---|---|
| Asthma | Blood, serum | Serum IgE | [65] | |
| Blood eosinophils | T2-high asthma, lung function | [51] | ||
| Serum periostin | T2-high asthma | [54] | ||
| Urine | LTE4 | Asthma severity, aspirin-sensitive asthma, | [66] | |
| susceptibility to leukotriene receptor antagonists | ||||
| Metabolomic profile | asthma severity, corticosteroid-resistant asthma, | [67–69] | ||
| early-onset asthma | ||||
| Exhaled breath | Volatile organic compounds | Eosinophilic asthma, neutrophilic asthma, | [70, 71] | |
| persistent asthma | ||||
| Fractional exhaled nitric oxide | Eosinophilic airway inflammation, response | [72] | ||
| to treatment | ||||
| Sputum | Eosinophils | T2-high asthma, asthma severity, lung function, | [51–53] | |
| predictor of exacerbations, response to treatment | ||||
| AD | Blood, serum | Serum TARC/CCL17 | AD severity | [56] |
| DNA | Filaggrin genotype | Screening and prognostic biomarker for | [55] | |
| AD risk, AD severity, early onset AD | ||||
| Skin | Transcriptome profile | Treatment response | [73] | |
| Microbiome profile | AD severity | [74] | ||
| Allergic rhinitis | Skin prick test | Allergen sensitization | Diagnosis, distinguishes allergic from | [63] |
| nonallergic rhinitis | ||||
| Nasal secretions | TARC/CCL17, endothelin-1 | Distinguishes allergic from nonallergic rhinitis | [75] | |
| Nasal lavage following | Eosinophils, IL-5, IL-6, macrophage | Diagnosis, monitoring of treatment | [76] | |
| nasal allergen challenge | inflammatory protein | |||
| Food allergy | Skin prick test | Allergen sensitization | Diagnosis | [77] |
| Blood, serum | Allergen-specific IgE levels | Diagnosis | [77] | |
| Blood | Basophil activation test | Diagnosis | [78] | |
| Plasma | Mast cell activation | Diagnosis | [79, 80] | |
| Blood | FOXP3 methylation in | Predictive of response to oral immunotherapy | [81] | |
| antigen-induced Treg cells | ||||
Genetic analysis of allergic diseases, including candidate gene studies, genome-wide linkage studies within pedigrees, and genome-wide association studies (GWAS), can provide useful biomarkers for endotyping of allergic diseases with an estimated heritability ranging from 35 to 95% [24, 82]. In the last 12 years, GWAS have identified loci shared across multiple allergic phenotypes, including asthma, allergic rhinitis, atopic dermatitis and food allergy. IL13, IL4, IL33, and TSLP are related to the “T2” immune response of type 2 helper T cells (Th2), and type 2 innate lymphoid cells (ILC2) are biologically linked with immune function and implicated in allergy [82–85]. There are some loci that do appear to act specifically for certain asthma phenotypes and European ancestry. The chromosomal 17q12–21 locus has been associated with childhood asthma [86]. In particular, ORMDL3/GSDMB/LRRC3C is linked to childhood-onset asthma [87–89]. Loci such as TLR1/TLR6 and ADAD1/IL2 are linked to Th17-related mechanisms. GWAS have successfully identified numerous FA association loci in European ancestry [90–93]. Recently, there has been a focus on ethnicity-specific effects. For instance, PYHIN1 is significantly associated with asthma, but only in individuals of African ancestry [88]. There is also an increasing focus on using admixture to map risk loci [24]. There is a strong heritable component to AD, and several candidate genes have been identified using association studies [94]. Filaggrin (FLG) mutations are found in 25–50% of AD patients [95] and are associated with severity and age of onset [96]. However, not all FLG mutation carriers develop AD [97], and FLG mutations alone are not sufficient to define a specific endotype.
Until recently, biomarkers for allergy were developed using hypothesis-based approaches focusing mainly on the abnormal immune responses to allergens. High-throughput omics technologies such as genomics, transcriptomics, epigenomics, proteomics, metabolomics, and microbiome analysis allow the screening for biomarkers in an unbiased fashion [98]. Biomarkers derived from an individual “omics” layer may not be sufficient to define all endotypes, underscoring the need for multi-omics approaches to achieve an integrated systems-level view of disease pathogenesis and endotypes [39]. The integration of endotypes, biomarker information, and data from omics technologies is creating large-scale biological databases [41]. These large-scale databases require strong computational power and new programing approaches to help sort and improve the prediction of outcomes for disease. Machine learning (ML) provides such opportunities, giving an improvement over conventional regression models by capturing complex, nonlinear relationships in multi-omics and clinical allergic data [40].
Immunophenotyping and Allergic Endotypes
Immunophenotyping has an important role in elucidating host immune responses. It is the analysis of heterogeneous populations of cells for the purpose of identifying the presence and proportions of cells in the populations of interest. Recent advances in RNA-sequencing and single-cell RNA-sequencing (scRNA-seq) technologies allow the detection of subpopulations that play important roles in host immune responses including the internal environment (obesity, co-morbidities, biome, etc) and external triggers (exposures, diet, infections, etc). ScRNA-seq studies have demonstrated that immune cells exhibit vastly heterogeneous expression profiles within seemingly homogeneous cell populations [99]. The immune response is affected by factors including immune predisposition based on immunologic history (including infection history), race, age, gender, and season. Immunophenotyping using cytometry by time-of-flight (CyTOF) has become the method of choice in identifying and sorting cells within complex populations, such as the analysis of immune cells in a blood sample [100, 101]. CyTOF profiles the immune cells in humans and identifies subpopulations and endotypes that are altered in allergic conditions[100, 101].Databases like the Immunology Database and Analysis Portal (IMMPORT) are readily available to explore immune phenotypes [102].
Leveraging Omics for Allergy Endotyping
Since molecular phenotypes are assumed to be downstream of genetic variants in the causal pathway such as gene expression, epigenetics,proteomics, or metabolomics data, an understanding of the causal link needs to be established. Currently, efforts are underway to deconvolute distinct endotypes using molecular profiling approaches (e.g., gene expression or epigenetics) to make therapeutic decisions feasible. For example, traditional diagnosis of AD depends on obtaining the patient’s family history and visual assessment of the skin, as there is no specific laboratory test clinically available for confirming a diagnosis or assessing likelihood of treatment outcomes. With the advances in high-throughput platforms for transcriptomic analyses, many gene expression datasets are regularly deposited on publicly accessible repositories such as the NCBI Gene Expression Omnibus (GEO). GEO currently hosts over 90,000 datasets comprising over two million samples and presents an enormous opportunity for data mining across multiple studies (https://www.ncbi.nlm.nih.gov/geo/). These genome-wide expression studies identify differentially expressed genes (DEGs) for diagnosis and molecular profiling of the disease condition and to evaluate treatment outcomes. The expression of selected/prioritized DEGs can be readily quantified and compared between disease or treatment states for quick, accurate, and cost-effective diagnosis. Gene expression profiling has been widely applied as a tool for definitive diagnosis for many complex diseases like cancers [103–106] as well as several autoimmune and inflammatory diseases [107–112].
Transcriptomes from whole blood, neutrophils, CD4+ T cells, lung and airway tissues, airway smooth muscle cells, induced sputum, and nasal lavage fluids have been studied in asthma [113]. For example, genome-wide profiling of bronchial epithelial brushings in individuals can distinguish between T2-high and T2-low asthma endotypes [114, 115]. Th2-high and Th2-low phenotypes have shown differential responses to available therapies, whereby patients with nonTh2-driven asthma may be less responsive to glucocorticoids compared with a predominantly Th2-high disease phenotype [114]. T2-high asthma is associated with high levels of eosinophils in sputum and peripheral blood. Eosinophilic asthma is the most common endotype found among severe asthmatics and is thought to be more glucocorticoid responsive. In contrast, neutrophilic asthma is marked by elevated peripheral blood neutrophils (> 7000 cells/μL in patients ages 6 to 12 years, or > 8000 cells/μL for those 12 years and older) and is commonly steroid nonresponsive [14, 116]. From 2003 to 2015, only one biologic was approved for the treatment of moderate to severe asthma in the USA. Since 2015, four new asthma biologics were approved by the US (FDA) [117]. Currently, asthma biologics cost US $30,000 or more annually [118].
Individuals with high levels of IL-5 and eosinophilic inflammation (determined by blood eosinophil count) are more likely to respond to mepolizumab, reslizumab, and benralizumab in addition to the IL-4alpha monoclonal antibody, dupilumab, which have been approved as add-on therapy for the management of severe eosinophilic asthma by the US FDA [119]. Significant strides in our understanding of the pathogenesis, pharmacotherapies, and therapeutic treatment strategies for asthma are made, and five biologic agents to target specific endotypes of inflammatory mediators that are important in the pathogenesis of asthma are now available. The five FDA-approved biologics for the treatment of asthma are as follows: omalizumab, mepolizumab, reslizumab, benralizumab, and dupilumab (Table 2). These targets include IgE (omalizumab), IL-5 (mepolizumab, reslizumab), IL-5 receptor (benralizumab) and IL-4 receptor alpha (dupilumab; shared part of receptor for both IL-4 and IL-13) [121]. Knowledge of a patient’s endotype can inform clinicians’ choice of appropriate therapies [122]. For example, asthmatic patients with eosinophilic airway inflammation respond better to inhaled glucocorticoids than patients without eosinophilic asthma [123]. Similarly, patients with the allergic asthma endotype, characterized by allergic hypersensitivity to airborne allergens, have been shown to respond better to omalizumab therapy than patients with other asthma endotypes.
Table 2.
Biologic agent for treating asthma [120]
| Agent | Mechanism of action | FDA-labeled indications | Route of application | Dosing | Comments |
|---|---|---|---|---|---|
| Xolair (omalizumab, Genentech) | Binds to IGE, preventing binding to mast and other inflammatory cells and subsequent inflammatory mediator release | IgE-mediated allergic asthma, not controlled ICS ≥ 6 years of age | SC | Determined by IgE level and body weight Every 2 to 4 weeks | Preferred biologic agent for T2 high allergic asthma Administered in clinic setting |
| Nucala (mepolizumab, GlaxoSmithKline) | Binds to IL-5 | Add-on therapy for severe asthma, eosinophilic phenotype ≥ 6 years of age | SC | Every 4 weeks | Administered in clinic setting |
| Cinqair (reslizumab, Teva Respiratory LLC) | Binds to IL-5 | Add-on therapy for severe asthma, eosinophilic phenotype ≥ 18 years of age | IV | Every 4 weeks | Not labeled for pediatric use Administered in clinic setting |
| Fasenra (benralizumab, AstraZeneca) | Binds to IL-5R subunit on eosinophils and basophils, inducing cell toxicity | Add-on therapy for severe asthma, eosinophilic phenotype ≥ 12 years of age | SC | Every 4 weeks for the first 3 doses, then once every 8 weeks | Administered in clinic setting |
| Dupixent (dupilumab, Regeneran Pharmaceuticals and Sanofi) | Binds to IL-4R alpha, inhibiting Il–4 and Il–13 signaling | Add-on therapy for moderate to -severe asthma, eosinophilic phenotype or CS dependent ≥ 12 years of age | SC | Every 2 weeks | Can be administered at home Avoid concomitant use with live vaccines |
CS, corticosteroid; ICS, inhaled corticosteroid; IgE, immunoglobulin E; IL, interleukin; IV, intravenous; SC, subcutaneous
Recently, we analyzed AD gene expression data generated from multiple independent studies that are publicly available on NCBI GEO database. We identified 89 genes (89 AD-gene signature panel, “89ADGES”) that were consistently up-/ downregulated (AD vs. controls; P < 0.001) in multiple independent transcriptomic datasets [124]. Using a support vector machine, the 89 genes discriminated AD from controls with 98% predictive accuracy [124]. Furthermore, we found that this gene panel consists of gene clusters related to barrier function, inflammation, and lipid metabolism, processes germane to AD [124]. A similar approach has also been successfully used in the diagnosis of EoE, for which no molecular diagnostic tool was previously available [125]. This genome-wide expression profiling work led to the development of an EoE Diagnostic Panel (EDP) that has been used to diagnose EoE, differentiate it from other forms of esophagitis, uncover disease mechanisms, and identify at least three EoE endotypes [125–128].
Epigenetic modifications have also been associated with allergic disease. Several genome-wide methylation studies of allergic disorders have been conducted, and many of them offer the first step for new diagnostic and targeted treatment plans. In one study, methylation patterns in peripheral blood mononuclear cells were characterized in inner city children with persistent atopic asthma compared to healthy controls. They found 81 differentially methylated regions (55 of which remained significant after accounting for cell mixture), most of which were hypo-methylated [129]. Their results suggest that the epigenome is dysregulated in asthmatic patients [129]. Total serum IgE was independently assessed in two different genome-wide methylation studies of Hispanic and European ancestry children [130, 131]. When eosinophil methylation patterns of asthmatics with high IgE and low IgE levels were compared, they found that certain genes were hypo-methylated in patients with high IgE levels compared to those with low IgE levels, suggesting potential therapeutic targets for the different subgroups [131]. More recently, a study examining DNA methylation in cord blood mononuclear cells from children with and without asthma found that the SMAD3 gene promoter was hyper-methylated in asthmatic versus nonasthmatic children (P = 0.005) [132], and these findings were replicated in three different birth cohorts. These types of studies are necessary to connect childhood asthma development to in utero exposures [132], and may help identify early life biomarkers that identify children at high risk for asthma development so prevention measures could be instituted earlier. Profiling molecular and cellular mechanisms that directly contribute to allergy pathogenesis and endotyping is likely to aid the discovery of novel biomarkers and drug targets (Fig. 3).
Fig. 3.

Multi-omics integration representing the use of bioinformatics to integrate omics and generate molecular endotypes for precision diagnostics. The development of multi-omics platform capable of measuring millions of analytes on a single specimen opened the flood gates for molecular phenotyping by vastly increasing the complexity and granularity of molecular data
Proteomics provide a way to characterize the inflammatory state in allergic disease, and this approach has been used to identify asthma endotypes. The U-BIOPRED cohort was used to identify clusters with distinct proteomic signatures that were associated with differential counts of granulocytes in sputum [133]. In a recent study involving 12 patients with severe asthma, protein composition in lung epithelial structures was explored using data-independent acquisition mass spectrometry. Proteome differences were found in patients with stable severe asthma compared with patients with poor symptom control, as well as in metaplastic versus normal epithelium [134]. Proteomics-based approaches have also been used to explore inflammation in AD. A recent study found evidence of an inflammatory and cardiovascular proteomics signature in nonlesional skin of patients with moderate to severe AD [135]. Furthermore, inflammatory proteomics signatures in serum of AD patients have been found to be associated with sensitization to specific allergen classes [136].
Recent studies leveraging human microbiota showed that there are distinct microbiota associated with particular endotypes of allergy. The commensal microbiome on epithelial barrier structures, such as the skin barrier, the gut epithelium, and the upper airways, plays an important role in maintaining homeostatic conditions. There is a symbiotic relationship and a great amount of cross-talk between epithelial barrier tissues, immune cells, and microbes [137, 138], and as part of the wider exposome, the microbiome is also intimately associated with lifestyle factors and a changing environment [138, 139]. Next generation sequencing has enabled the characterization of organ specific microbiomes in health and disease, and changes in the composition of the gut microbiota in infants have been associated with the development of childhood allergies, including food allergy [140, 141], AD development [142, 143], and asthma [138, 144]. It has also been shown that AD is associated with changes in the skin microbiome, including colonization by Staphylococcus aureus and decreased diversity [142, 145, 146]. An increased understanding of the interplay between host and microbes in allergic disease may lead to the development of novel biomarkers for endotyping and disease prediction.
It is well established that prenatal and postnatal environmental factors play a large role in the development of allergic disorders and often interact with genetic variation to modulate the risk of disease. These exposures include physical components of the external environment such as food allergens and aeroallergens, air pollutants, tobacco smoke, diet, drugs, microorganisms, and various chemicals. Furthermore, less specific factors such as climate, social environment, and behavioral factors can also modify the risk of allergies [147, 148]. The concept of the exposome encompasses all environmental stimuli that impact an individual from conception and onwards [149]. It is often defined to include the internal environment in the body, including oxidative stress, inflammation, and specific biomarkers that result from external exposures [150]. A better and more complete understanding of the way the environment interacts with other factors to promote the development of allergic disease holds particular promise for the area of preventive intervention. Among ongoing initiatives with relevance to the field of allergy is the Human Early Life Exposome (HELIX) project, which involves six existing European birth cohorts [151]. HELIX aims to obtain comprehensive, longitudinal pre- and postnatal data in order to characterize the early-life exposome and explore associations with health outcomes, including allergy and asthma. EXPOsOMICS is a European project that aims to assess exposure to high priority environmental pollutants mainly in air and water [152], whereas the Health and Environment-wide Associations based on Large population Surveys (HEALS; www.heals-eu.eu) aims to disentangle the influence of the exposome from genetic effects using monozygotic twin data. The Exposome-Explorer (http://exposome-explorer.iarc.fr) is a database with extensive information about biomarkers associated with exposures to environmental risk factors. The European multicohort study of allergies known as Mechanisms of the Development of Allergy (MeDALL), integrating 14 European birth cohorts that included 44,010 participants and 160 cohort follow-up study from pregnancy to age 20 [153], is a great example to advance this field through characterization of the exposome within multiple time windows, development of exposure assessment and analytic methods, and examination of exposome–response relationships related to a number of health end points [154]. In the USA, the National Institute of Environmental Health Sciences (NIEHS)has supported efforts to advance the field of exposome within environmental health research [155]. The NIEHS has established an infrastructure to promote the incorporation of the exposome within child health research studies. The Children’s Health Exposure Analysis Resource (CHEAR; https://www.niehs.nih.gov/research/supported/exposure/chear/) includes a network of laboratories with extensive analytic abilities for exposure assessment and measures of biologic response in a variety of biological samples. Although there are tremendous opportunities, exposomics is a young area of research, and challenges to be overcome include extremely complex statistics, exposure misclassification, and the limits of exposure assessment technology [156]. Table 3 shows omics cost, maturity of the technology, and applications.
Table 3.
Omics technology status, diagnostic, prognostic and therapeutic applications
| Sample type | Cost to assay | Ease of obtaining material | Sample size | Condition/context dependent | Relationship to endotype/phenotype | Completeness of assay | Maturity of analysis/technology | Potential applications |
|---|---|---|---|---|---|---|---|---|
| DNA | $+ | +++ | +++++++ | No | + | +++ | +++ | Screening, prognostic, predictive |
| RNA | $$ | + to ++ | ++(++) | Yes | ++ | +++ | ++ | Prognostic |
| Methylation | $+ | +++ | +++(++) | Yes | + to ++ | +++ | ++ | Screening, prognostic |
| Proteomics | $$+ | + to ++ | ++(+) | Yes | +++ | + | + | Monitoring, prognostic |
| Metabolomics | $$+ | + to ++ | ++(+) | Yes | +++ | + | + | Monitoring |
| Microbiome | $$+ | + to ++ | ++(++) | Yes | +++ | + | + | Screening, treatment |
| Exposome | $$+ | + to ++ | ++(++) | Yes | +++ | + | + | Prevention |
| cyTOF | $$+ | + to ++ | ++(+) | Yes | +++ | + | + | Monitoring, predictive |
| scRNA-seq | $$+ | + to ++ | ++(+) | Yes | +++ | + | + | Predictive/treatment |
From Omics to Multi-omics
The promise of the genome-scale multi-omics molecular phenotyping approach to decipher the endotypes of allergic diseases has been well-described, and several investigators have proposed potential biomarkers and endotypes using omics resources [39, 56, 157–159]. However, although the role of “multi-omics-level” studies have been very useful in understanding the mechanism of allergic diseases manifestation, current analyses are primarily based on individual omics data, and there have not been sufficient attempts to integrate multi-omics data including machine learning methods in allergy endotyping [160]. Each omics approach can capture one of many dimensions underlying allergic pathology. Recent high throughput next-generation sequencing technologies now make it possible to sequence and profile hundreds of thousands of biomarkers across the human genome. As a result, the development of multiple omic platforms capable of measuring millions of analytes on a single specimen opened the flood gates for molecular phenotyping by resolving the complexity and granularity of molecular data in allergic research (Fig. 4). Deep phenotyping into endotypes, multi-omics profiling, and ML methodology will bring allergic diseases to a new era of precision diagnosis [161].
Fig. 4.

Roadmap from genome-wide genotype to molecular and cellular phenotypes to endo-phenotype and diseases as influenced by environmental risk factors
MeDALL combined omics data that included GWAS, DNA methylation, targeted multiplex biomarker, and transcriptomic studies to define novel phenotypes and endotypes of allergic diseases. The multi-omics and longitudinal data were able to identify comorbidity of AD, AR, and asthma regardless of IgE sensitization [5], and two distinct phenotypes of monosensitization and polysensitization in allergies associated with specific IgE [5, 153]. The MeDALL project is an example of how multilayered integration of classic data collection and new and advanced analyses can come together to improve diagnosis of allergic diseases. Another resource is the Unbiased Biomarkers in Prediction of Respiratory Disease Outcomes (U-BIOPRED). Studies from U-BIOPRED cohort suggest multiple asthma endotype clusters based on transcriptomics data [162–167]. Recent studies have tried to understand the endotypes of pulmonary function impairment and their relationship with allergic diseases. A recent study by Kelly et al. used an integrative approach of transcriptomics and metabolomics to study pulmonary function in children with asthma [168]. Their approach involved using a weighted gene co-expression network analysis (WGCNA) to identify modules of co-regulated gene transcripts and metabolites (specifically 25,060 gene probes and 8185 metabolite features) from 325 children with asthma from the Genetic Epidemiology of Asthma in Costa Rica study. They found three gene modules and six metabolite modules associated with pulmonary function at p < 0.05. When they integrated the gene and metabolite modules, they were able to link FEV1/FVC ratio with ORMDL3 and dysregulated lipid metabolism. This study is one of the first studies to integrate omics to assess pulmonary function in asthma [168]. Using both microbiome and metabolomics data, a recent study identified intestinal microbial-derived sphingolipids that are inversely associated with food allergy [169]. Other studies have integrated genetic and metabolomic data using networks and identified biological relationships between an asthma gene, ORMDL3, lipid metabolism, and asthma phenotypes [170]. Additional examples include a multi-omics approach involving transcriptomics and metabolomics that characterized altered platelet function in severe food-associated respiratory allergy [171], and an ongoing study that aims to explore associations between AD and obesity by integrating metabolome, immune biomarkers, and microbiome biomarkers [172].
Machine Learning and Allergic Disease Diagnosis
ML approaches can use computer algorithms to identify homogeneous patterns with similar underlying biologic mechanisms of allergic diseases and are based on systematically collected large molecular profile data along with clinical metadata. There are many commonly used supervised and unsupervised ML algorithms such as latent class, random forest, support vector machine, naïve Bayes, K-means, decision tree, etc [173]. These methodologies can be employed to identify at-risk patients for which good risk prediction methods do not exist or are found to have relatively poor performance.
ML methods such as latent class analysis (LCA)—an unsupervised statistical method developed to identify a small set of underlying subgroups [174]—are an alternative to the traditional approaches where one or more disease phenotype is used to compare treatment response. Recent studies on allergy classification and diagnosis have taken advantage of LCA and have made progress in clustering disease phenotypes that will potentially represent more accurately defined disease endotypes. For example, the Wayne County Health, Environment, Allergy and Asthma Longitudinal Study (WHEALS) used LCA to characterize four different phenotypes in food allergy: (1) low to no sensitization, (2) highly sensitized, (3) milk and egg dominated sensitization, and (4) peanut and/or inhalant allergen—no milk sensitization. Total IgE levels showed variations among the groups [175]. Doctor-diagnosed AD and asthma (at age 4 years) also varied according to specific IgE levels. The highly sensitized group presented with the highest rates of diagnosis, and the low to no sensitized group showed the lowest rates of diagnosis. Data from this study suggests that the use of LCA, rather than the traditional definition of atopy for IgE presence (allergen-specific IgE), better identifies children with allergic disease diagnosis [175]. LCA was also used in the Protection Against Allergy Study in Rural Environment (PASTURE), a European-based birth cohort [176]. They found four phenotypes for eczema, two of them before age onset 2 years, namely: early transient, and early persistent; the late phenotype with onset 2 years or older, and finally the never/infrequent phenotype. Using multivariate logistic regressions, they found that early phenotypes were strongly associated with food allergy. Asthma diagnosis at age 6 years and allergic rhinitis were only associated with the late phenotype [176]. A very similar study was carried out using the Canadian Healthy Infant Longitudinal Development (CHILD) study [177]. They used LCA to investigate the patterns of sensitization and AD from infancy to age 3. The study yielded five distinct classes of phenotypes, and similar to the PASTURE study, children with persistent sensitization appeared to be prone to other allergic disease diagnosis with age [177]. These studies are examples of how ML can yield advancement not only in allergic disease prediction and diagnosis, but also in clinical care. Stratification of AD endotypes on the basis of allergen sensitization profiles by Leonard et al. [136] further reveals differential systemic inflammatory profiles and anti-Staphylococcus aureus toxin antibody levels, highlighting the need to further characterize allergen-specific responses in AD patients, since adequate therapeutic intervention may require specific treatment of systemic inflammatory components in addition to microbial responses.
Another ML method was used to predict asthma on the basis of response to controller medications in the Childhood Asthma Management Program (CAMP) study [178]. The authors of the study used an algorithm called predictor pursuit (PP), a method that has two capabilities: first, it iteratively discovers phenotypes in a dataset until there are no statistical differences that lead to further division, and second, it predicts clinical outcome on the basis of all available features (variables) sequentially dividing the feature space until there are no prediction performance improvements that can lead to further divisions [178]. PP identified four groups of phenotypes overall, namely allergic not obese (A+/O−), obese not allergic (A−/O+), allergic and obese (A+/O+), and not allergic not obese (A−/O−). Obesity was a factor for the effect of two control medications, nonobese children treated with budesonide instead of nedocromil had well-controlled asthma, whereas the opposite was true for obese children [178]. Pulmonary function impairment is recognized as an important endotype for asthma and AR diseases [179]. A recent study by the National Heart, Lung, and Blood Institute Severe Asthma Research Program (NHBLI-SARP) cohort indicated the predictive role of pulmonary function test in asthma [180, 181]. Pulmonary function tests include forced vital capacity (FVC, a measure of lung size), forced expiratory volume in 1 s (FEV1, a standard measure of lung function), and FEV1/FVC ratio (a commonly used outcome to assess airway obstruction). As outlined in the Guidelines of Asthma Severity [182], a patient is classified as having “mild” asthma if the FEV1 level is >80%, “moderate” asthma if the FEV1 level is 60 to 80%, and “severe” asthma if the FEV1 level is <60%. ML approaches are being used to explore allergy heterogeneity and to probe the pathophysiological patterns or “endotypes” [183]. Ideally, these large datasets would integrate existing clinical and “omics” data (e.g., genomic, proteomic, lipidomic, and microbiomic).The use of harmonized terminology for patient characteristics and outcomes and of standardized protocols and the open access to repositories for biospecimen collection are of critical importance. The challenge of accessing large sample sizes is that it introduces heterogeneity in cases and controls, thus obscuring findings, the so-called Faustian bargain [184]. A similar problem is the “winner’s curse” [185] where significant results in a genome-wide study tend to exhibit larger effect sizes than what they are in reality.
Omics-Driven Endotype Identification to Enable Allergic Disease Drug Discovery
Prior to the GWAS era, numerous pharmacological approaches including anti-IgE (omalizumab), anti-IL5 (mepolizumab), anti-IL13 (lebrikizumab), anti-IL4R (dupilmab), and anti-IL2RA (daclizumab) antibodies had been trialed for allergy with varying degrees of success [186–189]. Recently, other therapies (antiIL33, IL6R, TSLP, etc) have been tested [186, 190]. Recent high-throughput omics technologies such as genomics, epigenomics, transcriptomics, proteomics, metabolomics, microbiomics and exposomics have enabled the construction of regulatory networks and biological pathway models. Parallel advances in bioinformatics and computational techniques have enabled the integration and interpretation of these exponentially growing data sets and opens the possibility of endotype-based precision medicine for allergic diseases [171, 191]. High-throughput measurements of molecular phenotypes provide an unprecedented opportunity to model cellular processes and their impact on disease. By integrating proteomics and genomics data, proteo-genomics allows for the analysis of the correlation of mRNA and protein pairs across samples, of mutations, post-translational modifications, and signaling pathways, and of correlations of the regulatory effects on RNA and protein expression levels caused by genetic variants (eQTL), microRNAs (miRNAs), and copy number aberrations (CNAs) [192]. The growing availability of biologics for the treatment of allergic diseases makes it increasingly important to accurately characterize endotypes so that therapy can be appropriately prescribed to improve each condition. Understanding the impact of endotypes may ultimately help explain the disease etiology and help drug discovery.
The goals of the omics-based endotype-driven deep phenotyping approach in allergic diseases are to link drug responses with the biological profile and to provide a safe and efficacious drug for a selected patient population. Besides the therapeutic purpose, omics-driven approaches should be able to open new avenues for prevention in high-risk individuals, early diagnosis, and intervention strategies [23, 41, 45, 193–195]. Although there is no pharmacogenetics (PGx) testing for any allergic disease, PGx studies targeting the response to treatment of the main drugs used for asthma treatment (i.e., short- and long-acting β2-antagonists [SABA and LABA, respectively], inhaled corticosteroids [ICSs], and leukotriene modifiers [LTMs]) are being conducted [196]. Several factors involved in the response to drug reaction include host factors and the chemical structure of the drug. A major advance over the last few decades has been the discovery of genetic variants associated with specific drugs to a given allergicdisease[85,197,198].Association of specific HLA genotypes and vancomycin allergy as well as drug reaction with eosinophilia and systemic symptoms (DRESS) has been reported [199].
Recently, Thijs et al. published a retrospective study on the effects of mycophenolic acid (MPA) in 65 patients with severe AD with and without the presence of polymorphisms in the metabolizing enzyme uridine diphosphate-glucuronosyltransferase 1A9 (UGT1A9). Their results showed that 85% of patients with UGT1A9 polymorphisms were nonresponsive to MPA treatment [200]. An additional example of AD therapeutics focused on nemolizumab (CIM331), a humanized antibody against IL-31 receptor A. The IL-31 receptor plays a role in the occurrence of pruritus in AD, and a recent phase 2, randomized, double-blind, placebo-controlled, 12-week trial was conducted on 264 adults with moderate to severe AD who received subcutaneous nemolizumab [201]. This trial showed that nemolizumab significantly improved AD symptoms when compared to placebo by inhibiting the IL31 signaling, but longer studies should be carried out to confirm the results [201]. Similarly, a highly potent and specific inhibitor of human IL-13 activity in cell-based in vitro assay, QAX576, significantly improved intraepithelial esophageal eosinophil counts and dysregulated esophageal disease-related transcripts in adults with EoE; baseline transcripts in the esophagus were associated with responsiveness to therapy [202].
As we embrace the era of precision medicine aiming at developing diagnostics and therapeutics that account for individual’s unique genetic information, lifestyle, and environmental exposure to select the best treatment for their condition, large-scale omics, omics integration, and ML will be crucial for discovering biomarkers and designing therapeutic strategies for allergic diseases. Clinicians will be able to treat the molecular drivers of disease (endotypes), which can be better understood by analyzing a patient’s genetic and gene expression profiles, rather than clinical phenotypes or manifestations. In addition, pathway analysis will also facilitate the prediction of response to therapy and outcome, as well as identify novel therapeutic targets close to disease pathogenesis (Fig. 5). With current and emerging public resources, we can potentially match allergy-associated CpGs against the ChEMBL database (v22.1) (https://www.ebi.ac.uk/chembl/) to identify whether any are targets of approved drugs or drugs in development. Databases of Encyclopedia of DNA Elements (ENCODE), the Ensembl regulatory build, and Functional ANnoTation Of the Mammalian genome (FANTOM) project, the VISTA Enhancer Browser, expression quantitative trait loci (eQTLs), enhancer RNA (eRNA) co-expression, transcription factor (TF) co-expression, and capture Hi-C (CHi-C) are all useful to understand the role of genetic variants in health and diseases. Treatable targets can be identified in gene expression studies that encode proteins with predicted drug targets. We also can use the Ingenuity Pathway Analysis (IPA, www.ingenuity.com) to identify drug targets and upstream regulators of the gene lists. Studies have shown that several of the newly identified genes linked to lung function are treatable targets, highlighting the clinical relevance of an integrative omics approach [203, 204]. The long-term goal or roadmap is to tailor therapies to individual patients depending on their underlying endotype or pattern of pathophysiology (e.g., anti-T2 biologic treatment for allergic asthma; combined anti-Th2/ Th17 treatment for steroid-resistant disease), through the implementation of precision medicine.
Fig. 5.

An interaction and integrative molecular map showing an allergy phenotype (P) mapped to multiple endotypes (Enm) which intern profiling in certain organs and leading cellular pathways (Pathm) interacting with proteins (Pm) and target drugs (Dm) (adopted from Huang et al. [161])
Conclusion
The evolution of our understanding of allergic disease endotypes reflects the interplay of both the clinically recognized phenotypes observed and the technologies and treatments available to study them (Fig. 6). Asthma is an illustrative example of how a broadly defined clinical entity can be gradually subdivided based on pathophysiologically distinct endotypes. The first clinical definition of phenotype heterogeneity for asthma was made by Francis Rackemann in the 1940s [205], and the application of spirometric measurement of airflow limitation with reversible improvement as a recognized diagnosis was made in the 1980s [206]. Although not universally, clinical trials of broad anti-inflammatory therapy in the 1990s were largely successful for asthma. Around this time, researchers recognized the Th2 pathway as particularly relevant for many asthmatics, and broader availability of sequencing and early omics technologies reignited the interest in clinical heterogeneity of asthma [207, 208]. Only after the initially negative results of anti-IgE and anti-IL-5 antibody therapy in larger asthma clinical trials in the 2000s did the field begin to realize the importance of careful focus on targeted endotypes in allergic disease [209]. The last 10 years has brought significant advancement in treatment of allergic disease with the rollout of multiple biologics to target the Th2 pathway. The next 5–10 years will not only see an increased number of targeted therapeutics (especially non-Th2 targeted), but there will also be a large investment in multi-omics integration and the use of machine learning to help recognize patterns in large datasets/cohorts as these new therapeutics enter the field. The challenge of our field in the next 10– 20 years is using the multiple biologic/targeted therapies and the technological advances of multi-omics integration and machine learning to define the nuances of the many atopic endotypes, to set the stage for the next generation of trials in allergic disease.
Fig. 6.

Brief history of allergy phenotype/endotype research
Despite advances made in the understanding, diagnosis, and treatment of allergic diseases and an increasing interest in omics technologies, the search for endotype-specific biomarkers remains an unmet need. Recent efforts are underway to unravel allergy-specific endotypes and environmental exposures which impact the underlying biological systems [210]. One of the most promising sources of information to be used for endotyping of allergic diseases are multi-omics profiling, which now can be generated in real time for clinical use. With the advent of omics-based big data-driven and unbiased approaches to define and characterize endotypes, including more accessible tools for human immunophenotyping, we can now gather detailed molecular information to deconvolute and identify patterns from the data, and gather further insights into the biology of diseases and health states of individual patients [211, 212]. The availability of “omics” data and integrative omics approaches has created unique opportunities to unravel the molecular underpinnings of target endotypes to develop personalized risk stratification and therapy. Statistical methods to integrate multi-omics data are emerging to provide important insights into disease pathophysiology of allergic diseases [213]. As we move forward, accounting for individual’s unique race specific lifestyle and environmental exposure along with clinical and multi-omics-based data-driven ML framework and integration will be crucial for discovering and designing therapeutic strategies for specific allergic endotypes. Such an approach will help to develop precision treatment options tailored to distinct endotypes in allergic diseases [214, 215].
Funding Information
This work was supported by the National Institutes of Health (NIH) grant R01HL132344, as well as in part by NIH R37 AI045898, U19 AI070235, R01 AI057803, R01 DK107502, P30 DK078392 (Gene and Protein Expression Core), the Campaign Urging Research for Eosinophilic Disease (CURED), and the Sunshine Charitable Foundation and its supporters, Denise and David Bunning.
Footnotes
Compliance with Ethical Standards
Conflict of Interest The authors declare that they have no conflict of interest.
References
- 1.CDC, Gateway to Health Communication and Social Marketing Practice. Allergies. https://www.cdc.gov/healthcommunication/toolstemplates/entertainmented/tips/Allergies.html. Accessed Dec 2019.
- 2.Bacarese-Hamilton T, Gray J, Ardizzoni A, Crisanti A (2005) Allergen microarrays. Methods Mol Med 114:195–207 [DOI] [PubMed] [Google Scholar]
- 3.Pols DH, Wartna JB, van Alphen EI, Moed H, Rasenberg N, Bindels PJ, Bohnen AM (2015) Interrelationships between atopic disorders in children: a meta-analysis based on ISAAC questionnaires. PLoS One 10:e0131869. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Bröms K, Norbäck D, Eriksson M, Sundelin C, Svärdsudd K (2013) Prevalence and co-occurrence of parentally reported possible asthma and allergic manifestations in pre-school children. BMC Public Health 13:764. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Pinart M, Benet M, Annesi-Maesano I, von Berg A, Berdel D, Carlsen KC, Carlsen K-H, Bindslev-Jensen C, Eller E, Fantini MP (2014) Comorbidity of eczema, rhinitis, and asthma in IgEsensitised and non-IgE-sensitised children in MeDALL: a population-based cohort study. Lancet Respir Med 2:131–140 [DOI] [PubMed] [Google Scholar]
- 6.Zheng T, Yu J, Oh MH, Zhu Z (2011) The atopic march: progression from atopic dermatitis to allergic rhinitis and asthma. Allergy Asthma Immunol Res 3:67–73 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Hill DA, Spergel JM (2018) The atopic march: critical evidence and clinical relevance. Ann Allergy Asthma Immunol 120:131–137 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Hill DA, Grundmeier RW, Ramos M, Spergel JM (2018) Eosinophilic esophagitis is a late manifestation of the allergic march. J Allergy Clin Immunol Pract 6:1528–1533 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Lin SK, Sabharwal G, Ghaffari G (2015) A review of the evidence linking eosinophilic esophagitis and food allergy. Allergy Asthma Proc 36:26–33 [DOI] [PubMed] [Google Scholar]
- 10.Ram G, Lee J, Ott M, Brown-Whitehorn TF, Cianferoni A, Shuker M, Wang ML, Verma R, Liacouras CA, Spergel JM (2015) Seasonal exacerbation of esophageal eosinophilia in children with eosinophilic esophagitis and allergic rhinitis. Ann Allergy Asthma Immunol 115(224–228):e221. [DOI] [PubMed] [Google Scholar]
- 11.Hill DA, Spergel JM (2018) Is eosinophilic esophagitis a member of the atopic march? Ann Allergy Asthma Immunol 120:113–114 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Simpson A, Tan VY, Winn J, Svensén M, Bishop CM, Heckerman DE, Buchan I, Custovic A (2010) Beyond atopy: multiple patterns of sensitization in relation to asthma in a birth cohort study. Am J Respir Crit Care Med 181:1200–1206 [DOI] [PubMed] [Google Scholar]
- 13.Schoos A-MM, Chawes BL, Rasmussen MA, Bloch J, Bønnelykke K, Bisgaard H (2016) Atopic endotype in childhood. J Allergy Clin Immunol 137:844–851 e844 [DOI] [PubMed] [Google Scholar]
- 14.Casale TB (2017) Biologics and biomarkers for asthma, urticaria, and nasal polyposis. J Allergy Clin Immunol 139:1411–1421 [DOI] [PubMed] [Google Scholar]
- 15.Aaron SD, Boulet LP, Reddel HK, Gershon AS (2018) Underdiagnosis and overdiagnosis of asthma. Am J Respir Crit Care Med 198:1012–1020 [DOI] [PubMed] [Google Scholar]
- 16.Bernstein IL, Li JT, Bernstein DI, Hamilton R, Spector SL, Tan R, Sicherer S, Golden DB, Khan DA, Nicklas RA et al. (2008) Allergy diagnostic testing: an updated practice parameter. Ann Allergy Asthma Immunol 100:S1–S148 [DOI] [PubMed] [Google Scholar]
- 17.Hamilton RG (2014) Allergic sensitization is a key risk factor for but not synonymous with allergic disease. J Allergy Clin Immunol 134:360–361 [DOI] [PubMed] [Google Scholar]
- 18.Bock SA, Sampson HA, Atkins FM, Zeiger RS, Lehrer S, Sachs M, Bush RK, Metcalfe DD (1988) Double-blind, placebo controlled food challenge (DBPCFC) as an office procedure: a manual. J Allergy Clin Immunol 82:986–997 [DOI] [PubMed] [Google Scholar]
- 19.Lotvall J, Akdis CA, Bacharier LB, Bjermer L, Casale TB, Custovic A, Lemanske RF Jr, Wardlaw AJ, Wenzel SE, Greenberger PA (2011) Asthma endotypes: a new approach to classification of disease entities within the asthma syndrome. J Allergy Clin Immunol 127:355–360 [DOI] [PubMed] [Google Scholar]
- 20.Agache I, Akdis CA (2016) Endotypes of allergic diseases and asthma: an important step in building blocks for the future of precision medicine. Allergol Int 65:243–252 [DOI] [PubMed] [Google Scholar]
- 21.Desai M, Oppenheimer J (2016) Elucidating asthma phenotypes and endotypes: progress towards personalized medicine. Ann Allergy Asthma Immunol 116:394–401 [DOI] [PubMed] [Google Scholar]
- 22.Anderson GP (2008) Endotyping asthma: new insights into key pathogenic mechanisms in a complex, heterogeneous disease. Lancet 372:1107–1119 [DOI] [PubMed] [Google Scholar]
- 23.Agache I, Akdis C, Jutel M, Virchow JC (2012) Untangling asthma phenotypes and endotypes. Allergy 67:835–846 [DOI] [PubMed] [Google Scholar]
- 24.Gupta J, Johansson E, Bernstein JA, Chakraborty R, Khurana Hershey GK, Rothenberg ME, Mersha TB (2016) Resolving the etiology of atopic disorders by using genetic analysis of racial ancestry. J Allergy Clin Immunol 138:676–699 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Baye TM, Martin LJ, Hershey GKK (2010) Application of genetic/genomic approaches to allergic disorders. J Allergy Clin Immunol 126:425–436 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Leckie MJ, ten Brinke A, Khan J, Diamant Z, O’Connor BJ, Walls CM, Mathur AK, Cowley HC, Chung KF, Djukanovic R et al. (2000) Effects of an interleukin-5 blocking monoclonal antibody on eosinophils, airway hyper-responsiveness, and the late asthmatic response. Lancet 356:2144–2148 [DOI] [PubMed] [Google Scholar]
- 27.Flood-Page P, Swenson C, Faiferman I, Matthews J, Williams M, Brannick L, Robinson D, Wenzel S, Busse W, Hansel TT et al. (2007) A study to evaluate safety and efficacy of mepolizumab in patients with moderate persistent asthma. Am J Respir Crit Care Med 176:1062–1071 [DOI] [PubMed] [Google Scholar]
- 28.Rothenberg ME (2016) Humanized anti-IL-5 antibody therapy. Cell 165:509. [DOI] [PubMed] [Google Scholar]
- 29.Nair P, Pizzichini MM, Kjarsgaard M, Inman MD, Efthimiadis A, Pizzichini E, Hargreave FE, O’Byrne PM (2009) Mepolizumab for prednisone-dependent asthma with sputum eosinophilia. N Engl J Med 360:985–993 [DOI] [PubMed] [Google Scholar]
- 30.Benson M (2016) Clinical implications of omics and systems medicine: focus on predictive and individualized treatment. J Intern Med 279:229–240 [DOI] [PubMed] [Google Scholar]
- 31.Shalek AK, Benson M (2017) Single-cell analyses to tailor treatments. Sci Transl Med 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Stevenson LA, Gergen PJ, Hoover DR, Rosenstreich D, Mannino DM, Matte TD (2001) Sociodemographic correlates of indoor allergen sensitivity among United States children. J Allergy Clin Immunol 108:747–752 [DOI] [PubMed] [Google Scholar]
- 33.Litonjua AA, Celedon JC, Hausmann J, Nikolov M, Sredl D, Ryan L, Platts-Mills TA, Weiss ST, Gold DR (2005) Variation in total and specific IgE: effects of ethnicity and socioeconomic status. J Allergy Clin Immunol 115:751–757 [DOI] [PubMed] [Google Scholar]
- 34.Celedon JC, Sredl D, Weiss ST, Pisarski M, Wakefield D, Cloutier M (2004) Ethnicity and skin test reactivity to aeroallergens among asthmatic children in Connecticut. Chest 125:85–92 [DOI] [PubMed] [Google Scholar]
- 35.Joseph CL, Ownby DR, Peterson EL, Johnson CC (2000) Racial differences in physiologic parameters related to asthma among middle-class children. Chest 117:1336–1344 [DOI] [PubMed] [Google Scholar]
- 36.Hunninghake GM, Weiss ST, Celedon JC (2006) Asthma in Hispanics. Am J Respir Crit Care Med 173:143–163 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Ramsey CD, Celedon JC, Sredl DL, Weiss ST, Cloutier MM (2005) Predictors of disease severity in children with asthma in Hartford, Connecticut. Pediatr Pulmonol 39:268–275 [DOI] [PubMed] [Google Scholar]
- 38.Muraro A, Lemanske RF Jr, Castells M, Torres MJ, Khan D, Simon HU, Bindslev-Jensen C, Burks W, Poulsen LK, Sampson HA et al. (2017) Precision medicine in allergic disease-food allergy, drug allergy, and anaphylaxis—PRACTALL document of the European Academy of Allergy and Clinical Immunology and the American Academy of Allergy, Asthma and Immunology. Allergy 72:1006–1021 [DOI] [PubMed] [Google Scholar]
- 39.Ghosh D, Bernstein JA, Khurana Hershey GK, Rothenberg ME, Mersha TB (2018) Leveraging multilayered “omics” data for atopic dermatitis: a road map to precision medicine. Front Immunol 9: 2727. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Libbrecht MW, Noble WS (2015) Machine learning applications in genetics and genomics. Nat Rev Genet 16:321–332 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Agache I, Rogozea L (2017) Asthma biomarkers: do they bring precision medicine closer to the clinic? Allergy, Asthma Immunol Res 9:466–476 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Akdis CA, Ballas ZK (2016) Precision medicine and precision health: building blocks to foster a revolutionary health care model. J Allergy Clin Immunol 137:1359–1361 [DOI] [PubMed] [Google Scholar]
- 43.Berry A, Busse WW (2016) Biomarkers in asthmatic patients: has their time come to direct treatment? J Allergy Clin Immunol 137: 1317–1324 [DOI] [PubMed] [Google Scholar]
- 44.Canonica GW, Bachert C, Hellings P, Ryan D, Valovirta E, Wickman M, De Beaumont O, Bousquet J (2015) Allergen immunotherapy (AIT): a prototype of precision medicine. World Allergy Organ J 8:31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Muraro A, Lemanske RF Jr, Hellings PW, Akdis CA, Bieber T, Casale TB, Jutel M, Ong PY, Poulsen LK, Schmid-Grendelmeier P et al. (2016) Precision medicine in patients with allergic diseases: airway diseases and atopic dermatitis—PRACTALL document of the European Academy of Allergy and Clinical Immunology and the American Academy of Allergy, Asthma & Immunology. J Allergy Clin Immunol 137:1347–1358 [DOI] [PubMed] [Google Scholar]
- 46.Zissler UM, Esser-von Bieren J, Jakwerth CA, Chaker AM, Schmidt-Weber CB (2016) Current and future biomarkers in allergic asthma. Allergy 71:475–494 [DOI] [PubMed] [Google Scholar]
- 47.Saglani S (2017) Childhood severe asthma: new insights on remodelling and biomarkers. Paediatr Respir Rev 24:11–13 [DOI] [PubMed] [Google Scholar]
- 48.Richards LB, Neerincx AH, van Bragt JJ, Sterk PJ, Bel EH, Maitland-van der Zee AH (2018) Biomarkers and asthma management: analysis and potential applications. Curr Opin Allergy Clin Immunol 18:96–108 [DOI] [PubMed] [Google Scholar]
- 49.Mansouri Y, Guttman-Yassky E (2015) Immune pathways in atopic dermatitis, and definition of biomarkers through broad and targeted therapeutics. J Clin Med 4:858–873 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Perlikos F, Hillas G, Loukides S (2016) Phenotyping and endotyping asthma based on biomarkers. Curr Top Med Chem 16:1582–1586 [DOI] [PubMed] [Google Scholar]
- 51.Tiotiu A (2018) Biomarkers in asthma: state of the art. Asthma Res Pract 4:10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Licari A, Castagnoli R, Brambilla I, Marseglia A, Tosca MA, Marseglia GL, Ciprandi G (2018) Asthma endotyping and biomarkers in childhood asthma. Pediatr Allergy Immunol Pulmonol 31:44–55 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Hastie AT, Moore WC, Meyers DA, Vestal PL, Li H, Peters SP, Bleecker ER, National Heart L, Blood Institute Severe Asthma Research, P (2010) Analyses of asthma severity phenotypes and inflammatory proteins in subjects stratified by sputum granulocytes. J Allergy Clin Immunol 125:1028–1036 e1013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.van Rensen EL, Straathof KC, Veselic-Charvat MA, Zwinderman AH, Bel EH, Sterk PJ (1999) Effect of inhaled steroids on airway hyperresponsiveness, sputum eosinophils, and exhaled nitric oxide levels in patients with asthma. Thorax 54:403–408 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Carpagnano GE, Scioscia G, Lacedonia D, Soccio P, Lepore G, Saetta M, Foschino Barbaro MP, Barnes PJ (2018) Looking for airways periostin in severe asthma: could it be useful for clustering type 2 endotype? Chest 154:1083–1090 [DOI] [PubMed] [Google Scholar]
- 56.Thijs J, Krastev T, Weidinger S, Buckens CF, de Bruin-Weller M, Bruijnzeel-Koomen C, Flohr C, Hijnen D (2015) Biomarkers for atopic dermatitis: a systematic review and meta-analysis. Curr Opin Allergy Clin Immunol 15:453–460 [DOI] [PubMed] [Google Scholar]
- 57.Aral M, Arican O, Gul M, Sasmaz S, Kocturk SA, Kastal U, Ekerbicer HC (2006) The relationship between serum levels of total IgE, IL-18, IL-12, IFN-gamma and disease severity in children with atopic dermatitis. Mediat Inflamm 2006:73098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Trzeciak M, Glen J, Bandurski T, Sokolowska-Wojdylo M, Wilkowska A, Roszkiewicz J (2011) Relationship between serum levels of interleukin-18, IgE and disease severity in patients with atopic dermatitis. Clin Exp Dermatol 36:728–732 [DOI] [PubMed] [Google Scholar]
- 59.Morishima Y, Kawashima H, Takekuma K, Hoshika A (2010) Changes in serum lactate dehydrogenase activity in children with atopic dermatitis. Pediatr Int 52:171–174 [DOI] [PubMed] [Google Scholar]
- 60.Machura E, Rusek-Zychma M, Jachimowicz M, Wrzask M, Mazur B, Kasperska-Zajac A (2012) Serum TARC and CTACK concentrations in children with atopic dermatitis, allergic asthma, and urticaria. Pediatr Allergy Immunol 23:278–284 [DOI] [PubMed] [Google Scholar]
- 61.Johansson E, Biagini Myers JM, Martin LJ, He H, Pilipenko V, Mersha T, Weirauch M, Salomonis N, Ryan P, LeMasters GK et al. (2017) KIF3A genetic variation is associated with pediatric asthma in the presence of eczema independent of allergic rhinitis. J Allergy Clin Immunol 140(595–598):e595. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Marenholz I, Esparza-Gordillo J, Ruschendorf F, Bauerfeind A, Strachan DP, Spycher BD, Baurecht H, Margaritte-Jeannin P, Saaf A, Kerkhof M et al. (2015) Meta-analysis identifies seven susceptibility loci involved in the atopic march. Nat Commun 6:8804. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Fornadley JA (2014) Skin testing for inhalant allergy. Int Forum Allergy Rhinol 4(Suppl 2):S41–S45 [DOI] [PubMed] [Google Scholar]
- 64.Tsybikov NN, Egorova EV, Kuznik BI, Fefelova EV, Magen E (2016) Biomarker assessment in chronic rhinitis and chronic rhinosinusitis: endothelin-1, TARC/CCL17, neopterin, and alpha-defensins. Allergy Asthma Proc 37:35–42 [DOI] [PubMed] [Google Scholar]
- 65.Sonntag HJ, Filippi S, Pipis S, Custovic A (2019) Blood biomarkers of sensitization and asthma. Front Pediatr 7:251. 10.3389/fped.2019.00251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Hoffman BC, Rabinovitch N (2018) Urinary leukotriene E(4) as a biomarker of exposure, susceptibility, and risk in asthma: an update. Immunol Allergy Clin N Am 38:599–610 [DOI] [PubMed] [Google Scholar]
- 67.Carraro S, Bozzetto S, Giordano G, El Mazloum D, Stocchero M, Pirillo P, Zanconato S, Baraldi E (2018) Wheezing preschool children with early-onset asthma reveal a specific metabolomic profile. Pediatr Allergy Immunol 29:375–382 [DOI] [PubMed] [Google Scholar]
- 68.Park YH, Fitzpatrick AM, Medriano CA, Jones DP (2017) High resolution metabolomics to identify urine biomarkers in corticosteroid-resistant asthmatic children. J Allergy Clin Immunol 139:1518–1524.e1514 [DOI] [PubMed] [Google Scholar]
- 69.Papamichael MM, Katsardis C, Erbas B, Its iopoulos C, Tsoukalas D (2019) Urinary organic acids as biomarkers in the assessment of pulmonary function in children with asthma. Nutr Res 61:31–40 [DOI] [PubMed] [Google Scholar]
- 70.Cavaleiro Rufo J, Paciência I, Mendes FC, Farraia M, Rodolfo A, Silva D, de Oliveira Fernandes E, Delgado L, Moreira A (2019) Exhaled breath condensate volatilome allows sensitive diagnosis of persistent asthma. Allergy 74:527–534 [DOI] [PubMed] [Google Scholar]
- 71.Schleich FN, Zanella D, Stefanuto P-H, Bessonov K, Smolinska A, Dallinga JW, Henket M, Paulus V, Guissard F, Graff S et al. (2019) Exhaled volatile organic compounds are able to discriminate between neutrophilic and eosinophilic asthma. Am J Respir Crit Care Med 200:444–453 [DOI] [PubMed] [Google Scholar]
- 72.Menzies-Gow A, Mansur AH, Brightling CE (2020) Clinical utility of fractional exhaled nitric oxide in severe asthma management. Eur Respir J 26;55(3). 10.1183/13993003.01633-2019 [DOI] [PubMed] [Google Scholar]
- 73.Hamilton JD, Suárez-Fariñas M, Dhingra N, Cardinale I, Li X, Kostic A, Ming JE, Radin AR, Krueger JG, Graham N et al. (2014) Dupilumab improves the molecular signature in skin of patients with moderate-to-severe atopic dermatitis. J Allergy Clin Immunol 134:1293–1300 [DOI] [PubMed] [Google Scholar]
- 74.Kong HH, Oh J, Deming C, Conlan S, Grice EA, Beatson MA, Nomicos E, Polley EC, Komarow HD, Program NCS et al. (2012) Temporal shifts in the skin microbiome associated with disease flares and treatment in children with atopic dermatitis. Genome Res 22:850–859 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Tsybikov NN, Egorova EV, Kuznik BI, Fefelova EV, Magen E (2016) Biomarker assessment in chronic rhinitis and chronic rhinosinusitis: endothelin-1, TARC/CCL17, neopterin, and αdefensins. Allergy Asthma Proc 37:35–42 [DOI] [PubMed] [Google Scholar]
- 76.Badorrek P, Müller M, Koch W, Hohlfeld JM, Krug N (2017) Specificity and reproducibility of nasal biomarkers in patients with allergic rhinitis after allergen challenge chamber exposure. Ann Allergy Asthma Immunol 118:290–297 [DOI] [PubMed] [Google Scholar]
- 77.Oriel RC, Wang J (2019) Diagnosis and management of food allergy. Pediatr Clin N Am 66:941–954 [DOI] [PubMed] [Google Scholar]
- 78.Hemmings O, Kwok M, McKendry R, Santos AF (2018) Basophil activation test: old and new applications in allergy. Curr Allergy Asthma Rep 18:77–77 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Bahri R, Custovic A, Korosec P, Tsoumani M, Barron M, Wu J, Sayers R, Weimann A, Ruiz-Garcia M, Patel N et al. (2018) Mast cell activation test in the diagnosis of allergic disease and anaphylaxis. J Allergy Clin Immunol 142:485–496.e416 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Santos AF, Couto-Francisco N, Bécares N, Kwok M, Bahnson HT, Lack G (2018) A novel human mast cell activation test for peanut allergy. J Allergy Clin Immunol 142:689–691.e689 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Syed A, Garcia MA, Lyu S-C, Bucayu R, Kohli A, Ishida S, Berglund JP, Tsai M, Maecker H, O’Riordan G et al. (2014) Peanut oral immunotherapy results in increased antigen-induced regulatory T-cell function and hypomethylation of forkhead box protein 3 (FOXP3). J Allergy Clin Immunol 133:500–510 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Ober C, Yao TC (2011) The genetics of asthma and allergic disease: a 21st century perspective. Immunol Rev 242:10–30 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Portelli MA, Hodge E, Sayers I (2015) Genetic risk factors for the development of allergic disease identified by genome-wide association. Clin Exp Allergy 45:21–31 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Bonnelykke K, Sparks R, Waage J, Milner JD (2015) Genetics of allergy and allergic sensitization: common variants, rare mutations. Curr Opin Immunol 36:115–126 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Ortiz RA, Barnes KC (2015) Genetics of allergic diseases. Immunol Allergy Clin N Am 35:19–44 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Andiappan AK, Sio YY, Lee B, Suri BK, Matta SA, Lum J, Foo S, Koh G, Liu J, Zolezzi F et al. (2016) Functional variants of 17q1221 are associated with allergic asthma but not allergic rhinitis. J Allergy Clin Immunol 137(758–766):e753. [DOI] [PubMed] [Google Scholar]
- 87.Bonnelykke K, Sleiman P, Nielsen K, Kreiner-Moller E, Mercader JM, Belgrave D, den Dekker HT, Husby A, Sevelsted A, FauraTellez G et al. (2014) A genome-wide association study identifies CDHR3 as a susceptibility locus for early childhood asthma with severe exacerbations. Nat Genet 46:51–55 [DOI] [PubMed] [Google Scholar]
- 88.Torgerson DG, Ampleford EJ, Chiu GY, Gauderman WJ, Gignoux CR, Graves PE, Himes BE, Levin AM, Mathias RA, Hancock DB et al. (2011) Meta-analysis of genome-wide association studies of asthma in ethnically diverse North American populations. Nat Genet 43:887–892 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Moffatt MF, Gut IG, Demenais F, Strachan DP, Bouzigon E, Heath S, von Mutius E, Farrall M, Lathrop M, Cookson W et al. (2010) A large-scale, consortium-based genomewide association study of asthma. N Engl J Med 363:1211–1221 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Asai Y, Eslami A, van Ginkel CD, Akhabir L, Wan M, Yin D, Ellis G, Ben-Shoshan M, Marenholz I, Martino D et al. (2018) A Canadian genome-wide association study and meta-analysis confirm HLA as a risk factor for peanut allergy independent of asthma. J Allergy Clin Immunol 141:1513–1516 [DOI] [PubMed] [Google Scholar]
- 91.Marenholz I, Grosche S, Kalb B, Ruschendorf F, Blumchen K, Schlags R, Harandi N, Price M, Hansen G, Seidenberg J et al. (2017) Genome-wide association study identifies the SERPINB gene cluster as a susceptibility locus for food allergy. Nat Commun 8:1056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Liu X, Hong X, Tsai HJ, Mestan KK, Shi M, Kefi A, Hao K, Chen Q, Wang G, Caruso D et al. (2018) Genome-wide association study of maternal genetic effects and parent-of-origin effects on food allergy. Medicine (Baltimore) 97:e0043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Asai Y, Eslami A, van Ginkel CD, Akhabir L, Wan M, Ellis G, Ben-Shoshan M, Martino D, Ferreira MA, Allen K et al. (2018) Genome-wide association study and meta-analysis in multiple populations identifies new loci for peanut allergy and establishes C11orf30/EMSYas a genetic risk factor for food allergy. J Allergy Clin Immunol 141:991–1001 [DOI] [PubMed] [Google Scholar]
- 94.Brown SJ (2017) Molecular mechanisms in atopic eczema: insights gained from genetic studies. J Pathol 241:140–145 [DOI] [PubMed] [Google Scholar]
- 95.Osawa R, Akiyama M, Shimizu H (2011) Filaggrin gene defects and the risk of developing allergic disorders. Allergol Int 60:1–9 [DOI] [PubMed] [Google Scholar]
- 96.Brown SJ, Sandilands A, Zhao Y, Liao H, Relton CL, Meggitt SJ, Trembath RC, Barker JN, Reynolds NJ, Cordell HJ et al. (2008) Prevalent and low-frequency null mutations in the filaggrin gene are associated with early-onset and persistent atopic eczema. J Invest Dermatol 128:1591–1594 [DOI] [PubMed] [Google Scholar]
- 97.Henderson J, Northstone K, Lee SP, Liao H, Zhao Y, Pembrey M, Mukhopadhyay S, Smith GD, Palmer CN, McLean WH et al. (2008) The burden of disease associated with filaggrin mutations: a population-based, longitudinal birth cohort study. J Allergy Clin Immunol 121(872–877):e879. [DOI] [PubMed] [Google Scholar]
- 98.Kelly RS, Dahlin A, McGeachie MJ, Qiu W, Sordillo J, Wan ES, Wu AC, Lasky-Su J (2017) Asthma metabolomics and the potential for integrative omics in research and the clinic. Chest 151: 262–277 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Villani AC, Satija R, Reynolds G, Sarkizova S, Shekhar K, Fletcher J, Griesbeck M, Butler A4, Zheng S, Lazo S, Jardine L, Dixon D, Stephenson E, Nilsson E, Grundberg I, McDonald D, Filby A, Li W, De Jager PL, Rozenblatt-Rosen O, Lane AA, Haniffa M, Regev A, Hacohen N (2017) Single-cell RNA-seq reveals new types of human blood dendritic cells, monocytes, and progenitors. Science 356(6335). 10.1126/science.aah4573 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Kimball AK, Oko LM, Bullock BL, Nemenoff RA, van Dyk LF, Clambey ET (2018) A beginner’s guide to analyzing and visualizing mass cytometry data. J Immunol 200:3–22 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Nassar AF, Ogura H, Wisnewski AV (2015) Impact of recent innovations in the use of mass cytometry in support of drug development. Drug Discov Today 20:1169–1175 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Bhattacharya S, Dunn P, Thomas CG, Smith B, Schaefer H, Chen J, Hu Z, Zalocusky KA, Shankar RD, Shen-Orr SS et al. (2018) ImmPort, toward repurposing of open access immunological assay data for translational and clinical research. Sci Data 5:180015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Alizadeh AA, Eisen MB, Davis RE, Ma C, Lossos IS, Rosenwald A, Boldrick JC, Sabet H, Tran T, Yu X et al. (2000) Distinct types of diffuse large B-cell lymphoma identified by gene expression profiling. Nature 403:503–511 [DOI] [PubMed] [Google Scholar]
- 104.Bertucci F, Finetti P, Rougemont J, Charafe-Jauffret E, Cervera N, Tarpin C, Nguyen C, Xerri L, Houlgatte R, Jacquemier J et al. (2005) Gene expression profiling identifies molecular subtypes of inflammatory breast cancer. Cancer Res 65:2170–2178 [DOI] [PubMed] [Google Scholar]
- 105.Spira A, Beane JE, Shah V, Steiling K, Liu G, Schembri F, Gilman S, Dumas YM, Calner P, Sebastiani P et al. (2007) Airway epithelial gene expression in the diagnostic evaluation of smokers with suspect lung cancer. Nat Med 13:361–366 [DOI] [PubMed] [Google Scholar]
- 106.Rama swamy S, Ross KN, Lander ES, Golub TR (2003) A molecular signature of metastasis in primary solid tumors. Nat Genet 33: 49–54 [DOI] [PubMed] [Google Scholar]
- 107.Shivanna B, Maity S, Zhang S, Patel A, Jiang W, Wang L, Welty SE, Belmont J, Coarfa C, Moorthy B (2016) Gene expression profiling identifies cell proliferation and inflammation as the predominant pathways regulated by aryl hydrocarbon receptor in primary human fetal lung cells exposed to hyperoxia. Toxicol Sci 152:155–168 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Lau C, Olstad OK, Holden M, Nygard S, Fure H, Lappegard KT, Brekke OL, Espevik T, Hovig E, Mollnes TE (2015) Gene expression profiling of gram-negative bacteria-induced inflammation in human whole blood: the role of complement and CD14-mediated innate immune response. Genomics Data 5:176–183 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Hwang Y, Kim J, Shin JY, Kim JI, Seo JS, Webster MJ, Lee D, Kim S (2013) Gene expression profiling by mRNA sequencing reveals increased expression of immune/inflammation-related genes in the hippocampus of individuals with schizophrenia. Transl Psychiatry 3:e321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Trivedi NR, Gilliland KL, Zhao W, Liu W, Thiboutot DM (2006) Gene array expression profiling in acne lesions reveals marked upregulation of genes involved in inflammation and matrix remodeling. J Invest Dermatol 126:1071–1079 [DOI] [PubMed] [Google Scholar]
- 111.Fannin RD, Auman JT, Bruno ME, Sieber SO, Ward SM, Tucker CJ, Merrick BA, Paules RS (2005) Differential gene expression profiling in whole blood during acute systemic inflammation in lipopolysaccharide-treated rats. Physiol Genomics 21:92–104 [DOI] [PubMed] [Google Scholar]
- 112.Adarichev VA, Vermes C, Hanyecz A, Mikecz K, Bremer EG, Glant TT (2005) Gene expression profiling in murine autoimmune arthritis during the initiation and progression of joint inflammation. Arthritis Res Ther 7:R196–R207 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Kan M, Shumyatcher M, Himes BE (2017) Using omics approaches to understand pulmonary diseases. Respir Res 18:149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Woodruff PG, Modrek B, Choy DF, Jia G, Abbas AR, Ellwanger A, Koth LL, Arron JR, Fahy JV (2009) T-helper type 2-driven inflammation defines major subphenotypes of asthma. Am J Respir Crit Care Med 180:388–395 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Svenningsen S, Nair P (2017) Asthma endotypes and an overview of targeted therapy for asthma. Front Med (Lausanne) 4:158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Panettieri RA Jr (2018) The role of neutrophils in asthma. Immunol Allergy Clin N Am 38:629–638 [DOI] [PubMed] [Google Scholar]
- 117.Inselman JW, Jeffery MM, Maddux JT, Shah ND, Rank MA (2019) Trends and disparities in asthma biologic use in the United States. J Allergy Clin Immunol Pract 8(2):549–554.e1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Institute for Clinical and Economic Review, 2018. Biologic therapies for treatment of asthma associated with type 2 inflammation: effectiveness, value, and value-based price benchmarks: final evidence report. Available from: http://resource.nlm.nih.gov/101745064. Accessed Feb 2020.
- 119.Croteau-Chonka DC, Qiu W, Martinez FD, Strunk RC, Lemanske RF Jr, Liu AH, Gilliland FD, Millstein J, Gauderman WJ, Ober C (2017) Gene expression profiling in blood provides reproducible molecular insights into asthma control. Am J Respir Crit Care Med 195:179–188 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Bell E (2020) New biologic drugs for asthma: what PCPs need to know: https://www.healio.com/pediatrics/news/print/infectiousdiseases-in-children. Accessed Feb 2020
- 121.Pepper AN, Renz H, Casale TB, Garn H (2017) Biologic therapy and novel molecular targets of severe asthma. J Allergy Clin Immunol Pract 5:909–916 [DOI] [PubMed] [Google Scholar]
- 122.Campo P, Rodriguez F, Sanchez-Garcia S, Barranco P, Quirce S, Perez-Frances C, Gomez-Torrijos E, Cardenas R, Olaguibel JM, Delgado J et al. (2013) Phenotypes and endotypes of uncontrolled severe asthma: new treatments. J Investig Allergol Clin Immunol 23:76–88 quiz 71 p follow 88 [PubMed] [Google Scholar]
- 123.Berry M, Morgan A, Shaw DE, Parker D, Green R, Brightling C, Bradding P, Wardlaw AJ, Pavord ID (2007) Pathological features and inhaled corticosteroid response of eosinophilic and noneosinophilic asthma. Thorax 62:1043–1049 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Ghosh D, Ding L, Sivaprasad U, Geh E, Biagini Myers J, Bernstein JA, Khurana Hershey GK, Mersha TB (2015) Multiple transcriptome data analysis reveals biologically relevant atopic dermatitis signature genes and pathways. PLoS One 10: e0144316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Wen T, Stucke EM, Grotjan TM, Kemme KA, Abonia JP, Putnam PE, Franciosi JP, Garza JM, Kaul A, King EC et al. (2013) Molecular diagnosis of eosinophilic esophagitis by gene expression profiling. Gastroenterology 145:1289–1299 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Abonia JP, Wen T, Stucke EM, Grotjan T, Griffith MS, Kemme KA, Collins MH, Putnam PE, Franciosi JP, von Tiehl KF et al. (2013) High prevalence of eosinophilic esophagitis in patients with inherited connective tissue disorders. J Allergy Clin Immunol 132:378–386 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Molina-Infante J, Bredenoord AJ, Cheng E, Dellon ES, Furuta GT, Gupta SK, Hirano I, Katzka DA, Moawad FJ, Rothenberg ME et al. (2016) Proton pump inhibitor-responsive oesophageal eosinophilia: an entity challenging current diagnostic criteria for eosinophilic oesophagitis. Gut 65:524–531 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Shoda T, Wen T, Aceves SS, Abonia JP, Atkins D, Bonis PA, Caldwell JM, Capocelli KE, Carpenter CL, Collins MH et al. (2018) Eosinophilic oesophagitis endotype classification by molecular, clinical, and histopathological analyses: a cross-sectional study. Lancet Gastroenterol Hepatol 3:477–488 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Yang IV, Pedersen BS, Liu A, O’Connor GT, Teach SJ, Kattan M, Misiak RT, Gruchalla R, Steinbach SF, Szefler SJ et al. (2015) DNA methylation and childhood asthma in the inner city. J Allergy Clin Immunol 136:69–80 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Chen W,Wang T, Pino-Yanes M, Forno E, Liang L, Yan Q, Hu D, Weeks DE, Baccarelli A, Acosta-Perez E, Eng C, Han YY, Boutaoui N, Laprise C, Davies GA, Hopkin JM, Moffatt MF, Cookson WOCM, Canino G, Burchard EG, Celedón JC (2017) An epigenome-wide association study of total serum IgE in Hispanic children. J Allergy Clin Immunol 140(2):571–577. 10.1016/j.jaci.2016.11.030 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Liang L, Willis-Owen SA, Laprise C, Wong KC, Davies GA, Hudson TJ, Binia A, Hopkin JM, Yang IV, Grundberg E (2015) An epigenome-wide association study of total serum immunoglobulin E concentration. Nature 520:670–674 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.DeVries A, Wlasiuk G, Miller SJ, Bosco A, Stern DA, Lohman IC, Rothers J, Jones AC, Nicodemus-Johnson J, Vasquez MM et al. (2017) Epigenome-wide analysis links SMAD3 methylation at birth to asthma in children of asthmatic mothers. J Allergy Clin Immunol 140:534–542 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Schofield JPR, Burg D, Nicholas B, Strazzeri F, Brandsma J, Staykova D, Folisi C, Bansal AT, Xian Y, Guo Y et al. (2019) Stratification of asthma phenotypes by airway proteomic signatures. J Allergy Clin Immunol 144:70–82 [DOI] [PubMed] [Google Scholar]
- 134.Weitoft M, Muller C, Ahrman E, Bjermer L, Hoffmann HJ, Erjefalt J, Westergren-Thorsson G (2019) Comparison of normal and metaplastic epithelium in patients with stable versus persistently symptomatic severe asthma using laser-capture microdissection and data-independent acquisition-mass spectrometry. Am J Pathol 189:2358–2365 [DOI] [PubMed] [Google Scholar]
- 135.Pavel AB, Zhou L, Diaz A, Ungar B, Dan J, He H, Estrada YD, Xu H, Fernandes M, Renert-Yuval Y, Krueger JG, Guttman-Yassky E (2020) The proteomic skin profile ofmoderate-to-severe atopic dermatitis patients shows an inflammatory signature. J Am Acad Dermatol 82(3):690–699. 10.1016/j.jaad.2019.10.039 [DOI] [PubMed] [Google Scholar]
- 136.Leonard A, Wang J, Yu L, Liu H, Estrada Y, Greenlees L, McPhee R, Ruzin A, Guttman-Yassky E, Howell MD (2019) Atopic dermatitis endotypes based on allergen sensitization, reactivity to Staphylococcus aureus antigens, and underlying systemic inflammation. J Allergy Clin Immunol Pract 8(1):236–247.e3 [DOI] [PubMed] [Google Scholar]
- 137.Honda K, Littman DR (2016) The microbiota in adaptive immune homeostasis and disease. Nature 535:75–84 [DOI] [PubMed] [Google Scholar]
- 138.Borbet TC, Zhang X, Muller A, Blaser MJ (2019) The role of the changing human microbiome in the asthma pandemic. J Allergy Clin Immunol 144:1457–1466 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Gschwendtner S, Kang H, Thiering E, Kublik S, Fosel B, Schulz H, Krauss-Etschmann S, Heinrich J, Scholer A, Schloter M et al. (2019) Early life determinants induce sustainable changes in the gut microbiome of six-year-old children. Sci Rep 9:12675. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Rachid R, Chatila TA (2016) The role of the gut microbiota in food allergy. Curr Opin Pediatr 28:748–753 [DOI] [PubMed] [Google Scholar]
- 141.Kourosh A, Luna RA, Balderas M, Nance C, Anagnostou A, Devaraj S, Davis CM (2018) Fecal microbiome signatures are different in food-allergic children compared to siblings and healthy children. Pediatr Allergy Immunol 29:545–554 [DOI] [PubMed] [Google Scholar]
- 142.Powers CE, McShane DB, Gilligan PH, Burkhart CN, Morrell DS (2015) Microbiome and pediatric atopic dermatitis. J Dermatol 42: 1137–1142 [DOI] [PubMed] [Google Scholar]
- 143.Abrahamsson TR, Jakobsson HE, Andersson AF, Bjorksten B, Engstrand L, Jenmalm MC (2012) Low diversity of the gut microbiota in infants with atopic eczema. J Allergy Clin Immunol 129:434–440 440 e431–432 [DOI] [PubMed] [Google Scholar]
- 144.Abrahamsson TR, Jakobsson HE, Andersson AF, Bjorksten B, Engstrand L, Jenmalm MC (2014) Low gut microbiota diversity in early infancy precedes asthma at school age. Clin Exp Allergy 44:842–850 [DOI] [PubMed] [Google Scholar]
- 145.Kennedy EA, Connolly J, Hourihane JO, Fallon PG, McLean WHI, Murray D, Jo JH, Segre JA, Kong HH, Irvine AD (2017) Skin microbiome before development of atopic dermatitis: early colonization with commensal staphylococci at 2 months is associated with a lower risk of atopic dermatitis at 1 year. J Allergy Clin Immunol 139:166–172 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Chng KR, Tay AS, Li C, Ng AH, Wang J, Suri BK, Matta SA, McGovern N, Janela B, Wong XF et al. (2016) Whole metagenome profiling reveals skin microbiome-dependent susceptibility to atopic dermatitis flare. Nat Microbiol 1:16106. [DOI] [PubMed] [Google Scholar]
- 147.Cecchi L, D’Amato G, Annesi-Maesano I (2018) External exposome and allergic respiratory and skin diseases. J Allergy Clin Immunol 141:846–857 [DOI] [PubMed] [Google Scholar]
- 148.Subramanian A, Khatri SB (2019) The exposome and asthma. Clin Chest Med 40:107–123 [DOI] [PubMed] [Google Scholar]
- 149.Siroux V, Agier L, Slama R (2016) The exposome concept: a challenge and a potential driver for environmental health research. Eur Respir Rev 25:124–129 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Vrijheid M (2014) The exposome: a new paradigm to study the impact of environment on health. Thorax 69:876–878 [DOI] [PubMed] [Google Scholar]
- 151.Vrijheid M, Slama R, Robinson O, Chatzi L, Coen M, van den Hazel P, Thomsen C, Wright J, Athersuch TJ, Avellana N et al. (2014) The human early-life exposome (HELIX): project rationale and design. Environ Health Perspect 122:535–544 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Vineis P, Chadeau-Hyam M, Gmuender H, Gulliver J, Herceg Z, Kleinjans J, Kogevinas M, Kyrtopoulos S, Nieuwenhuijsen M, Phillips DH et al. (2017) The exposome in practice: design of the EXPOsOMICS project. Int J Hyg Environ Health 220:142–151 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Anto JM, Bousquet J, Akdis M, Auffray C, Keil T, Momas I, Postma DS, Valenta R, Wickman M, Cambon-Thomsen A (2017) Mechanisms of the development of allergy (MeDALL): introducing novel concepts in allergy phenotypes. J Allergy Clin Immunol 139:388–399 [DOI] [PubMed] [Google Scholar]
- 154.Karjalainen T (2014) Bridging the science-policy gap: EU-funded research for better environmental health. Available from: https://op.europa.eu/en/publication-detail/-/publication/593b6ff1-882c4adf-899f-43a56b80a221. Accessed Mar 2020
- 155.Cui Y, Balshaw DM, Kwok RK, Thompson CL, Collman GW, Birnbaum LS (2016) The exposome: embracing the complexity for discovery in environmental health. Environ Health Perspect 124:A137–A140 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Agache I, Miller R, Gern JE, Hellings PW, Jutel M, Muraro A, Phipatanakul W, Quirce S, Peden D (2019) Emerging concepts and challenges in implementing the exposome paradigm in allergic diseases and asthma: a PRACTALL document. Allergy 74: 449–463 [DOI] [PubMed] [Google Scholar]
- 157.Ardern-Jones MR. Characterisation of atopic dermatitis (AD) endotypes and novel treatment targets: towards a molecular classification. Exp Dermatol. 2018. April;27(4):433–434. 10.1111/exd.13152. Epub 2016 Dec 27. [DOI] [PubMed] [Google Scholar]
- 158.Thijs JL, Strickland I, Bruijnzeel-Koomen CAFM, Nierkens S, Giovannone B, Csomor E, Sellman BR, Mustelin T, Sleeman MA, de Bruin-Weller MS, Herath A, Drylewicz J, May RD, Hijnen D (2017) Moving toward endotypes in atopic dermatitis: Identification of patient clusters based on serum biomarker analysis. J Allergy Clin Immunol 140(3):730–737. 10.1016/j.jaci.2017.03.023 [DOI] [PubMed] [Google Scholar]
- 159.Higdon R, Earl RK, Stanberry L, Hudac CM, Montague E, Stewart E, Janko I, Choiniere J, Broomall W, Kolker N et al. (2015) The promise of multi-omics and clinical data integration to identify and target personalized healthcare approaches in autism spectrum disorders. Omics 19:197–208 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Mirza B, Wang W, Wang J, Choi H, Chung NC, Ping P (2019) Machine learning and integrative analysis of biomedical big data. Genes (Basel) 10(2). 10.3390/genes10020087 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Huang SM, Abernethy DR, Wang Y, Zhao P, Zineh I (2013) The utility of modeling and simulation in drug development and regulatory review. J Pharm Sci 102:2912–2923 [DOI] [PubMed] [Google Scholar]
- 162.Shaw DE, Sousa AR, Fowler SJ, Fleming LJ, Roberts G, Corfield J, Pandis I, Bansal AT, Bel EH, Auffray C, Compton CH, Bisgaard H, Bucchioni E, Caruso M, Chanez P, Dahlen B, Dahlen S-E, Dyson K, Frey U, Geiser T, de Verdier MG, Gibeon D, Guo Yk, Hashimoto S, Hedlin G, Jeyasingham E, Hekking P-PW, Higenbottam T, Horvath I, Knox AJ, Krug N, Erpenbeck VJ, Larsson LX, Lazarinis N, Matthews JG, Middelveld R, Montuschi P, Musial J, Myles D, Pahus L, Sandstrom T, Seibold W, Singer F, Strandberg K, Vestbo J, Vissing N, von Garnier C, Adcock IM, Wagers S, Rowe A, Howarth P, Wagener AH, Djukanovic R, Sterk PJ, Kian Fan Chung on behalf of the U-BIOPRED Study Group (2015) Clinical and inflammatory characteristics of the European U-BIOPRED adult severe asthma cohort. Eur Respir J 46:1308–1321 [DOI] [PubMed] [Google Scholar]
- 163.Shaw DE, Sousa AR, Fowler SJ, Fleming LJ, Roberts G, Corfield J, Pandis I, Bansal AT, Bel EH, Auffray C et al. (2015) Clinical and inflammatory characteristics of the European U-BIOPRED adult severe asthma cohort. Eur Respir J 46:1308–1321 [DOI] [PubMed] [Google Scholar]
- 164.Altman MC, Busse WW (2017) A deep dive into asthma transcriptomics. Lessons from U-BIOPRED. Am J Respir Crit Care Med 195:1279–1280 [DOI] [PubMed] [Google Scholar]
- 165.Kuo CS, Pavlidis S, Loza M, Baribaud F, Rowe A, Pandis I, Hoda U, Rossios C, Sousa A, Wilson SJ et al. (2017) A transcriptomedriven analysis of epithelial brushings and bronchial biopsies to define asthma phenotypes in U-BIOPRED. Am J Respir Crit Care Med 195:443–455 [DOI] [PubMed] [Google Scholar]
- 166.Simpson AJ, Fowler SJ, Group, U.B.S (2018) Reclassification of bronchodilator reversibility in the U-BIOPRED adult asthma cohort using z scores. Chest 153:1070–1072 [DOI] [PubMed] [Google Scholar]
- 167.Takahashi K, Pavlidis S, Ng Kee Kwong F, Hoda U, Rossios C, Sun K, Loza M, Baribaud F, Chanez P, Fowler SJ, Horvath I, Montuschi P, Singer F, Musial J, Dahlen B, Dahlen SE, Krug N, Sandstrom T, Shaw DE, Lutter R, Bakke P, Fleming LJ, Howarth PH, Caruso M, Sousa AR, Corfield J, Auffray C, De Meulder B, Lefaudeux D, Djukanovic R, Sterk PJ, Guo Y, Adcock IM, Chung KF, on behalf of the U-BIOPRED study group (2018) Sputum proteomics and airway cell transcripts of current and ex-smokers with severe asthma in U-BIOPRED: an exploratory analysis. Eur Respir J 51(5). 10.1183/13993003.02173-2017 [DOI] [PubMed] [Google Scholar]
- 168.Kelly RS, Chawes BL, Blighe K, Virkud YV, Croteau-Chonka DC, McGeachie MJ, Clish CB, Bullock K, Celedón JC, Weiss ST (2018) An integrative transcriptomic and metabolomic study of lung function in children with asthma. Chest 154:335–348 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Lee-Sarwar K, Kelly RS, Lasky-Su J, Moody DB, Mola AR, Cheng TY, Comstock LE, Zeiger RS, O’Connor GT, Sandel MT et al. (2018) Intestinal microbial-derived sphingolipids are inversely associated with childhood food allergy. J Allergy Clin Immunol 142(335–338):e339. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Zhang Y, Willis-Owen SA, Spiegel S, Lloyd CM, Moffatt MF, Cookson WO (2019) The ORMDL3 asthma gene regulates ICAM1 and has multiple effects on cellular inflammation. Am J Respir Crit Care Med 199:478–488 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Obeso D, Mera-Berriatua L, Rodriguez-Coira J, Rosace D, Fernandez P, Martin-Antoniano IA, Santaolalla M, Marco Martin G, Chivato T, Fernandez-Rivas M et al. (2018) Multi-omics analysis points to altered platelet functions in severe food associated respiratory allergy. Allergy 73:2137–2149 [DOI] [PubMed] [Google Scholar]
- 172.Son MJ, Yang GJ, Jo EH, Shim YH, Kang SJ, Hong JE, Kim YE, Lee JE, Chun J, Park S et al. (2019) Association of atopic dermatitis with obesity via a multi-omics approach: a protocol for a casecontrol study. Medicine (Baltimore) 98:e16527. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Ross EG, Shah NH, Dalman RL, Nead KT, Cooke JP, Leeper NJ (2016) The use of machine learning for the identification of peripheral artery disease and future mortality risk. J Vasc Surg 64: 1515–1522.e1513 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Lanza ST, Rhoades BL (2013) Latent class analysis: an alternative perspective on subgroup analysis in prevention and treatment. Prev Sci 14:157–168 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Havstad S, Johnson CC, Kim H, Levin AM, Zoratti EM, Joseph CL, Ownby DR, Wegienka G (2014) Atopic phenotypes identified with latent class analyses at age 2 years. J Allergy Clin Immunol 134:722–727. e722 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Roduit C, Frei R, Depner M, Karvonen AM, Renz H, Braun-Fahrländer C, Schmausser-Hechfellner E, Pekkanen J, Riedler J, Dalphin J-C (2017) Phenotypes of atopic dermatitis depending on the timing of onset and progression in childhood. JAMA Pediatr 171:655–662 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Dharma C, Lefebvre D, Tran M, Lou W, Subbarao P, Becker A, Mandhane P, Turvey S, Sears M, investigators, C.S (2018) Patterns of allergic sensitization and atopic dermatitis from 1 to 3 years: effects on allergic diseases. Clin Exp Allergy 48:48–59 [DOI] [PubMed] [Google Scholar]
- 178.Ross MK, Yoon J, van der Schaar A, van der Schaar M (2018) Discovering pediatric asthma phenotypes on the basis of response to controller medication using machine learning. Ann Am Thoracs Soc 15:49–58 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Togias A (2003) Rhinitis and asthma: evidence for respiratory system integration. J Allergy Clin Immunol 111:1171–1183 [DOI] [PubMed] [Google Scholar]
- 180.Moore WC, Meyers DA, Wenzel SE, Teague WG, Li H, Li X, D’Agostino R Jr, Castro M, Curran-Everett D, Fitzpatrick AM et al. (2010) Identification of asthma phenotypes using cluster analysis in the severe asthma research program. Am J Respir Crit Care Med 181:315–323 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Li X, Howard TD, Moore WC, Ampleford EJ, Li H, Busse WW, Calhoun WJ, Castro M, Chung KF, Erzurum SC et al. (2011) Importance of hedgehog interacting protein and other lung function genes in asthma. J Allergy Clin Immunol 127:1457–1465 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.National Asthma Education and Prevention Program. Expert Panel Report 3 (EPR-3): Guidelines for the Diagnosis and Management of Asthma-Summary Report 2007. J Allergy Clin Immunol. 2007. November;120(5 Suppl):S94–138 [DOI] [PubMed] [Google Scholar]
- 183.Tang HHF, Sly PD, Holt PG, Holt KE, Inouye M (2020) Systems biology and big data in asthma and allergy: recent discoveries and emerging challenges. Eur Respir J 55(1). 10.1183/13993003.00844-2019 [DOI] [PubMed] [Google Scholar]
- 184.Vercelli D, Martinez FD (2006) The Faustian bargain of genetic association studies: bigger might not be better, or at least it might not be good enough. J Allergy Clin Immunol 117:1303–1305 [DOI] [PubMed] [Google Scholar]
- 185.Huang QQ, Ritchie SC, Brozynska M, Inouye M (2018) Power, false discovery rate and winner’s curse in eQTL studies. Nucleic Acids Res 46:e133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.McGregor MC, Krings JG, Nair P, Castro M (2019) Role of biologics in asthma. Am J Respir Crit Care Med 199:433–445 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Wenzel S, Wilbraham D, Fuller R, Getz EB, Longphre M (2007) Effect of an interleukin-4 variant on late phase asthmatic response to allergen challenge in asthmatic patients: results of two phase 2a studies. Lancet 370:1422–1431 [DOI] [PubMed] [Google Scholar]
- 188.Luo J, Liu D, Liu CT (2016) The efficacy and safety of antiinterleukin 13, a monoclonal antibody, in adult patients with asthma: a systematic review and meta-analysis. Medicine (Baltimore) 95:e2556. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Busse WW, Israel E, Nelson HS, Baker JW, Charous BL, Young DY, Vexler V, Shames RS, Daclizumab Asthma Study, G (2008) Daclizumab improves asthma control in patients with moderate to severe persistent asthma: a randomized, controlled trial. Am J Respir Crit Care Med 178:1002–1008 [DOI] [PubMed] [Google Scholar]
- 190.Farzan N, Vijverberg SJ, Kabesch M, Sterk PJ, Maitland-van der Zee AH (2018) The use of pharmacogenomics, epigenomics, and transcriptomics to improve childhood asthma management: where do we stand? Pediatr Pulmonol 53:836–845 [DOI] [PubMed] [Google Scholar]
- 191.Dhondalay GK, Rael E, Acharya S, Zhang W, Sampath V, Galli SJ, Tibshirani R, Boyd SD, Maecker H, Nadeau KC et al. (2018) Food allergy and omics. J Allergy Clin Immunol 141:20–29 [DOI] [PubMed] [Google Scholar]
- 192.Barbieri R, Guryev V, Brandsma CA, Suits F, Bischoff R, Horvatovich P (2016) Proteogenomics: key driver for clinical discovery and personalized medicine. Adv Exp Med Biol 926:21–47 [DOI] [PubMed] [Google Scholar]
- 193.Jameson JL, Longo DL (2015) Precision medicine—personalized, problematic, and promising. N Engl J Med 372:2229–2234 [DOI] [PubMed] [Google Scholar]
- 194.Muraro A, Fokkens WJ, Pietikainen S, Borrelli D, Agache I, Bousquet J, Costigliola V, Joos G, Lund VJ, Poulsen LK et al. (2016) European symposium on precision medicine in allergy and airways diseases: report of the European Union parliament symposium (October 14, 2015). Allergy 71:583–587 [DOI] [PubMed] [Google Scholar]
- 195.Diamant Z, Vijverberg S, Alving K, Bakirtas A, Bjermer L, Custovic A, Dahlen SE, Gaga M, Gerth van Wijk R, Giacco SD, Hamelmann E, Heaney LG, Heffler E, Kalayci Ö, Kostikas K, Lutter R, Olin AC, Sergejeva S, Simpson A, Sterk P, Tufvesson E, Agache I, Seys SF (2019) Toward clinically applicable biomarkers for asthma: An EAACI position paper. Allergy 74(10): 1835–1851. 10.1111/all.13806 [DOI] [PubMed] [Google Scholar]
- 196.Kersten ET, Koppelman GH (2017) Pharmacogenetics of asthma: toward precision medicine. Curr Opin Pulm Med 23:12–20 [DOI] [PubMed] [Google Scholar]
- 197.Laprise C, Bouzigon E (2013) The genetics of asthma and allergic diseases: pieces of the puzzle are starting to come together. Curr Opin Allergy Clin Immunol 13:461–462 [DOI] [PubMed] [Google Scholar]
- 198.Bellanti JA, Settipane RA (2018) Allergic diseases: a collection of interactive immunologic disorders at the crossroads of genetics, environment, and immunity. Allergy Asthma Proc 39:83–85 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Redwood AJ, Pavlos RK, White KD, Phillips EJ (2018) HLAs: key regulators of T-cell-mediated drug hypersensitivity. HLA 91: 3–16 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Thijs JL, Van Der Geest BA, Van Der Schaft J, Van Den Broek MP, Van Seggelen WO, Bruijnzeel-Koomen C, Hijnen D-J, Van Schaik RH, De Bruin-Weller M (2017) Predicting therapy response to mycophenolic acid using UGT1A9 genotyping: towards personalized medicine in atopic dermatitis. J Dermatol Treat 28: 242–245 [DOI] [PubMed] [Google Scholar]
- 201.Ruzicka T, Hanifin JM, Furue M, Pulka G, Mlynarczyk I, Wollenberg A, Galus R, Etoh T, Mihara R, Yoshida H (2017) Anti–interleukin-31 receptor A antibody for atopic dermatitis. N Engl J Med 376:826–835 [DOI] [PubMed] [Google Scholar]
- 202.Rothenberg ME, Wen T, Greenberg A, Alpan O, Enav B, Hirano I, Nadeau K, Kaiser S, Peters T, Perez A et al. (2015) Intravenous anti-IL-13 mAb QAX576 for the treatment of eosinophilic esophagitis. J Allergy Clin Immunol 135:500–507 [DOI] [PubMed] [Google Scholar]
- 203.Wain LV, Shrine N, Artigas MS, Erzurumluoglu AM, Noyvert B, Bossini-Castillo L, Obeidat M, Henry AP, Portelli MA, Hall RJ et al. (2017) Genome-wide association analyses for lung function and chronic obstructive pulmonary disease identify new loci and potential druggable targets. Nat Genet 49:416–425 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Henry AP, Probert K, Stewart CE, Thakker D, Bhaker S, Azimi S, Hall IP, Sayers I (2019) Defining a role for lung function associated gene GSTCD in cell homeostasis. Respir Res 20:172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Rackemann FM (1947) A working classification of asthma. Am J Med 3:601–606 [DOI] [PubMed] [Google Scholar]
- 206.— (1987) Standards for the diagnosis and care of patients with chronic obstructive pulmonary disease (COPD) and asthma. This official statement of the American Thoracic Society was adopted by the ATS Board of Directors, November 1986. Am Rev Respir Dis 136(1):225–244 [DOI] [PubMed] [Google Scholar]
- 207.Brightling CE, Woltmann G, Wardlaw AJ, Pavord ID (1999) Development of irreversible airflow obstruction in a patient with eosinophilic bronchitis without asthma. Eur Respir J 14:1228–1230 [DOI] [PubMed] [Google Scholar]
- 208.Wenzel SE, Schwartz LB, Langmack EL, Halliday JL, Trudeau JB, Gibbs RL, Chu HW (1999) Evidence that severe asthma can be divided pathologically into two inflammatory subtypes with distinct physiologic and clinical characteristics. Am J Respir Crit Care Med 160:1001–1008 [DOI] [PubMed] [Google Scholar]
- 209.Wenzel SE (2012) Asthma phenotypes: the evolution from clinical to molecular approaches. Nat Med 18:716–725 [DOI] [PubMed] [Google Scholar]
- 210.North ML, Brook JR, Lee EY, Omana V, Daniel NM, Steacy LM, Evans GJ, Diamond ML, Ellis AK (2017) The Kingston Allergy Birth Cohort: exploring parentally reported respiratory outcomes through the lens of the exposome. Ann Allergy Asthma Immunol 118:465–473 [DOI] [PubMed] [Google Scholar]
- 211.Holzinger A, Dehmer M, Jurisica I (2014) Knowledge discovery and interactive data mining in bioinformatics—state-of-the-art, future challenges and research directions. BMC Bioinformatics 15(Suppl 6):I1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.Yu KH, Snyder M (2016) Omics profiling in precision oncology. Mol Cell Proteomics 15:2525–2536 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213.Reinke SN, Gallart-Ayala H, Gomez C, Checa A, Fauland A, Naz S, Kamleh MA, Djukanovic R, Hinks TS, Wheelock CE (2017) Metabolomics analysis identifies different metabotypes of asthma severity. Eur Respir J 49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214.Wills-Karp M, Ewart SL (2004) Time to draw breath: asthmasusceptibility genes are identified. Nat Rev Genet 5:376–387 [DOI] [PubMed] [Google Scholar]
- 215.Zosky GR, Sly PD (2007) Animal models of asthma. Clin Exp Allergy 37:973–988 [DOI] [PubMed] [Google Scholar]
