Next Article in Journal
Comparing the Impact of Different Antiarrhythmic Classes on Clinical Outcomes Following Atrial Fibrillation Catheter Ablation
Previous Article in Journal
Spray-Dried Polymeric Microspheres for Lipophilic Drugs: Formulation Design, Physicochemical Characterization, and In Vitro Release Evaluation
Previous Article in Special Issue
Therapeutic Potential of Hexahydrocurcumin in the Regeneration and Protection of the Retinal Pigment Epithelium
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Targeted Biologic Therapies in Severe Asthma: Mechanisms, Biomarkers, and Clinical Applications

by
Renata Maria Văruț
1,†,
Dop Dalia
2,†,
Kristina Radivojevic
1,
Diana Maria Trasca
3,*,
George-Alin Stoica
4,*,
Niculescu Stefan Adrian
5,
Niculescu Elena Carmen
2 and
Cristina Elena Singer
2
1
Research Methodology Department, Faculty of Pharmacy, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
2
Department of Mother and Baby, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
3
Department of Internal Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
4
Department of Pediatric Surgery, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
5
Department of Orthopedics, University of Medicine and Pharmacy Craiova, 200349 Craiova, Romania
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Pharmaceuticals 2025, 18(7), 1021; https://doi.org/10.3390/ph18071021
Submission received: 19 June 2025 / Revised: 3 July 2025 / Accepted: 4 July 2025 / Published: 10 July 2025

Abstract

Asthma represents a heterogeneous disorder characterized by a dynamic balance between pro-inflammatory and anti-inflammatory forces, with allergic sensitization contributing substantially to airway hyperresponsiveness and remodeling. Central to its pathogenesis are cytokines such as IL-4, IL-5, IL-13, IL-17, and IL-33, which drive recruitment of eosinophils, neutrophils, and other effector cells, thereby precipitating episodic exacerbations in response to viral and environmental triggers. Conventional biomarkers, including blood and sputum eosinophil counts, IgE levels, and fractional exhaled nitric oxide, facilitate phenotypic classification and guide the emerging biologic era. Monoclonal antibodies targeting IgE (omalizumab) and IL-5 (mepolizumab, benralizumab, reslizumab, depemokimab) have demonstrated the ability to reduce exacerbation frequency and improve lung function, with newer agents such as depemokimab offering extended dosing intervals. Itepekimab, an anti-IL-33 antibody, effectively engages its target and mitigates tissue eosinophilia, while CM310-stapokibart, tralokinumab, and lebrikizumab inhibit IL-4/IL-13 signaling with variable efficacy depending on patient biomarkers. Comparative analyses of these biologics, encompassing affinity, dosing regimens, and trial outcomes, underscore the imperative of personalized therapy to optimize disease control in severe asthma.

Graphical Abstract

1. Introduction

Asthma affects roughly 358 million individuals worldwide, and its occurrence varies widely by region [1]. Rates tend to be higher in cities and among people exposed to known risk factors, such as allergens, tobacco smoke, and air pollution. In many high-income countries, new cases have leveled off after decades of rise; by contrast, low- and middle-income nations are seeing rapid increases in asthma prevalence, likely fueled by worsening fossil fuel emissions and shifts toward Western lifestyles. Limited access to precise diagnostic tools and uniform treatment protocols in these regions further amplifies the burden on patients, families, and healthcare systems [2].
The hallmark of asthma is expiratory airflow obstruction, which is generally reversible yet stems from narrowing of the airway lumen. Chronic inflammation of the airway wall, driven by infiltrating and activated immune cells (eosinophils, neutrophils, lymphocytes, dendritic cells, innate lymphoid cells, and mast cells), triggers bronchial hyperresponsiveness, mucus overproduction, and structural remodeling of the airways.
Clinically, asthma presents with symptoms such as shortness of breath, coughing, wheezing, chest tightness, progressive loss of lung function, frequent exacerbations, and variable disease severity. These diverse features reflect multiple underlying mechanisms in which interactions between structural cells and immune elements give rise to the pathological traits of asthma. Genetic differences and environmental exposures further shape each patient’s profile, leading to considerable heterogeneity in symptoms and biomarker patterns.
The 2023–2024 GINA updates classify several common asthma phenotypes: allergic asthma, nonallergic asthma, adult-onset (late-onset) asthma, asthma with persistent airflow limitation, and obesity-associated asthma [3]. In parallel, animal studies—particularly murine models of allergic airway inflammation—have confirmed a central role for T helper 2 (TH2) cells in driving airway inflammation and hyperreactivity [4,5]. TH2 cytokines mediate key asthma characteristics: IL-5 for tissue eosinophilia, IL-13 for bronchial hyperresponsiveness, and IL-4 plus IL-13 for goblet cell metaplasia [6].
More recently, other innate immune populations—mast cells, basophils, group 2 ILCs, “M2” macrophages activated by IL-4/IL-13, and a subset of IL-4-secreting NK/NKT cells—have been shown to contribute to type 2 cytokine production. This has prompted a shift in terminology from “TH2-high” to “type 2-high” asthma [7]. Roughly half of asthma patients exhibit this eosinophil-rich, type 2-high profile [8]. The remainder—classified as “type 2-low” asthma—lack eosinophilia and often display airway neutrophilia, obesity-related systemic inflammation, or minimal immune activation [9].
Asthma management underwent a paradigm shift in the early 2000s with the advent of the first biologic therapies (omalizumab). Before this, oral corticosteroids represented the ultimate treatment option for severe cases. Chronic use of systemic steroids, however, carries a substantial risk of serious side effects, ranging from adrenal suppression and Cushingoid features to glaucoma, cataract formation, steroid-induced diabetes, and bone demineralization [10]. Moreover, oral steroids fail to halt the structural changes in the airway driven by IL-33, a key cytokine in airway remodeling [11].
Even with strict adherence to conventional treatments, about 15 percent of asthma sufferers continue to experience poor disease control. For these individuals, both GINA and the National Heart, Lung, and Blood Institute recommend escalating to biologic agents [11]. Such therapies are indicated when patients’ symptoms remain uncontrolled or deteriorate despite optimized use of inhaled β2-agonists, anticholinergics, and corticosteroids—whether administered singly or in combination [12]. Clinical trials and real-world studies suggest that initiating a biologic is particularly beneficial in patients requiring two or more courses of oral steroids annually to manage exacerbations [13].
Prior to starting a biologic, confirming the asthma diagnosis and assessing biomarkers, such as peripheral eosinophil counts, total IgE, and fractional exhaled nitric oxide (FeNO), is essential. These measures help predict which patients are most likely to derive benefit from targeted biologic interventions [14]. Data from the International Severe Asthma Registry indicate that over 60 percent of enrolled patients meet eligibility criteria for biologics and experience high exacerbation rates; upon starting biologic therapy, their exacerbation frequency drops markedly [15]. Conversely, discontinuation of biologic treatment is associated with worsened outcomes and increased emergency healthcare utilization compared with those who maintain therapy [16]. As the role of biologics in severe asthma continues to broaden, further studies are required to define optimal treatment algorithms and long-term management strategies [17].

2. Asthma

2.1. Pro-Inflammatory and Anti-Inflammatory Aspects of Asthma

Under the influence of chemotactic signals released in the lungs, a variety of inflammatory cells migrate from the bloodstream into the airway tissue, where they contribute to asthma pathogenesis. In addition to these recruited leukocytes, resident structural cells, such as the airway epithelium, fibroblasts, and smooth muscle cells, serve as important sources of inflammatory mediators, actively sustaining the inflammatory milieu. In asthmatic patients, both arms of the immune system are engaged: innate effectors (mast cells, dendritic cells, eosinophils, neutrophils, basophils, innate lymphoid cells, monocytes, and macrophages) and adaptive lymphocytes (T and B cells) form the cellular landscape of airway inflammation [18].
Central to the eosinophilic inflammation seen in many asthma phenotypes are TH2 lymphocytes, which secrete IL-4, IL-5, and IL-13 in large amounts [19]. IL-4 drives allergic sensitization and class switching to IgE, IL-5 sustains eosinophil survival, and IL-13 orchestrates multiple pathological changes, mucus hypersecretion, goblet cell transformation, bronchial hyperresponsiveness, and airway remodeling [20]. By contrast, TH1 cells produce IL-2, IFN-γ, and TNF-α, which can counterbalance TH2-driven processes and thus may exert protective effects against eosinophil-dominant inflammation [21]. Supporting this, administration of IL-12 (a cytokine that promotes TH1 differentiation) in murine models reduces antigen-induced airway hyperresponsiveness and inflammation via IFN-γ-mediated pathways [22,23]. Yet, in some severe, corticosteroid-resistant asthma cases, an abundance of IFN-γ-producing TH1 cells has been linked to more intractable disease [24,25].
Beyond TH2 and TH1 subsets, TH17 cells and their hallmark cytokine IL-17A have emerged as key players in neutrophil-predominant severe asthma phenotypes [26]. IL-17 and IL-22 enhance neutrophil recruitment to the airways [27], while neutrophil extracellular traps and cytoplasts further skew the immune response toward a TH17-driven, neutrophilic pattern [28]. However, IL-17’s role is dualistic: in some stages it confers protection, whereas in others it exacerbates airway inflammation [29,30]. In chronic models of asthma, IL-17A promotes fibroblast proliferation [31], undermines regulatory T cell-mediated suppression [32], and directly induces contraction of bronchial smooth muscle cells [33] (Figure 1).
The identification of TH9 cells, a CD4+ subset that secretes IL-9, has added another layer of complexity. Differentiation into TH9 is driven by IL-4 and TGF-β, which enable transcription factors PU.1 and IRF4 to activate the Il9 gene [34,35]. IL-25 (also known as IL-17E) further amplifies IL-9 production by TH9 cells [36]. Functionally, IL-9 promotes IgE synthesis and eosinophilic inflammation [37], and in experimental allergic models, mast cell accumulation and protease expression are IL-9 dependent [38,39]. Deletion of critical regulatory elements in the Il9 locus, which impairs TH9 maturation, markedly reduces allergic lung inflammation in mice [40,41]. Moreover, human trials of the anti-IL-9 monoclonal antibody MEDI-528 have shown promising efficacy in asthma patients [42].
Despite asthma’s well-recognized heterogeneity, a skewed immune balance underpins its development. It is the shifting interplay among T cell subsets and macrophage phenotypes, from disease initiation through progression, that shapes the overall inflammatory network. Rather than a simple overabundance of one cell type, it is the disruption of “pro-versus anti-inflammatory” regulatory circuits that drives pathology [43]. While the classic TH2-dominant paradigm remains fundamental, current evidence reveals a far more nuanced in vivo landscape, marked by uneven subpopulation dynamics and context-dependent cytokine expression.

2.2. Allergies and Asthma

The hallmark of allergy is an overzealous immune response to ordinarily harmless substances, pollen, house dust mites, pet dander or saliva, particular foods, and insect venoms among them. Upon exposure, sensitized individuals release mediators such as histamine, precipitating classic allergic manifestations: sneezing, itching, rhinorrhea or nasal congestion, tearing, and cutaneous eruptions. While most reactions are mild, severe anaphylaxis can occur [44].
Asthma and allergy frequently coexist, with allergens often acting as precipitating factors for bronchial hyperreactivity. In allergic asthma, inhaled triggers amplify airway inflammation, edema, and mucus secretion, further constricting the airways and provoking or intensifying wheezing and breathlessness.
Effective control of allergic asthma hinges on rigorous allergy management. It was demonstrated, as in many locales, that a high proportion of asthma sufferers exhibit atopic sensitization. Multiple epidemiological surveys have aimed to quantify asthma and its allergic subtype. For instance, Al Ghobain et al. (2018) used the ECRHS questionnaire in 2405 participants, finding an 18.2 percent rate of wheezing in the past year (absent upper respiratory infection), with no significant sex differences (p = 0.107) [45]. Physician-diagnosed asthma was reported by 11.3 percent (p = 0.239 between genders), and 10.6 percent were on asthma medications. Asthma prevalence did not vary significantly by residence (p = 0.07), education level (p = 0.11), or smoking status (p = 0.06), but showed a strong association with nasal allergy (p < 0.001). Long recognized as linked entities, asthma and allergic rhinitis share underlying immune mechanisms. Emerging evidence suggests that intense lower-airway inflammation can provoke or exacerbate upper-airway allergic responses, underscoring the continuum of airway disease in atopic individuals [46].

2.3. Cytokines in Asthma

T helper lymphocytes play a pivotal role in orchestrating immune reactions. In asthma, the interplay of cytokines, immune cell populations, and their signaling cascades forms a highly dynamic, intricate network. Cytokines, small protein mediators released by various immune cells, are critical for regulating inflammation, shaping immune responses, and facilitating cell-to-cell communication. Table 1 summarizes the key cytokines implicated in asthma and describes their respective contributions to the disease’s inflammatory and immunological processes.
In addition to classical Th2 cytokines such as IL-4, IL-5, and IL-13, the epithelial-derived alarmin thymic stromal lymphopoietin (TSLP) has emerged as a critical upstream mediator of airway inflammation in asthma, and is now recognized as a major therapeutic target, most notably for the monoclonal antibody tezepelumab.

2.4. Exacerbations in Asthma

Asthma flare-ups significantly contribute to patient morbidity, elevate healthcare expenditures, and, for some individuals, accelerate the decline in pulmonary function [60]. Although exacerbation rates can be lowered through optimized therapies, complete prevention is not always achievable [61]. Since acute attacks may occur despite adherence to standard regimens, early recognition of high-risk patients and implementation of personalized action plans are vital for improving overall control and quality of life.
Episodes leading to emergency department visits or hospital admissions are not only costly but also predict a higher likelihood of future exacerbations, regardless of baseline asthma severity, demographic factors, or previous control levels [62]. This underscores the ongoing necessity for more effective preventive and therapeutic strategies.
The most frequent precipitants of these attacks are viral respiratory pathogens, especially human rhinovirus subtypes A and C [63]. In school-aged children, and similarly in adults, hospital admissions for asthma spike in autumn (September–December) and again in spring, mirroring seasonal peaks in rhinovirus circulation [64,65].
Beyond rhinoviruses, other viruses play roles in acute worsening. During the 2009 H1N1 influenza A pandemic, asthma patients represented a disproportionate share of ICU admissions and fatalities [66,67]. Respiratory syncytial virus, well known for causing wheezing in infants, can also trigger exacerbations in older adults, particularly those over 65 [68]. Although coronaviruses, metapneumoviruses, parainfluenza viruses, adenoviruses, and bocaviruses are detected less commonly in asthma attacks, their presence has been confirmed [69].
Bacterial infections may impair mucociliary clearance and heighten mucus production, potentially sustaining chronic airway inflammation [70]. While direct links between bacteria and sudden asthma flares remain limited, viral infections can weaken macrophage-mediated antibacterial defenses and alter the airway microbiome, which could predispose to secondary bacterial involvement [71].
Atopic sensitization further compromises innate antiviral responses, rendering individuals more susceptible to virus-induced wheezing; allergen-driven inflammation also amplifies airway reactivity to rhinoviruses. In addition, sensitization to molds such as Alternaria alternata, which correlates with five-fold higher odds of asthma, increased bronchial hyperresponsiveness, wheezing, and inhaler use, aligns with seasonal exacerbation trends tied to airborne spore counts [72].
Environmental irritants likewise act as triggers. Tobacco smoke exposure is linked to persistent wheezing, more severe asthma, and a higher rate of ED visits and hospitalizations [73]. Pollutants, including particulate matter, ozone, nitrogen dioxide, sulfur dioxide, and diesel exhaust, intensify airway inflammation and hyperreactivity and can synergize with viral infections to provoke attacks [74].

3. Targeted Immune Modulation in Severe Asthma

3.1. Conventional Biomarkers in Clinical Practice

Asthma management increasingly relies on measurable indicators, such as eosinophil and neutrophil counts, total IgE, fractional exhaled nitric oxide (FeNO), leukotriene levels, and others, that bridge underlying biology with clinical presentation. Although no single marker is without limitations, these tests are invaluable for tailoring the use of targeted biologic treatments.

3.1.1. Eosinophils

Peripheral blood and sputum eosinophil counts correlate with disease activity in both atopic and non-atopic asthma. Elevated eosinophils signal greater severity, and serial blood measurements are routinely used to monitor the response to anti-IL-5 therapies [75]. To date, blood eosinophilia remains one of the most reliable biomarkers for classifying asthma subtypes and gauging treatment effectiveness.

3.1.2. Neutrophils

High airway neutrophil levels often characterize severe, non-type 2 asthma phenotypes driven by TH17 pathways [76]. While sputum analysis can reveal pronounced neutrophilia, the relationship between airway and blood neutrophil counts is inconsistent in some cohorts [77], underscoring the need for complementary assessments.

3.1.3. Total IgE

IgE production is promoted by TH2 lymphocyte–B cell interactions, leading to mast cell and basophil activation [78]. Although serum IgE does not reliably track disease severity or therapeutic response, anti-IgE monoclonal antibodies are approved for patients with persistent atopy, particularly children with allergic asthma and adults who also suffer from allergic rhinitis, chronic sinusitis, nasal polyposis, or chronic urticaria [79].

3.1.4. Fractional Exhaled Nitric Oxide (FeNO)

Nitric oxide synthesis in the airway epithelium is upregulated by IL-4 and IL-13 signaling via IL-4Rα [76]. FeNO measurement offers a noninvasive readout of eosinophilic inflammation and type 2-driven asthma; it can be performed at home to guide therapy adjustments [80]. Together, FeNO and blood eosinophil levels form the cornerstone of biomarker-guided management in severe asthma.

3.1.5. Leukotrienes

Leukotrienes are synthesized through the 5-lipoxygenase (5-LOX) enzymatic route. These cysteinyl leukotrienes contribute to bronchoconstriction and drive inflammatory processes. In asthmatic airways, they provoke smooth muscle tightening, increase vascular leak and mucus output, and recruit plus activate multiple inflammatory cell types [81].

3.2. Exacerbation with Biologics

Biologic agents have proven their worth in cutting the frequency of severe asthma exacerbations [82]. In the MEX trial, researchers characterized the nature of breakthrough exacerbations in patients with severe eosinophilic asthma receiving mepolizumab. Despite profound reductions in circulating eosinophils, about half of these residual attacks were still driven by eosinophilic inflammation, while the other half showed a non-eosinophilic profile [83]. Importantly, FeNO measurements taken during an exacerbation reliably distinguished high-eosinophil from low-eosinophil events. When FeNO was ≤20 ppb, the negative predictive value for an eosinophilic flare was 100 percent (95% CI 0.8–1.0), suggesting that systemic steroids might be avoidable in this setting. Conversely, FeNO ≥ 50 ppb corresponded to a positive predictive value of 76.9 percent (95% CI 0.6–0.9) for eosinophilic attacks, indicating that oral corticosteroids were likely warranted [84].
A separate prospective investigation by Poznanski et al. evaluated why some patients on benralizumab continued to experience exacerbations or failed to halve their prednisone dose, a definition of suboptimal response, over a median follow-up of 14 months [85]. Among 74 treated individuals (23 of whom underwent NK cell quantification), 27 percent met the suboptimal response criteria. These patients’ sputum was neutrophil-rich (mean 19.5 ± 37 × 106 cells/g vs. 4.5 ± 6 × 106 cells/g in responders; p = 0.01) and frequently linked to viral or bacterial infections (e.g., H. influenzae, M. catarrhalis, S. pneumoniae, S. aureus) [84]. They also had experienced more infections in the prior year (median 0.5 vs. 0) and exhibited lower circulating NK cell counts alongside elevated NKT cell numbers, cells known to drive neutrophil recruitment and activation.
Together, these findings underscore that even under targeted T2-inflammation blockade, persistent exacerbations remain heterogeneous. Profiling each flare, distinguishing eosinophilic from neutrophilic phenotypes, emerges as a critical step toward precision interventions and, ultimately, achieving remission in severe asthma [86].

3.3. Anti-IgE: Omalizumab

3.3.1. Mechanism of Action and Efficacy

Omalizumab is a recombinant humanized monoclonal antibody targeting immunoglobulin E (IgE), and it was the first biologic to receive approval for asthma treatment in both the U.S. and Europe. Since approximately 70% of asthma cases are linked to allergic mechanisms, IgE plays a pivotal role in the inflammatory cascade underlying these conditions [87,88]. Produced by B lymphocytes in response to allergen exposure, IgE binds to high-affinity receptors (FcεRI) located on mast cells and basophils, promoting the release of inflammatory mediators. Omalizumab interrupts this process by blocking IgE from binding to its receptor, thereby attenuating downstream allergic signaling [89,90]. Additionally, it reduces the expression of FcεRI receptors on effector cells, further dampening the immune response [90] (Figure 2).
Interestingly, beyond these established pathways, clinical research has noted a decrease in asthma exacerbations during viral seasons among patients on omalizumab. This effect has been associated with increased production of interferon-alpha (IFN-α) in response to rhinovirus, suggesting the antibody may also enhance antiviral immunity [91].
Omalizumab has been utilized in clinical practice for over 15 years, demonstrating consistent efficacy in allergic asthma management. A comprehensive Cochrane meta-analysis conducted in 2014 analyzed 25 randomized controlled trials and confirmed that omalizumab significantly reduced asthma exacerbations (by roughly 25%), decreased hospital admissions, and enabled reductions in inhaled corticosteroid (ICS) usage [92,93,94,95,96]. While some studies have noted slight improvements in pulmonary function, others did not observe significant changes [97]. Data supporting reductions in oral corticosteroid (OCS) use remain inconclusive.
Initial trials mainly included patients with moderate allergic asthma; however, more recent studies extended to severe cases, showing comparable effectiveness [98]. Observational studies from real-world settings have similarly reported fewer exacerbations and hospital visits among patients treated with omalizumab [99,100].
Ongoing investigations aim to identify patient profiles that respond best to this therapy. Retrospective assessments indicate that individuals with elevated eosinophil counts and exhaled nitric oxide (FeNO) levels experience more substantial reductions in exacerbation rates [101]. This may be attributable to their initially higher inflammation burden, allowing for greater visible improvement with treatment. Nonetheless, patients with low type 2 (T2) biomarker levels can also derive benefit from omalizumab. A recent pragmatic trial supported its effectiveness across both high and low T2 biomarker groups (absolute eosinophil count < 300 vs. ≥300 cells/μL and FeNO < 25 vs. ≥25 ppb) [100].
Further, the antibody has shown efficacy in patients with IgE levels outside the U.S.-approved range (30–700 IU/mL), including those above 700 IU/mL [100]. Notably, a pilot study found that omalizumab could downregulate FcεRI expression even in nonatopic asthma patients, opening the door for broader therapeutic applications [102].

3.3.2. Indications, Administration, and Safety

In the U.S., omalizumab is authorized for patients aged 6 years and older who have moderate to severe persistent asthma, uncontrolled on ICS therapy, and show evidence of allergic sensitization. Eligibility includes total serum IgE levels ranging from 30 to 1300 IU/mL for children aged 6–11 and 30 to 700 IU/mL for those 12 and older (in the EU, the range extends to 1500 IU/mL).
Administration involves subcutaneous injections every 2 to 4 weeks, with dosing based on patient weight and baseline IgE levels. Routine monitoring of IgE during treatment is not advised. A trial period of 3–6 months is recommended to assess treatment response. If beneficial, long-term continuation is supported by findings from the XPORT study (Xolair Persistency of Response after Long-Term Therapy) [103]. Omalizumab is generally safe, with an anaphylaxis incidence estimated between 0.1% and 0.2% [104]. Due to this rare but serious risk, the FDA mandates a black box warning, and initial administrations should occur in clinical settings equipped to manage allergic emergencies. Patients are advised to remain under observation for 2 h following their first three injections, then for 30 min after subsequent doses (Table 2).

3.4. Anti-Interleukin-5 Monoclonal Antibodies

Targeting eosinophils in severe asthma requires understanding both circulating and airway-resident eosinophil dynamics. While peripheral eosinophil counts (AECs) generally mirror sputum eosinophil levels in patients on low-to-moderate inhaled corticosteroids [108], this correlation breaks down in those maintained on systemic steroids [109]. Sustained AECs above 400 cells/µL usually indicate sputum eosinophilia, but normal AECs do not rule it out. Such mismatches hint at local eosinophil-driven inflammation within the airway wall, mechanisms that anti-IL-5 therapy may not fully suppress and which can perpetuate symptoms [110,111,112] (Figure 2).
Weight-based IV reslizumab demonstrated more uniform suppression, both sputum eosinophils and eosinophil peroxidase levels declined, and asthma control improved [113]. However, without head-to-head trials comparing mepolizumab, reslizumab, and benralizumab under identical enrollment criteria, definitive conclusions about optimal dosing and agent selection remain elusive.
Intriguingly, patients on low-dose mepolizumab sometimes show increased free IL-5 and IgG–IL-5 immune complexes in sputum, despite the presence of drug–cytokine aggregates. Such complexes can paradoxically enhance the bioactivity of IL-5 in vivo, potentially worsening inflammation [114]. These observations, concurrent rises in ILC2 counts, free and Ig-bound IL-5 in those who deteriorate on low-dose anti-IL-5 therapy, underscore the need to tailor both dosing and agent choice to each patient’s eosinophil biology [114]. In one study, a single 750 mg IV dose of mepolizumab in patients with persistent sputum eosinophilia markedly lowered both blood and sputum eosinophils, enabled an 87 percent reduction in oral steroid requirements, and improved asthma control [115]. By contrast, the SIRIUS trial showed only modest steroid-sparing effects when mepolizumab was given as 100 mg SC injections [116]. A small series of ten patients on the 100 mg dose experienced significant falls in AEC but had persisting or even increased sputum eosinophils that correlated with clinical exacerbations [111]. This suggests low-dose SC mepolizumab may not fully inhibit airway eosinophilopoiesis, likely driven by local IL-5–producing ILC2s and eosinophil progenitors [111,112] (Table 3).

3.4.1. Mepolizumab

Eosinophil activation within the bronchial walls contributes to airway constriction, excessive mucus production, and structural changes over time. Consequently, therapies targeting eosinophils have been central to asthma care. Among the many cytokines and surface molecules that govern eosinophil survival, growth, and activation, interleukin-5 (IL-5) and its receptor have been studied most intensively (Figure 2).
Mepolizumab (brand name Nucala) became the inaugural anti-IL-5 monoclonal antibody approved for adjunctive, long-term treatment of severe eosinophilic asthma. By blocking IL-5 from engaging its receptor (IL-5Rα) on eosinophils, mepolizumab disrupts the maturation of these cells in the bone marrow and leads to reduced deposition of extracellular matrix components in the reticular basement membrane of the airway mucosa. Mepolizumab has a favorable safety profile, with the most common adverse effects being headache, injection site reactions, and back pain. Hypersensitivity reactions are rare. Long-term safety data have not revealed new safety signals [121] (Table 4).
In addition to severe eosinophilic asthma, mepolizumab is also approved for the treatment of eosinophilic granulomatosis with polyangiitis (EGPA), further broadening its clinical utility in patients with overlapping comorbidities [122].
Table 4. Key studies supporting the clinical use of mepolizumab in severe asthma.
Table 4. Key studies supporting the clinical use of mepolizumab in severe asthma.
Study/Trial NameStudy TypeKey FindingsReference
General IL-5 ResearchMechanistic/PreclinicalIL-5 is critical for eosinophil activation, recruitment, and survival.[123]
DREAM Study
(Phase 2)
Randomized Controlled TrialIdentified elevated eosinophils and prior exacerbations as predictors for better response to Mepolizumab.[124]
SIRIUS Trial
(Phase 3)
Randomized Controlled TrialShowed reduction in oral corticosteroid (OCS) use with Mepolizumab therapy.[125]
MUSCA Study
(Phase 3)
Randomized Controlled TrialDemonstrated improved quality of life and safety profile comparable to placebo.[126]
MENSA Trial
(Phase 3)
Randomized Controlled TrialReported a 53% reduction in asthma exacerbations with Mepolizumab vs. placebo in patients on high-dose ICS + LABA.[124]
Clinical Use OverviewReal-world/Approved IndicationApproved for patients ≥ 6 years old with eosinophils ≥ 150 cells/μL; administered as 100 mg SC every 4 weeks.[124,125,126,127]
Comparative Use StudyObservational/Clinical PracticeRecommended for patients with poor response to Omalizumab; effective in both allergic and non-allergic asthma.[128,129]
Off-label ApplicationRegulatory/Clinical SummaryCurrently the only approved anti-IL-5 agent for eosinophilic granuloma and vasculitis.[130]

3.4.2. Reslizumab

Reslizumab (brand name Cinqair) gained FDA approval in 2016 as an add-on therapy for severe eosinophilic asthma. Its action hinges on binding free interleukin-5, thereby preventing this cytokine from engaging its receptor on eosinophils (Figure 2). Since IL-5 is a key driver of eosinophil maturation, recruitment, activation, and longevity, sequestering it disrupts these processes and leads to a reduction in eosinophil numbers and activity. The most frequently reported adverse effects are oropharyngeal pain, increased creatine phosphokinase, and myalgia. Anaphylaxis has been reported in rare cases. A comprehensive evaluation of the long-term safety profile is still ongoing [131] (Table 5).

3.4.3. Benralizumab

Benralizumab is a humanized monoclonal antibody that binds selectively to the α-chain of the interleukin-5 receptor (IL-5Rα), blocking IL-5 signaling on eosinophils, basophils, and ILC2s (Figure 2). By engaging FcγRIIIa on natural killer cells, it induces antibody-dependent cell-mediated cytotoxicity, leading to targeted eosinophil apoptosis [137]. The drug is approved for patients aged 12 years and older with moderate to severe asthma and a baseline peripheral blood eosinophil count of at least 150 cells/µL.
Benralizumab is indicated for individuals aged 12 years and above whose asthma remains uncontrolled and who have an absolute eosinophil count exceeding 300 cells/µL [138]. The recommended regimen begins with a 30 mg subcutaneous injection once every four weeks for the initial three doses, an induction phase aimed at depleting eosinophils in the tissues, followed by maintenance injections every eight weeks. A minimum treatment period of four months is advised to evaluate therapeutic effectiveness. Overall, benralizumab is well tolerated, although rare hypersensitivity events such as anaphylaxis, angioedema, and urticaria have been reported (Table 6).
More recently, benralizumab has also demonstrated efficacy in eosinophilic granulomatosis with polyangiitis (EGPA), providing an alternative therapeutic option for patients with this comorbidity [139].
Table 6. Summary of key clinical evidence supporting benralizumab in severe eosinophilic asthma.
Table 6. Summary of key clinical evidence supporting benralizumab in severe eosinophilic asthma.
Study/Trial NameStudy TypeKey FindingsReference
General Efficacy OverviewClinical RecommendationApproved for patients aged ≥ 12 with eosinophils > 300 cells/μL unresponsive to GINA Step 4–5 therapies.[140]
SIROCCO Trial (Phase 3)RCT, Placebo-Controlled (n = 1204)Achieved 51% reduction in annual asthma exacerbations and improved FEV1 in patients with high eosinophil counts.[141]
CALIMA Trial (Phase 3)RCTShowed reduced exacerbations and enhanced lung function after 56 weeks of treatment.[142]

3.5. Dupilumab

Dupilumab exerts its effects by binding to the α-subunit of the interleukin-4 receptor (IL-4Rα), thereby simultaneously interrupting both IL-4 and IL-13 signaling pathways. The type 1 IL-4 receptor, composed of IL-4Rα paired with the common γ-chain, is expressed on B and T lymphocytes, monocytes, eosinophils, and fibroblasts (Figure 2). The type 2 receptor, a heterodimer of IL-4Rα and IL-13Rα1, is found on monocytes, fibroblasts, eosinophils, activated B cells, epithelial and goblet cells, and airway smooth muscle. IL-4 can engage both receptor types, whereas IL-13 signals solely through the type 2 complex. By targeting the shared IL-4Rα component, dupilumab blocks downstream cascades initiated by both cytokines [143]. IL-4 is crucial for TH2 differentiation and expansion, B-cell proliferation, class-switch recombination to IgE, and eosinophil migration. IL-13 overlaps in function but additionally drives goblet cell hyperplasia, mucus hypersecretion, and smooth muscle contraction and hypertrophy. Both cytokines also induce epithelial and endothelial cells to secrete chemokines such as eotaxin-3, which guides eosinophils to inflamed tissues. IL-4 further upregulates adhesion molecules on the endothelium, facilitating eosinophil extravasation under the direction of chemokines like TARC [144,145,146]. Because IL-4 and IL-13 share many activities, inhibiting only one of these cytokines yields incomplete suppression of type 2 inflammation. Dupilumab’s dual blockade of IL-4 and IL-13 signaling via IL-4Rα is therefore necessary to fully interrupt key pathogenic processes—TH2-driven antigen presentation, eosinophil recruitment to the lungs, and the production of inflammatory mediators and chemokines. By targeting this central IL-4/IL-13 axis, dupilumab addresses fundamental mechanisms common to a range of type 2 inflammatory disorders, mitigating epithelial hyperplasia, basement membrane thickening, barrier disruption, and tissue infiltration that underlie clinical symptoms. The safety profile of dupilumab is well established; most adverse events are mild, including injection site reactions and transient eosinophilia. Conjunctivitis is also reported more frequently than with a placebo [147] (Table 7). Recent evidence suggests that dupilumab may be effective even in patients who have not achieved adequate disease control with anti-IL-5 or anti-IL-5R therapies. In a recent study, patients with severe eosinophilic asthma who experienced a suboptimal response to anti-IL-5/5R biologics demonstrated clinically meaningful improvements following a switch to anti-IL-4R therapy with dupilumab [148].

3.6. Tezepelumab

Tezepelumab (TZP) is a fully human IgG2κ monoclonal antibody designed to occupy the thymic stromal lymphopoietin (TSLP) binding site required for heterodimeric TSLP receptor (TSLPR) engagement, thereby blocking TSLP–TSLPR signaling (Figure 2) [156]. Both TSLPR and interleukin-7 receptor (IL-7R) co-localize on eosinophil surfaces, rendering these cells responsive to TSLP’s survival and activation cues, which include release of cytotoxic granule proteins and proinflammatory chemokines [157,158]. Signaling downstream of TSLPR normally proceeds via MAP kinase and NF-κB pathways, but TZP’s antagonism interrupts this cascade, resulting in broad suppression of cytokine production and inflammatory cell activation [158]. Clinical studies have demonstrated that TZP significantly lowers tissue eosinophil counts, blood eosinophil counts (BECs), total IgE, IL-5, IL-13, and fractional exhaled nitric oxide (FeNO), while also promoting restoration of epithelial barrier integrity [159]. Furthermore, recent real-world evidence suggests that higher baseline T2 biomarker levels are associated with a greater likelihood of clinical remission with tezepelumab in patients with severe uncontrolled asthma [160]. In 2018, the U.S. FDA granted tezepelumab breakthrough therapy status for patients with severe asthma lacking a high type 2 phenotype, who remain symptomatic despite treatment with inhaled corticosteroids plus long-acting β2-agonists (with or without oral corticosteroids) and additional controllers [161]. To date, TZP stands alone among biologics in offering targeted therapy for type 2–low asthma, though it may also be considered for patients exhibiting both atopy and eosinophilia. Tezepelumab is associated with a low rate of adverse events, most commonly nasopharyngitis and headache. No new safety concerns have been identified in ongoing extension studies, but safety profile evaluation is still ongoing.
In addition to asthma, tezepelumab has shown clinical benefit in patients with nasal polyposis, expanding its potential indications, particularly in those with overlapping upper and lower airway disease [162].

4. Perspectives

4.1. Depemokimab-Anti-IL5

Surveys and discrete choice experiments across various therapeutic fields, including severe asthma, consistently show that both patients and clinicians favor regimens requiring fewer injections [163]. Depemokimab, a next-generation anti-IL-5 monoclonal antibody, exhibits roughly 29-fold greater potency and about half the clearance rate of mepolizumab in single-dose pharmacokinetic and pharmacodynamic evaluations performed in cynomolgus monkeys (GSK data on file) (Figure 2). In an initial human trial involving individuals with mild to moderate asthma and baseline blood eosinophil counts ≥ 200 cells/μL, subcutaneous depemokimab (doses 2–300 mg) displayed a favorable safety profile, predictable, dose-proportional pharmacokinetics, and an extended terminal half-life of 38–53 days, substantially longer than the 16–22 days for mepolizumab and 15.5 days for benralizumab, supporting dosing intervals of up to six months [164,165]. That first-in-human study used subcutaneous injections prepared from vials by specialized laboratory and pharmacy teams [164]. In routine practice, most biologics for severe asthma are delivered subcutaneously, either in the patient’s home or a clinic, via a prefilled syringe, which can be incorporated into a safety syringe device (SSD) or an autoinjector (AI) [166]. With an SSD, the user inserts the needle into the skin, depresses the plunger to administer the drug, and the needle automatically retracts into the housing afterward. In contrast, an AI conceals the needle entirely and deploys the medication automatically upon activation. Both delivery systems facilitate convenient therapy administration, though individual preferences among patients and healthcare providers may differ based on factors such as simplicity of use and perceived injection discomfort [167,168,169].
Existing biologic treatments for asthma require injections every four to eight weeks [170]. Due to its prolonged elimination half-life, depemokimab has the potential to extend this interval further. Currently, multiple Phase III studies are evaluating depemokimab in various eosinophil-driven conditions: asthma (NCT05243680, NCT04718389), chronic rhinosinusitis with nasal polyps (NCT05274750, NCT05281523), eosinophilic granulomatosis with polyangiitis (NCT05263934), and hypereosinophilic syndrome (NCT05334368). A separate Phase III trial, NIMBLE, is investigating the effects—both in terms of efficacy and safety—of switching patients from existing anti-IL-5 therapies (mepolizumab or benralizumab) to depemokimab, with topline data anticipated in late 2026. In addition, allowing patients to choose among several injection sites can be beneficial for personal comfort and practicality during self-administration, especially if certain areas become tender or hard to reach [167,171]. Pharmacokinetic analyses have shown that depemokimab’s absorption and disposition remain consistent whether administered into the upper arm, abdomen, or thigh, independent of the delivery device. Therefore, offering multiple, rotatable injection locations provides greater flexibility and autonomy in patient self-management.

4.2. Itepekimab

IL-33 functions as an alarmin released from airway epithelial and endothelial cells when they experience stress or injury caused by inhaled allergens, viral infections, tobacco smoke, or pollutants. Once secreted, IL-33 binds to the ST2 receptor on immune cells, recruiting the IL-1 receptor accessory protein (IL-1RAcP) and triggering downstream MAP kinase and NF-κB signaling. This activation cascade promotes the secretion of proinflammatory cytokines that initiate and amplify both innate and adaptive immune responses [172] (Figure 2). The specific roles of IL-33, together with other epithelial-derived alarmins such as IL-25 and thymic stromal lymphopoietin, in sustaining chronic airway inflammation in diseases like asthma and COPD, as well as the degree to which these alarmins overlap or compensate for one another, remain active areas of research [173].
Itepekimab (formerly REGN3500 and SAR440340) is a fully human IgG4P monoclonal antibody that targets IL-33. It was developed using VelocImmune® technology, which employs a specially engineered mouse with a humanized immune system to generate high-affinity human antibodies [174,175]. The IgG4P format features a serine-to-proline mutation in the hinge region, promoting stable dimer formation [176]. In a preclinical house dust mite (HDM) mouse model of airway inflammation, itepekimab’s blockade of IL-33 significantly attenuated lung tissue injury and reduced inflammatory cell infiltration, supporting its potential to disrupt epithelial-driven inflammatory pathways in respiratory diseases [177]. Currently, the Phase III AERIFY-1 (NCT04701983) and AERIFY-2 (NCT04751487) trials are evaluating itepekimab’s safety and efficacy in COPD patients.
Pharmacokinetic analyses revealed that, after a single intravenous dose in healthy volunteers and following multiple doses in individuals with moderate asthma, itepekimab displayed linear, dose-proportional kinetics and a long elimination half-life, consistent with once-monthly or less frequent dosing schedules. Early single-dose cohorts (0.3 mg/kg) prompted extended follow-up for later cohorts to ensure thorough characterization of PK and pharmacodynamic (PD) endpoints [178]. No treatment-emergent anti-drug antibodies (ADAs) were detected, and itepekimab was generally well tolerated across all studies.
Baseline total IL-33 levels in serum were typically undetectable in both healthy subjects and those with asthma, mirroring findings in COPD populations with low systemic IL-33 concentrations [179]. Upon itepekimab administration, serum IL-33 levels rose, reflecting formation of a stable IL-33–itepekimab complex. Given the negligible free IL-33 at baseline, the increase in detectable total IL-33 post-treatment likely represents bound alarmin, serving as a pharmacodynamic marker of target engagement. Notably, the decline of total IL-33 concentrations occurred more slowly than itepekimab’s own half-life, suggesting that the clearance of the IL-33–itepekimab complex is protracted relative to the antibody alone. Dose-dependent elevations in total IL-33 were seen in the single-dose study; profiles for participants receiving 3 mg/kg versus 10 mg/kg intravenous doses overlapped, indicating saturation of IL-33 binding at higher doses [180].
In summary, itepekimab exhibits favorable tolerability, robust subcutaneous absorption, linear and dose-proportional pharmacokinetics, and no detectable ADA induction in both healthy volunteers and moderate asthma patients. The observed increases in total serum IL-33 confirm effective target engagement, and concomitant reductions in blood eosinophil counts demonstrate pharmacodynamic activity.

4.3. CM310-Stapokibart

Stapokibart is a humanized monoclonal antibody directed against IL-4Rα, engineered to inhibit both IL-4 and IL-13 signaling, in contrast to dupilumab, which is fully human (Figure 2). Its safety and efficacy have been evaluated in healthy volunteers and in patients with atopic dermatitis (AD) or chronic rhinosinusitis with nasal polyps (CRSwNP) during phase 1b/2a (NCT04893941), phase 2b (NCT04805411), and phase 2 (NCT04805398) trials. In a study targeting severe eosinophilic CRSwNP, stapokibart significantly decreased nasal polyp size and improved nasal congestion, both coprimary endpoints [181]. For moderate to severe AD, a phase III trial demonstrated that 66.9 percent of stapokibart-treated participants achieved EASI-75 and 44.2 percent reached an IGA score of 0/1 (≥2-point reduction), outperforming placebo rates of 25.8 percent and 16.1 percent, respectively [182]. Details regarding stapokibart’s underlying mechanism and preclinical data have not yet been publicly disclosed. Although stapokibart and dupilumab have not been directly compared in the same trial, separate studies suggest comparable efficacy in type 2 inflammatory conditions. In the CROWNS-1 phase II trial for severe CRSwNP, 79 percent (22/28) of patients receiving stapokibart experienced at least a 2-point reduction in Nasal Polyp Score by week 16 [181]. By comparison, dupilumab achieved this outcome in 46 percent of participants in the SINUS-24 trial (66/143) and 46 percent in SINUS-52 (136/295) [183]. For moderate to severe AD, 66.9 percent of stapokibart recipients met EASI-75 in its phase III study, versus 57.3 percent for dupilumab in a separate trial with a similar ethnic composition; placebo rates were 25.8 percent and 14.5 percent, respectively. Similarly, IGA 0/1 (≥2-point reduction) occurred in 44.2 percent of stapokibart-treated subjects compared to 26.8 percent with dupilumab, while placebo groups showed 16.1 percent and 4.8 percent, respectively [184]. Definitive head-to-head trials are needed to confirm these observations. In vitro assays reveal stapokibart’s ability to block IL-4 and IL-13/IL-13Rα1 binding to IL-4Rα, matching or slightly surpassing dupilumab’s neutralizing potency. In vivo pharmacology studies demonstrate that stapokibart’s IL-4/IL-13 blockade effectively protects rats in various type 2 allergy models. Toxicology evaluations further indicate a favorable safety margin, with a no-observed-adverse-effect level (NOAEL) of 150 mg/kg when dosed weekly for 26 weeks.

4.4. Tralokinumab

Tralokinumab underwent evaluation in three Phase II studies involving asthma patients (NCT00640016, NCT00873860 [185], and NCT01402986 [186]), although one trial was terminated early due to enrollment difficulties. In the two trials that completed enrollment (NCT00873860 and NCT01402986), the primary endpoints were not reached when assessing all enrolled participants. Nonetheless, both studies demonstrated that inhibiting IL-13 with tralokinumab led to improvements in pulmonary function among individuals with moderate to severe asthma that was not adequately controlled on medium- or high-dose ICS/LABA. Post hoc analyses from these trials revealed that patients exhibiting biomarkers indicative of IL-13 activity experienced greater clinical benefit [185,186]. In the Phase IIa trial (NCT00873860), a subgroup of moderate to severe uncontrolled asthmatics who had elevated sputum IL-13 at baseline showed trends toward meaningful enhancements in Asthma Control Questionnaire (ACQ-6) scores and FEV1 when compared with placebo [185] (Figure 2). Recognizing that sputum collection is impractical in larger studies, the Phase IIb trial (NCT01402986) instead used serum periostin and DPP-4 levels as surrogate markers for IL-13 signaling [186]. Post hoc findings in this cohort, specifically participants with severe uncontrolled asthma, baseline FEV1 reversibility ≥ 12%, no regular OCS use at entry, and elevated DPP-4 or periostin, showed that 300 mg of tralokinumab administered subcutaneously every two weeks yielded improvements in lung function, exacerbation frequency, symptom control, and quality of life [186]. Across both Phase II trials, no new safety concerns emerged: in the Phase IIa study, no serious adverse events (AEs) were attributed to tralokinumab, and in Phase IIb, treatment-emergent AEs occurred at similar rates in both tralokinumab and placebo arms [185].
A combined pharmacokinetic–pharmacodynamic (PK–PD) model applied to these Phase II data predicted that a 300 mg subcutaneous dose every two weeks would achieve near-maximal FEV1 increases, leading to its selection for the Phase III program [187]. The ATMOSPHERE trials were designed to identify whether a subset of patients would exhibit an enhanced response to tralokinumab, potentially enabling more targeted management of IL-13-driven asthma. Previous attempts with other IL-13-targeted agents have produced mixed results: lebrikizumab, for instance, failed to show consistent benefit in both all-comers and serum periostin- or eosinophil-high subgroups during Phase III trials [188], whereas dupilumab, targeting both IL-4 and IL-13, demonstrated efficacy in a broad severe asthma population as well as in those with elevated blood eosinophils in a Phase IIb trial [189]. The experience with tralokinumab mirrors that of lebrikizumab, as no clearly defined patient subset has yet been identified that consistently achieves a robust, clinically meaningful response (Table 8).

4.5. Lebrikizumab

Lebrikizumab is an IgG4 monoclonal antibody that binds IL-13 with very high affinity and a slow dissociation rate, selectively blocking IL-13 signaling via the IL-13Rα1/IL-4Rα receptor complex and thus potently inhibiting downstream IL-13-mediated effects. Because its activity is confined to IL-13 neutralization, lebrikizumab was hypothesized to benefit patients exhibiting a type 2 inflammatory signature (Figure 2). To evaluate this, trials were conducted in individuals with moderate to severe asthma that remained uncontrolled despite standard therapies. However, Phase III studies did not consistently demonstrate reductions in exacerbation rates across the broad asthmatic population [193,194], calling into question whether isolated IL-13 blockade, without concurrent IL-4 inhibition, would suffice in difficult-to-treat cases. In the ACOUSTICS trial, adolescents received lebrikizumab over a 52-week, placebo-controlled period. Those treated with 125 mg (n = 116) showed a greater decrease in annualized exacerbation rate (AER) compared to placebo than did the 37.5 mg cohort (n = 113). Specifically, the adjusted rate ratios (RRs) versus placebo were 0.49 (95% CI, 0.28–0.83) for 125 mg and 0.60 (95% CI, 0.35–1.03) for 37.5 mg, corresponding to 51% and 40% reductions in AER, respectively. When focusing on participants with baseline blood eosinophils ≥ 300 cells/μL and at least one exacerbation in the prior year, the AER declined even further: RR = 0.41 (95% CI, 0.19–0.88) for 125 mg and RR = 0.36 (95% CI, 0.15–0.87) for 37.5 mg, representing reductions of 59% and 64% compared to placebo [193,194].
A secondary endpoint in the pooled LAVOLTA trials assessed lung function via absolute change from baseline in pre-bronchodilator FEV1. Post hoc analyses revealed that both 125 mg and 37.5 mg doses of lebrikizumab yielded greater FEV1 improvements versus placebo, suggesting that, when targeted to patients with prior exacerbations and evidence of type 2 inflammation, lebrikizumab can be effective [195,196].
The data indicates that selective IL-13 inhibition significantly lowers exacerbation risk in moderate to severe eosinophilic asthma. Still, these findings cannot be generalized to other IL-13 inhibitors like tralokinumab, because binding characteristics differ markedly. In vitro assays show that lebrikizumab’s affinity for IL-13 is approximately 140-fold greater than that of tralokinumab, and IL-13 dissociates more slowly from lebrikizumab than from tralokinumab [197].

5. Real-World Evidence for Biologic Therapies in Severe Asthma

While RCTs have established the efficacy and safety of biologic therapies in severe asthma, patients enrolled in RCTs often represent a select population that may not reflect the full spectrum of disease encountered in routine clinical practice. Real-world evidence (RWE) studies have become increasingly important in validating the generalizability of trial findings and assessing the effectiveness of these therapies across more diverse patient groups, including those with multiple comorbidities, varying adherence, and more complex phenotypes. One notable example is the REALITI-A study, a prospective, international observational cohort that evaluated mepolizumab in patients with severe eosinophilic asthma [198]. This study confirmed significant reductions in exacerbation rates and oral corticosteroid use, as well as improvements in asthma control, mirroring outcomes from pivotal RCTs. Similarly, the CHRONICLE study [199] provided key insights into the real-world use of benralizumab, mepolizumab, and reslizumab in the United States, demonstrating substantial reductions in exacerbations and OCS dependence among biologic-treated patients.
These and other RWE publications highlight that biologic therapies remain effective and well tolerated even in broader, less-selected asthma populations. The findings support the utility of blood eosinophil count, exacerbation history, and other biomarkers as predictors of response, not only in clinical trials but also in daily practice. Incorporating real-world evidence thus strengthens the case for personalized and biomarker-driven use of biologics in severe asthma.

6. Methodology

A comprehensive literature search was performed in accordance with PRISMA guidelines, utilizing PubMed, Google Scholar, and ScienceDirect as primary databases. Emphasis was placed on articles published within the past five years to capture the most current insights into asthma immunobiology and biologic therapies, although foundational studies predating this window were included when they provided essential mechanistic or clinical context. Search terms combined core disease descriptors—such as “asthma”, “airway inflammation”, “cytokines”, “type 2 inflammation”, and “asthma exacerbations”—with names of established and investigational biologics (e.g., “omalizumab”, “mepolizumab”, “benralizumab”, “depemokimab”, “itepekimab”, “dupilumab”, “tralokinumab”, “lebrikizumab”, “tezepelumab”, “stapokibart”) as well as key upstream mediators (“IL-4”, “IL-5”, “IL-13”, “IL-33”, “TSLP”). In addition, combinations of “biomarkers”, “eosinophils”, “IgE”, “FeNO”, “clinical trials”, and “mechanism of action” were employed to ensure comprehensive coverage of preclinical and clinical data. Inclusion criteria required studies to address one or more of the following: asthma pathophysiology (particularly pro-/anti-inflammatory dynamics and the role of allergic sensitization), cytokine-mediated mechanisms, exacerbation biology, biomarker-driven phenotyping, or the efficacy, safety, and pharmacokinetics of biologic agents. Review articles, randomized controlled trials, observational cohorts, and mechanistic investigations were considered, provided they offered substantive data on biologic therapy selection or outcomes. Exclusion criteria ruled out non-peer-reviewed content, case reports with fewer than ten participants, non-English publications, and studies lacking relevance to severe or poorly controlled asthma. Initial identification via Google Scholar was followed by targeted retrieval of full texts from PubMed and ScienceDirect, with cross-referencing to capture any additional pertinent publications (Figure 3).

7. Conclusions

Asthma inherent heterogeneity has long challenged clinicians, as traditional anti-inflammatory and bronchodilator regimens fail to address the diverse immunological pathways underpinning severe disease. The advent of biologic therapies marks a paradigm shift, enabling direct interception of key molecular drivers, namely IgE, IL-5, IL-4, IL-13, IL-33, and TSLP, that orchestrate airway inflammation, remodeling, and exacerbations. By leveraging biomarkers such as blood and sputum eosinophils, serum IgE, and fractional exhaled nitric oxide, clinicians can more precisely identify patients whose disease is driven by type 2 cytokines or alarmin pathways, thereby optimizing the selection of agents most likely to yield clinical benefit.
Anti-IgE therapy established proof of concept, demonstrating reduced exacerbation rates and improved lung function in allergic asthmatics. Subsequent anti-IL-5 agents (mepolizumab, benralizumab, reslizumab, and now depemokimab) extend this success to eosinophilic phenotypes, with trials confirming significant steroid-sparing effects and fewer hospitalizations. The longer half-life of newer constructs like depemokimab supports extended dosing intervals, easing treatment burden. Targeting upstream alarmins—through agents such as itepekimab (anti-IL-33) and anti-TSLP antibodies—shows promise in mitigating tissue eosinophilia and broad inflammatory cascades before they fully develop. Meanwhile, dual IL-4Rα blockers (dupilumab, stapokibart) and IL-13-specific inhibitors (tralokinumab, lebrikizumab) highlight the critical interplay between IL-4 and IL-13 in sustaining type 2 inflammation, though their ultimate efficacy hinges on careful patient stratification via biomarker profiles.
Collectively, these biologics illustrate how elucidating asthma’s molecular heterogeneity enables tailored interventions that not only improve symptom control and lung function but also reduce exacerbations and healthcare utilization. Future research should focus on head-to-head comparisons, refinement of predictive biomarkers, and exploration of combination strategies to achieve durable remission. As our understanding of airway immunobiology deepens, integrating novel biologic agents into treatment algorithms will be essential to deliver truly personalized, mechanism-based care for patients with severe asthma.

Author Contributions

Conceptualization, R.M.V., C.E.S., K.R.; Methodology, D.D., N.S.A.; Software, N.E.C., K.R.; Validation, R.M.V., G.-A.S.; Formal analysis, D.M.T., D.D.; Investigation, N.S.A., K.R.; Resources, C.E.S., N.E.C.; Data curation, D.M.T., G.-A.S.; Writing—original draft preparation, K.R., D.D., N.E.C.; Writing—review and editing, N.S.A., R.M.V., C.E.S.; Visualization, K.R., G.-A.S., D.M.T.; Supervision, R.M.V., N.E.C.; Project administration, D.M.T., C.E.S.; Funding acquisition, D.D., G.-A.S. All authors have read and agreed to the published version of the manuscript.

Funding

The Article Processing Charges were funded by the University of Medicine and Pharmacy of Craiova, Romania.

Data Availability Statement

No new data were created or analyzed in this study.

Acknowledgments

There was no support received (administrative and technical support, or donations in kind) to realize the study.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Collaborators GBDS. Global, regional, and national burden of stroke and its risk factors, 1990–2019: A systematic analysis for the Global Burden of Disease Study 2019. Lancet Neurol. 2021, 20, 795–820. [Google Scholar] [CrossRef]
  2. Meghji, J.; Mortimer, K.; Agusti, A.; Allwood, B.W.; Asher, I.; Bateman, E.D.; Bissell, K.; Bolton, C.E.; Bush, A.; Celli, B.; et al. Improving lung health in low-income and middle-income countries: From challenges to solutions. Lancet 2021, 397, 928–940. [Google Scholar] [CrossRef]
  3. Venkatesan, P. 2023 Gina report for asthma. Lancet Respir. Med. 2023, 11, 589. [Google Scholar] [CrossRef] [PubMed]
  4. Wills-Karp, M.; Luyimbazi, J.; Xu, X.; Schofield, B.; Neben, T.Y.; Karp, C.L.; Donaldson, D.D. Interleukin-13: Central mediator of allergic asthma. Science 1998, 282, 2258–2261. [Google Scholar] [CrossRef] [PubMed]
  5. GrünIg, G.; Warnock, M.; Wakil, A.E.; Venkayya, R.; Brombacher, F.; Rennick, D.M.; Sheppard, D.; Mohrs, M.; Donaldson, D.D.; Locksley, R.M.; et al. Requirement for IL-13 independently of IL-4 in experimental asthma. Science 1998, 282, 2261–2263. [Google Scholar] [CrossRef] [PubMed]
  6. Lambrecht, B.N.; Hammad, H.; Fahy, J.V. The cytokines of asthma. Immunity 2019, 50, 975–991. [Google Scholar] [CrossRef]
  7. Ohki, Y.; Tokuyama, K.; Mayuzumi, H.; Sato, A.; Koyama, H.; Takizawa, T.; Arakawa, H.; Mochizuki, H.; Morikawa, A. Characteristic features of allergic airway inflammation in a murine model of infantile asthma. Int. Arch. Allergy Immunol. 2005, 138, 51–58. [Google Scholar] [CrossRef]
  8. Fahy, J.V. Type 2 inflammation in asthma–present in most, absent in many. Nat. Rev. Immunol. 2015, 15, 57–65. [Google Scholar] [CrossRef]
  9. Sze, E.; Bhalla, A.; Nair, P. Mechanisms and therapeutic strategies for non-T2 asthma. Allergy 2020, 75, 311–325. [Google Scholar] [CrossRef]
  10. Bikov, A.; Ŏguzülgen, I.K.; Baiardini, I.; Contoli, M.; Emelyanov, A.; Fassio, O.; Ivancevich, J.C.; Kaidashev, I.; Kowal, K.; Labor, M.; et al. Beliefs and Preferences Regarding Biological Treatments for Severe Asthma. World Allergy Organ. J. 2020, 13, 100441. [Google Scholar] [CrossRef]
  11. Olin, J.T.; Wechsler, M.E. Asthma: Pathogenesis and Novel Drugs for Treatment. BMJ 2014, 349, g5517. [Google Scholar] [CrossRef] [PubMed]
  12. Bender, B.; Oppenheimer, J.; George, M.; Brown, R.; Patel, A.; Hill, T.; Boe, A.; Herrera, E.M.; Dotiwala, Z.; Casciano, J.; et al. Assessment of Real-World Escalation to Biologics in US Patients with Asthma. J. Allergy Clin. Immunol. Pract. 2022, 10, 2941–2948. [Google Scholar] [CrossRef]
  13. Ogulur, I.; Pat, Y.; Ardicli, O.; Barletta, E.; Cevhertas, L.; Fernandez-Santamaria, R.; Huang, M.; Bel Imam, M.; Koch, J.; Ma, S.; et al. Advances and highlights in biomarkers of allergic diseases. Allergy 2021, 76, 3659–3686. [Google Scholar] [CrossRef] [PubMed]
  14. Mavissakalian, M.; Brady, S. The Current State of Biologic Therapies for Treatment of Refractory Asthma. Clin. Rev. Allergy Immunol. 2020, 59, 195–207. [Google Scholar] [CrossRef]
  15. Whittington, M.D.; McQueen, R.B.; Ollendorf, D.A.; Tice, J.A.; Chapman, R.H.; Pearson, S.D.; Campbell, J.D. Assessing the Value of Mepolizumab for Severe Eosinophilic Asthma: A Cost-Effectiveness Analysis. Ann. Allergy Asthma Immunol. 2017, 118, 220–225. [Google Scholar] [CrossRef]
  16. Jeffery, M.M.; Inselman, J.W.; Maddux, J.T.; Lam, R.W.; Shah, N.D.; Rank, M.A. Asthma Patients Who Stop Asthma Biologics Have a Similar Risk of Asthma Exacerbations as Those Who Continue Asthma Biologics. J. Allergy Clin. Immunol. Pract. 2021, 9, 2742–2750. [Google Scholar] [CrossRef]
  17. Opina, M.T.; Moore, W.C. Phenotype-Driven Therapeutics in Severe Asthma. Curr. Allergy Asthma Rep. 2017, 17, 10. [Google Scholar] [CrossRef] [PubMed]
  18. Barnes, P.J. Cellular and molecular mechanisms of asthma and COPD. Clin. Sci. 2017, 131, 1541–1558. [Google Scholar] [CrossRef]
  19. Harker, J.A.; Lloyd, C.M. T helper 2 cells in asthma. J. Exp. Med. 2023, 220, e20221094. [Google Scholar] [CrossRef]
  20. Finkelman, F.D.; Hogan, S.P.; Hershey, G.K.; Rothenberg, M.E.; Wills-Karp, M. Importance of cytokines in murine allergic airway disease and human asthma. J. Immunol. 2010, 184, 1663–1674. [Google Scholar] [CrossRef]
  21. Szabo, S.J.; Sullivan, B.M.; Peng, S.L.; Glimcher, L.H. Molecular mechanisms regulating th1 immune responses. Annu. Rev. Immunol. 2003, 21, 713–758. [Google Scholar] [CrossRef]
  22. Gavett, S.H.; O’Hearn, D.J.; Li, X.; Huang, S.K.; Finkelman, F.D.; Wills-Karp, M. Interleukin 12 inhibits antigen-induced airway hyperresponsiveness, inflammation, and th2 cytokine expression in mice. J. Exp. Med. 1995, 182, 1527–1536. [Google Scholar] [CrossRef] [PubMed]
  23. Cohn, L.; Homer, R.J.; Niu, N.; Bottomly, K. T helper 1 cells and interferon gamma regulate allergic airway inflammation and mucus production. J. Exp. Med. 1999, 190, 1309–1318. [Google Scholar] [CrossRef] [PubMed]
  24. Wisniewski, J.A.; Muehling, L.M.; Eccles, J.D.; Capaldo, B.J.; Agrawal, R.; Shirley, D.A.; Patrie, J.T.; Workman, L.J.; Schuyler, A.J.; Lawrence, M.G.; et al. T(H)1 signatures are present in the lower airways of children with severe asthma, regardless of allergic status. J. Allergy Clin. Immunol. 2018, 141, 2048–2060.e13. [Google Scholar] [CrossRef]
  25. Britt, R.D., Jr.; Thompson, M.A.; Sasse, S.; Pabelick, C.M.; Gerber, A.N.; Prakash, Y.S. Th1 cytokines tnf-alpha and ifn-gamma promote corticosteroid resistance in developing human airway smooth muscle. Am. J. Physiol. Lung Cell. Mol. Physiol. 2019, 316, L71–L81. [Google Scholar] [CrossRef] [PubMed]
  26. Newcomb, D.C.; Peebles, R.S., Jr. Th17-mediated inflammation in asthma. Curr. Opin. Immunol. 2013, 25, 755–760. [Google Scholar] [CrossRef]
  27. Bullone, M.; Carriero, V.; Bertolini, F.; Folino, A.; Mannelli, A.; Di Stefano, A.; Gnemmi, I.; Torchio, R.; Ricciardolo, F.L. Elevated serum IgE, oral corticosteroid dependence and IL-17/22 expression in highly neutrophilic asthma. Eur. Respir. J. 2019, 54, 1900068. [Google Scholar] [CrossRef]
  28. Krishnamoorthy, N.; Douda, D.N.; Brüggemann, T.R.; Ricklefs, I.; Duvall, M.G.; Abdulnour, R.-E.E.; Martinod, K.; Tavares, L.; Wang, X.; Cernadas, M.; et al. Neutrophil cytoplasts induce T(H)17 differentiation and skew inflammation toward neutrophilia in severe asthma. Sci. Immunol. 2018, 3, eaao4747. [Google Scholar] [CrossRef]
  29. Schnyder-Candrian, S.; Togbe, D.; Couillin, I.; Mercier, I.; Brombacher, F.; Quesniaux, V.; Fossiez, F.; Ryffel, B.; Schnyder, B. Interleukin-17 is a negative regulator of established allergic asthma. J. Exp. Med. 2006, 203, 2715–2725. [Google Scholar] [CrossRef]
  30. Wakashin, H.; Hirose, K.; Maezawa, Y.; Kagami, S.-I.; Suto, A.; Watanabe, N.; Saito, Y.; Hatano, M.; Tokuhisa, T.; Iwakura, Y.; et al. IL-23 and th17 cells enhance th2-cell-mediated eosinophilic airway inflammation in mice. Am. J. Respir. Crit. Care Med. 2008, 178, 1023–1032. [Google Scholar] [CrossRef]
  31. Bellini, A.; Marini, M.A.; Bianchetti, L.; Barczyk, M.; Schmidt, M.; Mattoli, S. Interleukin (IL)-4, IL-13, and IL-17a differentially affect the profibrotic and proinflammatory functions of fibrocytes from asthmatic patients. Mucosal Immunol. 2012, 5, 140–149. [Google Scholar] [CrossRef] [PubMed]
  32. Zhao, J.; Lloyd, C.M.; Noble, A. Th17 responses in chronic allergic airway inflammation abrogate regulatory T-cell-mediated tolerance and contribute to airway remodeling. Mucosal Immunol. 2013, 6, 335–346. [Google Scholar] [CrossRef] [PubMed]
  33. Kudo, M.; Melton, A.C.; Chen, C.; Engler, M.B.; Huang, K.E.; Ren, X.; Wang, Y.; Bernstein, X.; Li, J.T.; Atabai, K.; et al. IL-17a produced by alphabeta T cells drives airway hyper-responsiveness in mice and enhances mouse and human airway smooth muscle contraction. Nat. Med. 2012, 18, 547–554. [Google Scholar] [CrossRef]
  34. Veldhoen, M.; Uyttenhove, C.; Van Snick, J.; Helmby, H.; Westendorf, A.; Buer, J.; Martin, B.; Wilhelm, C.; Stockinger, B. Transforming growth factor-beta ‘Reprograms’ the differentiation of T helper 2 cells and promotes an interleukin 9-producing subset. Nat. Immunol. 2008, 9, 1341–1346. [Google Scholar] [CrossRef]
  35. Kaplan, M.H. The transcription factor network in th9 cells. Semin. Immunopathol. 2017, 39, 11–20. [Google Scholar] [CrossRef] [PubMed]
  36. Angkasekwinai, P.; Chang, S.H.; Thapa, M.; Watarai, H.; Dong, C. Regulation of IL-9 expression by IL-25 signaling. Nat. Immunol. 2010, 11, 250–256. [Google Scholar] [CrossRef]
  37. Koch, S.; Sopel, N.; Finotto, S. Th9 and other IL-9-producing cells in allergic asthma. Semin. Immunopathol. 2017, 39, 55–68. [Google Scholar] [CrossRef]
  38. Pajulas, A.; Fu, Y.; Cheung, C.C.; Chu, M.; Cannon, A.; Alakhras, N.; Zhang, J.; Ulrich, B.J.; Nelson, A.S.; Zhou, B.; et al. Interleukin-9 promotes mast cell progenitor proliferation and CCR2-dependent mast cell migration in allergic airway inflammation. Mucosal Immunol. 2023, 16, 432–445. [Google Scholar] [CrossRef] [PubMed]
  39. Sehra, S.; Yao, W.; Nguyen, E.T.; Glosson-Byers, N.L.; Akhtar, N.; Zhou, B.; Kaplan, M.H. Th9 cells are required for tissue mast cell accumulation during allergic inflammation. J. Allergy Clin. Immunol. 2015, 136, 433–440.e1. [Google Scholar] [CrossRef]
  40. Lloyd, C.M.; Harker, J.A. Epigenetic control of interleukin-9 in asthma. N. Engl. J. Med. 2018, 379, 87–89. [Google Scholar] [CrossRef]
  41. Parker, J.M.; Oh, C.K.; LaForce, C.; Miller, S.D.; Pearlman, D.S.; Le, C.; Robbie, G.J.; White, W.I.; White, B.; Molfino, N.A. Safety profile and clinical activity of multiple subcutaneous doses of medi-528, a humanized antiinterleukin-9 monoclonal antibody, in two randomized phase 2a studies in subjects with asthma. BMC Pulm. Med. 2011, 11, 14. [Google Scholar] [CrossRef] [PubMed]
  42. Oh, C.K.; Leigh, R.; McLaurin, K.K.; Kim, K.; Hultquist, M.; Molfino, N.A. A randomized, controlled trial to evaluate the effect of an anti-interleukin-9 monoclonal antibody in adults with uncontrolled asthma. Respir. Res. 2013, 14, 93. [Google Scholar] [CrossRef]
  43. Liu, X.; Fang, L.; Guo, T.B.; Mei, H.; Zhang, J.Z. Drug targets in the cytokine universe for autoimmune disease. Trends Immunol. 2013, 34, 120–128. [Google Scholar] [CrossRef]
  44. Janssens, T.; Ritz, T. Perceived triggers of asthma: Key to symptom perception and management. Clin. Exp. Allergy 2013, 43, 1000–1008. [Google Scholar] [CrossRef] [PubMed]
  45. Al Ghobain, M.O.; Algazlan, S.S.; Oreibi, T.M. Asthma prevalence among adults in Saudi Arabia. Saudi Med. J. 2018, 39, 179–184. [Google Scholar] [CrossRef] [PubMed]
  46. Alqahtani, J.M. Asthma and other allergic diseases among Saudi schoolchildren in Najran: The need for a comprehensive intervention program. Ann. Saudi Med. 2016, 36, 379–385. [Google Scholar] [CrossRef]
  47. Sahoo, A.; Wali, S.; Nurieva, R. T helper 2 and T follicular helper cells: Regulation and function of interleukin 4. Cytokine Growth Factor Rev. 2016, 30, 29–37. [Google Scholar] [CrossRef]
  48. Ramirez, G.A.; Yacoub, M.R.; Ripa, M.; Mannina, D.; Cariddi, A.; Saporiti, N.; Ciceri, F.; Castagna, A.; Colombo, G.; Dagna, L. Eosinophils from physiology to disease: A comprehensive review. BioMed Res. Int. 2018, 2018, 9095275. [Google Scholar] [CrossRef]
  49. Pan, R.; Kuai, S.; Li, Q.; Zhu, X.; Wang, T.; Cui, Y. Diagnostic value of IL-6 for patients with asthma: A meta-analysis. Allergy Asthma Clin. Immunol. 2023, 19, 39. [Google Scholar] [CrossRef]
  50. Pelaia, C.; Heffler, E.; Crimi, C.; Maglio, A.; Vatrella, A.; Pelaia, G.; Canonica, G.W. Interleukins 4 and 13 in asthma: Key pathophysiologic cytokines and druggable molecular targets. Front. Pharmacol. 2022, 13, 851940. [Google Scholar] [CrossRef]
  51. Coomes, S.M.; Kannan, Y.; Pelly, V.S.; Entwistle, L.J.; Guidi, R.; Perez-Lloret, J.; Nikolov, N.; Müller, W.; Wilson, M.S. CD4+ Th2 cells are directly regulated by IL-10 during allergic airway inflammation. Mucosal Immunol. 2017, 10, 150–161. [Google Scholar] [CrossRef]
  52. Shik, D.; Tomar, S.; Lee, J.B.; Chen, C.Y.; Smith, A.; Wang, Y.H. IL-9-producing cells in the development of IgE-mediated food allergy. Semin. Immunopathol. 2017, 39, 69–77. [Google Scholar] [CrossRef]
  53. Evasovic, J.M.; Singer, C.A. Regulation of IL-17A and implications for TGF-β1 co modulation of airway smooth muscle remodeling in severe asthma. Am. J. Physiol. Lung Cell. Mol. Physiol. 2019, 316, L843–L868. [Google Scholar] [CrossRef]
  54. Chan, B.C.L.; Lam, C.W.K.; Tam, L.S.; Wong, C.K. IL33: Roles in allergic inflammation and therapeutic perspectives. Front. Immunol. 2019, 10, 364. [Google Scholar] [CrossRef] [PubMed]
  55. Li, H.; Zhang, C.; Lu, D.; Liu, F.; Wang, C.; Wu, J.; Dong, L. IL-25 promotes Th2 immunity responses in airway inflammation of asthmatic mice via activation of dendritic cells. Inflammation 2014, 37, 1070–1077. [Google Scholar]
  56. Ojiaku, C.A.; Yoo, E.J.; Panettieri, R.A., Jr. Transforming growth factor β1 function in airway remodelling and hyperresponsiveness. The missing link? Am. J. Respir. Cell Mol. Biol. 2017, 56, 432–442. [Google Scholar] [CrossRef] [PubMed]
  57. Zhang, X.; Xu, Z.; Wen, X.; Huang, G.; Nian, S.; Li, L.; Guo, X.; Ye, Y.; Yuan, Q. The onset, development, and pathogenesis of severe neutrophilic asthma. Immunol. Cell Biol. 2022, 100, 144–159. [Google Scholar] [CrossRef]
  58. Hammad, H.; Lambrecht, B.N. The basic immunology of asthma. Cell 2021, 184, 1469–1485. [Google Scholar] [CrossRef]
  59. Smolinska, S.; Antolín-Amérigo, D.; Popescu, F.-D.; Jutel, M. Thymic Stromal Lymphopoietin (TSLP), Its Isoforms and the Interplay with the Epithelium in Allergy and Asthma. Int. J. Mol. Sci. 2023, 24, 12725. [Google Scholar] [CrossRef]
  60. Bai, T.R.; Vonk, J.M.; Postma, D.S.; Boezen, H.M. Severe exacerbations predict excess lung function decline in asthma. Eur. Respir. J. 2007, 30, 452–456. [Google Scholar] [CrossRef]
  61. O’Byrne, P.M.; Pedersen, S.; Lamm, C.J.; Tan, W.C.; Busse, W.W. Severe exacerbations and decline in lung function in asthma. Am. J. Respir. Crit. Care Med. 2009, 179, 19–24. [Google Scholar] [CrossRef] [PubMed]
  62. Miller, M.K.; Lee, J.H.; Miller, D.P.; Wenzel, S.E.; TENOR Study Group. Recent asthma exacerbations: A key predictor of future exacerbations. Respir. Med. 2007, 101, 481–489. [Google Scholar] [CrossRef]
  63. Nicholson, K.G.; Kent, J.; Ireland, D.C. Respiratory viruses and exacerbations of asthma in adults. BMJ 1993, 307, 982–986. [Google Scholar] [CrossRef]
  64. Johnston, S.L.; Pattemore, P.K.; Sanderson, G.; Smith, S.; Campbell, M.J.; Josephs, L.K.; Cunningham, A.; Robinson, B.S.; Myint, S.H.; Ward, M.E.; et al. The relationship between upper respiratory infections and hospital admissions for asthma: A time-trend analysis. Am. J. Respir. Crit. Care Med. 1996, 154, 654–660. [Google Scholar] [CrossRef] [PubMed]
  65. Johnston, N.W.; Johnston, S.L.; Norman, G.R.; Dai, J.; Sears, M.R. The September epidemic of asthma hospitalization: School children as disease vectors. J. Allergy Clin. Immunol. 2006, 117, 557–562. [Google Scholar] [CrossRef] [PubMed]
  66. O’riordan, S.; Barton, M.; Yau, Y.; Read, S.E.; Allen, U.; Tran, D. Risk factors and outcomes among children admitted to hospital with pandemic H1N1 influenza. Can. Med Assoc. J. 2010, 182, 39–44. [Google Scholar] [CrossRef]
  67. Plessa, E.; Diakakis, P.; Gardelis, J.; Thirios, A.; Koletsi, P.; Falagas, M.E. Clinical features, risk factors, and complications among pediatric patients with pandemic influenza A (H1N1). Clin. Pediatr. 2010, 49, 777–781. [Google Scholar] [CrossRef] [PubMed]
  68. Libster, R.; Bugna, J.; Coviello, S.; Hijano, D.R.; Dunaiewsky, M.; Reynoso, N.; Cavalieri, M.L.; Guglielmo, M.C.; Areso, M.S.; Gilligan, T.; et al. Pediatric hospitalizations associated with 2009 pandemic influenza A (H1N1) in Argentina. N. Engl. J. Med. 2010, 362, 45–55. [Google Scholar] [CrossRef]
  69. Yoo, J.-K.; Kim, T.S.; Hufford, M.M.; Braciale, T.J. Viral infection of the lung: Host response and sequelae. J. Allergy Clin. Immunol. 2013, 132, 1263–1276. [Google Scholar] [CrossRef]
  70. Von, H.L. Role of persistent infection in the control and severity of asthma: Focus on Chlamydia pneumoniae. Eur. Respir. J. 2002, 19, 546–556. [Google Scholar]
  71. Oliver, B.G.; Lim, S.; Wark, P.; Laza-Stanca, V.; King, N.; Black, J.L.; Burgess, J.K.; Roth, M.; Johnston, S.L. Rhinovirus exposure impairs immune responses to bacterial products in human alveolar macrophages. Thorax 2008, 63, 519–525. [Google Scholar] [CrossRef]
  72. Message, S.D.; Laza-Stanca, V.; Mallia, P.; Parker, H.L.; Zhu, J.; Kebadze, T.; Contoli, M.; Sanderson, G.; Kon, O.M.; Papi, A.; et al. Rhinovirus-induced lower respiratory illness is increased in asthma and related to virus load and Th1/2 cytokine and IL-10 production. Proc. Natl. Acad. Sci. USA 2008, 105, 13562–13567. [Google Scholar] [CrossRef]
  73. Eisner, M.D.; Klein, J.; Hammond, S.K.; Koren, G.; Lactao, G.; Iribarren, C. Directly measured second hand smoke exposure and asthma health outcomes. Thorax 2005, 60, 814–821. [Google Scholar] [CrossRef]
  74. Hansel, N.N.; Breysse, P.N.; McCormack, M.C.; Matsui, E.C.; Curtin-Brosnan, J.; Williams, D.L.; Moore, J.L.; Cuhran, J.L.; Diette, G.B. A longitudinal study of indoor nitrogen dioxide levels and respiratory symptoms in inner-city children with asthma. Environ. Health Perspect. 2008, 116, 1428–1432. [Google Scholar] [CrossRef] [PubMed]
  75. Petsky, H.L.; Cates, C.J.; Lasserson, T.J.; Li, A.M.; Turner, C.; Kynaston, J.A.; Chang, A.B. A systematic review and meta-analysis: Tailoring asthma treatment on eosinophilic markers (exhaled nitric oxide or sputum eosinophils). Thorax 2012, 67, 199–208. [Google Scholar] [CrossRef] [PubMed]
  76. Little, S.A.; MacLeod, K.J.; Chalmers, G.W.; Love, J.G.; McSharry, C.; Thomson, N.C. Association of forced expiratory volume with disease duration and sputum neutrophils in chronic asthma. Am. J. Med. 2002, 112, 446–452. [Google Scholar] [CrossRef] [PubMed]
  77. Gans, M.D.; Gavrilova, T. Understanding the immunology of asthma: Pathophysiology, biomarkers, and treatments for asthma endotypes. Paediatr. Respir. Rev. 2020, 36, 118–127. [Google Scholar] [CrossRef]
  78. Agache, I.; Eguiluz-Gracia, I.; Cojanu, C.; Laculiceanu, A.; del Giacco, S.; Zemelka-Wiacek, M.; Kosowska, A.; Akdis, C.A.; Jutel, M. Advances and highlights in asthma in 2021. Allergy 2021, 76, 3390–3407. [Google Scholar] [CrossRef]
  79. Schleich, F.; Bougard, N.; Moermans, C.; Sabbe, M.; Louis, R. Cytokine-targeted therapies for asthma and COPD. Eur. Respir. Rev. 2023, 32, 220193. [Google Scholar] [CrossRef]
  80. Dweik, R.A.; Boggs, P.B.; Erzurum, S.C.; Irvin, C.G.; Leigh, M.W.; Lundberg, J.O.; Olin, A.-C.; Plummer, A.L.; Taylor, D.R. An official ATS clinical practice guideline: Interpretation of exhaled nitric oxide levels (FENO) for clinical applications. Am. J. Respir. Crit. Care Med. 2011, 184, 602–615. [Google Scholar] [CrossRef]
  81. Peters-Golden, M.; Henderson, W.R., Jr. Leukotrienes. N. Engl. J. Med. 2007, 357, 1841–1854. [Google Scholar] [CrossRef] [PubMed]
  82. Pavord, I.D.; Korn, S.; Howarth, P.; Bleecker, E.R.; Buhl, R.; Keene, O.N.; Ortega, H.; Chanez, P. Mepolizumab for severe eosinophilic asthma (DREAM): A multicentre, double-blind, placebo-controlled trial. Lancet 2012, 380, 651–659. [Google Scholar] [CrossRef] [PubMed]
  83. McDowell, P.J.; Diver, S.; Yang, F.; Borg, C.; Busby, J.; Brown, V.; Shrimanker, R.; Cox, C.; Brightling, C.E.; Chaudhuri, R.; et al. The inflammatory profile of exacerbations in patients with severe refractory eosinophilic asthma receiving mepolizumab (the MEX study): A prospective observational study. Lancet Respir. Med. 2021, 9, 1174–1184. [Google Scholar] [CrossRef] [PubMed]
  84. Bafadhel, M.; Haldar, K.; Barker, B.; Patel, H.; Mistry, V.; Barer, M.R.; Pavord, I.D.; Brightling, C. AAirway bacteria measured by quantitative polymerase chain reaction and culture in patients with stable COPD: Relationship with neutrophilic airway inflammation, exacerbation frequency, and lung function. Int. J. Chronic Obstr. Pulm. Dis. 2015, 10, 1075–1083. [Google Scholar] [CrossRef]
  85. Poznanski, S.M.; Mukherjee, M.; Zhao, N.; Huang, C.; Radford, K.; Ashkar, A.A.; Nair, P. Asthma exacerbations on benralizumab are largely non-eosinophilic. Allergy 2021, 76, 375–379. [Google Scholar] [CrossRef]
  86. Nair, P.; O’Byrne, P.M. Medical algorithms: Approach to adult asthma exacerbations. Allergy 2021, 76, 3556–3559. [Google Scholar] [CrossRef]
  87. Humbert, M.; Busse, W.; Hanania, N.A.; Lowe, P.J.; Canvin, J.; Erpenbeck, V.J.; Holgate, S. Omalizumab in asthma: An update on recent developments. J. Allergy Clin. Immunol. Pract. 2014, 2, 525–536.e1. [Google Scholar] [CrossRef]
  88. Novak, N.; Bieber, T. Allergic and nonallergic forms of atopic diseases. J. Allergy Clin. Immunol. 2003, 112, 252–262. [Google Scholar] [CrossRef]
  89. Manka, L.A.; Wechsler, M.E. Selecting the right biologic for your patients with severe asthma. Ann. Allergy Asthma Immunol. 2018, 121, 406–413. [Google Scholar] [CrossRef]
  90. Kavanagh, J.E.; Hearn, A.P.; Jackson, D.J. A pragmatic guide to choosing biologic therapies in severe asthma. Breathe 2022, 17, 210144. [Google Scholar] [CrossRef]
  91. Teach, S.J.; Gill, M.A.; Togias, A.; Sorkness, C.A.; Arbes, S.J., Jr.; Calatroni, A.; Wildfire, J.J.; Gergen, P.J.; Cohen, R.T.; Pongracic, J.A.; et al. Preseasonal treatment with either omalizumab or an inhaled corticosteroid boost to prevent fall asthma exacerbations. J. Allergy Clin. Immunol. 2015, 136, 1476–1485. [Google Scholar] [CrossRef] [PubMed]
  92. Normansell, R.; Walker, S.; Milan, S.J.; Walters, E.H.; Nair, P. Omalizumab for Asthma in Adults and Children. Cochrane Database Syst. Rev. 2014, 1, CD003559. [Google Scholar] [CrossRef] [PubMed]
  93. Holgate, S.T.; Chuchalin, A.G.; Hebert, J.; Lötvall, J.; Persson, G.B.; Chung, K.F.; Bousquet, J.; Kerstjens, H.A.; Fox, H.; Thirlwell, J.; et al. Omalizumab 011 International Study Group. Efficacy and safety of a recombinant anti-immunoglobulin E antibody (omalizumab) in severe allergic asthma. Clin. Exp. Allergy 2004, 34, 632–638. [Google Scholar] [CrossRef] [PubMed]
  94. Soler, M.; Matz, J.; Townley, R.; Buhl, R.; O’brien, J.; Fox, H.; Thirlwell, J.; Gupta, N.; Della Cioppa, G.A. The anti-IgE antibody omalizumab reduces exacerbations and steroid requirement in allergic asthmatics. Eur. Respir. J. 2001, 18, 254–261. [Google Scholar] [CrossRef]
  95. Busse, W.; Corren, J.; Lanier, B.Q.; McAlary, M.; Fowler-Taylor, A.; Della Cioppa, G.; van As, A.; Gupta, N. Omalizumab, anti-IgE recombinant humanized monoclonal antibody, for the treatment of severe allergic asthma. J. Allergy Clin. Immunol. 2001, 108, 184–190. [Google Scholar] [CrossRef]
  96. Busse, W.W.; Morgan, W.J.; Gergen, P.J.; Mitchell, H.E.; Gern, J.E.; Liu, A.H.; Gruchalla, R.S.; Kattan, M.; Teach, S.J.; Pongracic, J.A.; et al. Randomized trial of omalizumab (anti-IgE) for asthma in inner-city children. N. Engl. J. Med. 2011, 364, 1005–1015. [Google Scholar] [CrossRef]
  97. Menzella, F.; Piro, R.; Facciolongo, N.; Castagnetti, C.; Simonazzi, A.; Zucchi, L. Long-term benefits of omalizumab in a patient with severe non-allergic asthma. Allergy Asthma Clin. Immunol. 2011, 7, 9. [Google Scholar] [CrossRef]
  98. Bousquet, J.; Humbert, M.; Gibson, P.G.; Kostikas, K.; Jaumont, X.; Pfister, P.; Nissen, F. Real-World Effectiveness of Omalizumab in Severe Allergic Asthma: A Meta-Analysis of Observational Studies. J Allergy Clin Immunol Pract. 2021, 9, 2702–2714. [Google Scholar] [CrossRef]
  99. Niven, R.M.; Saralaya, D.; Chaudhuri, R.; Masoli, M.; Clifton, I.; Mansur, A.H.; Hacking, V.; McLain-Smith, S.; Menzies-Gow, A. Impact of omalizumab on treatment of severe allergic asthma in UK clinical practice: A UK multicentre observational study (the APEX II study). BMJ Open 2016, 6, e011857. [Google Scholar] [CrossRef]
  100. Casale, T.B.; Luskin, A.T.; Busse, W.; Zeiger, R.S.; Trzaskoma, B.; Yang, M.; Griffin, N.M.; Chipps, B.E. Omalizumab effectiveness by biomarker status in patients with asthma: Evidence from PROSPERO, a prospective real-world study. J. Allergy Clin. Immunol. Pract. 2019, 7, 156–164.e1. [Google Scholar] [CrossRef]
  101. Hanania, N.A.; Wenzel, S.; Rosén, K.; Hsieh, H.J.; Mosesova, S.; Choy, D.F.; Lal, P.; Arron, J.R.; Harris, J.M.; Busse, W. Exploring the effects of omalizumab in allergic asthma: An analysis of biomarkers in the EXTRA study. Am. J. Respir. Crit. Care Med. 2013, 187, 804–811. [Google Scholar] [CrossRef] [PubMed]
  102. Garcia, G.; Magnan, A.; Chiron, R.; Contin-Bordes, C.; Berger, P.; Taillé, C.; Devouassoux, G.; de Blay, F.; Couderc, L.-J.; Didier, A.; et al. A proof-of-concept, randomized, controlled trial of omalizumab in patients with severe, difficult-to-control, nonatopic asthma. Chest 2013, 144, 411–419. [Google Scholar] [CrossRef]
  103. Ledford, D.; Busse, W.; Trzaskoma, B.; Omachi, T.A.; Rosén, K.; Chipps, B.E.; Luskin, A.T.; Solari, P.G. A randomized multicenter study evaluating Xolair persistence of response after long-term therapy. J. Allergy Clin. Immunol. 2017, 140, 162–169.e2. [Google Scholar] [CrossRef] [PubMed]
  104. Corren, J.; Casale, T.B.; Lanier, B.; Buhl, R.; Holgate, S.; Jimenez, P. Safety and tolerability of omalizumab. Clin Exp Allergy 2009, 39, 788–797. [Google Scholar] [CrossRef] [PubMed]
  105. Kardas, G.; Kuna, P.; Panek, M. Biological Therapies of Severe Asthma and Their Possible Effects on Airway Remodeling. Front. Immunol. 2020, 11, 1134. [Google Scholar] [CrossRef]
  106. McCracken, J.L.; Tripple, J.W.; Calhoun, W.J. Biologic Therapy in the Management of Asthma. Curr. Opin. Allergy Clin. Immunol. 2016, 16, 375–382. [Google Scholar] [CrossRef]
  107. Humbert, M.; Beasley, R.; Ayres, J.; Slavin, R.; Hébert, J.; Bousquet, J.; Beeh, K.M.; Ramos, S.; Canonica, G.W.; Hedgecock, S.; et al. Benefits of Omalizumab as Add-on Therapy in Patients with Severe Persistent Asthma Who Are Inadequately Controlled despite Best Available Therapy (Gina 2002 Step 4 Treatment): Innovate. Allergy 2005, 60, 309–316. [Google Scholar] [CrossRef]
  108. Fowler, S.J.; Tavernier, G.; Niven, R. High blood eosinophil counts predict sputum eosinophilia in patients with severe asthma. J. Allergy Clin. Immunol. 2015, 135, 822–824.e2. [Google Scholar] [CrossRef]
  109. Mukherjee, M.; Nair, P. Blood or sputum eosinophils to guide asthma therapy? Lancet Respir. Med. 2015, 3, 824–825. [Google Scholar] [CrossRef]
  110. Bhalla, A.; Mukherjee, M.; Nair, P. Airway eosinophilopoietic and autoimmune mechanisms of eosinophilia in severe asthma. Immunol. Allergy Clin. N. Am. 2018, 38, 639–654. [Google Scholar] [CrossRef]
  111. Sehmi, R.; Smith, S.G.; Kjarsgaard, M.; Radford, K.; Boulet, L.; Lemiere, C.; Prazma, C.M.; Ortega, H.; Martin, J.G.; Nair, P. Role of local eosinophilopoietic processes in the development of airway eosinophilia in prednisone-dependent severe asthma. Clin. Exp. Allergy 2016, 46, 793–802. [Google Scholar] [CrossRef] [PubMed]
  112. Menzies-Gow, A.; Flood-Page, P.; Sehmi, R.; Burman, J.; Hamid, Q.; Robinson, D.S.; Kay, A.; Denburg, J. Anti-IL-5 (mepolizumab) therapy induces bone marrow eosinophil maturational arrest and decreases eosinophil progenitors in the bronchial mucosa of atopic asthmatics. J. Allergy Clin. Immunol. 2003, 111, 714–719. [Google Scholar] [CrossRef] [PubMed]
  113. Mukherjee, M.; Paramo, F.A.; Kjarsgaard, M.; Salter, B.; Nair, G.; LaVigne, N.; Radford, K.; Sehmi, R.; Nair, P. Weight-adjusted intravenous reslizumab in severe asthma with inadequate response to fixed-dose subcutaneous mepolizumab. Am. J. Respir. Crit. Care Med. 2018, 197, 38–46. [Google Scholar] [CrossRef] [PubMed]
  114. Mukherjee, M.; Lim, H.F.; Thomas, S.; Miller, D.; Kjarsgaard, M.; Tan, B.; Sehmi, R.; Khalidi, N.; Nair, P. Airway autoimmune responses in severe eosinophilic asthma following low-dose mepolizumab therapy. Allergy Asthma Clin. Immunol. 2017, 13, 2. [Google Scholar] [CrossRef]
  115. Nair, P.; Pizzichini, M.M.; Kjarsgaard, M.; Inman, M.D.; Efthimiadis, A.; Pizzichini, E.; Hargreave, F.E.; O’BYrne, P.M. Mepolizumab for prednisone-dependent asthma with sputum eosinophilia. N. Engl. J. Med. 2009, 360, 985–993. [Google Scholar] [CrossRef]
  116. Bel, E.H.; Wenzel, S.E.; Thompson, P.J.; Prazma, C.M.; Keene, O.N.; Yancey, S.W.; Ortega, H.G.; Pavord, I.D.; SIRIUS Investigators. Oral glucocorticoid-sparing effect of mepolizumab in eosinophilic asthma. N. Engl. J. Med. 2014, 371, 1189–1197. [Google Scholar] [CrossRef]
  117. Pavord, I.D.; Menzies-Gow, A.; Buhl, R.; Chanez, P.; Dransfield, M.; Lugogo, N.; Keene, O.N.; Bradford, E.S.; Yancey, S.W. Clinical Development of Mepolizumab for the Treatment of Severe Eosinophilic Asthma: On the Path to Personalized Medicine. J. Allergy Clin. Immunol. Pract. 2021, 9, 1121–1132.e7. [Google Scholar] [CrossRef]
  118. Máspero, J. Reslizumab in the Treatment of Inadequately Controlled Asthma in Adults and Adolescents with Elevated Blood Eosinophils: Clinical Trial Evidence and Future Prospects. Ther. Adv. Respir. Dis. 2017, 11, 311–325. [Google Scholar] [CrossRef]
  119. Pilette, C.; Canonica, G.W.; Chaudhuri, R.; Chupp, G.; Lee, F.E.; Lee, J.K.; Almonacid, C.; Welte, T.; Alfonso-Cristancho, R.; Jakes, R.W.; et al. REALITI-A Study: Real-World Oral Corticosteroid-Sparing Effect of Mepolizumab in Severe Asthma. J. Allergy Clin. Immunol. Pract. 2022, 10, 2646–2656. [Google Scholar] [CrossRef]
  120. Mihaltan, F.; Csoma, Z.; Pauk, N.; Irás, B.; Baukienė, J.; Teodorescu, G.; Malakauskas, K.; Staevska, M. Benralizumab Outcomes in Patients with Severe Eosinophilic Asthma Treated in Real-Life Settings: Results of the BREEZE Study in 5 Countries from Central Eastern Europe and Baltics. J. Asthma Allergy 2025, 18, 195–210. [Google Scholar] [CrossRef]
  121. Carstens, D.D.; Genofre, E.H.; Young, J.A.; Yang, D.; Mu, F.; Cook, E.E.; Betts, K.A.; Chung, Y. Long-Term Real-World Effectiveness of Benralizumab in Patients with Severe Eosinophilic Asthma: Results from the Zephyr 2 Study. Chest 2022, 162, A7. [Google Scholar] [CrossRef]
  122. Wechsler, M.E.; Faverio, P.; Sussman, G.; Lang, D.M.; Heaney, L.G.; Kolbeck, R.; Yancey, S.W.; Lee, D.; Le, M.; Kwon, N.; et al. Mepolizumab or Benralizumab for Eosinophilic Granulomatosis with Polyangiitis. N. Engl. J. Med. 2024, 390, 911–921. [Google Scholar] [CrossRef] [PubMed]
  123. Rosenberg, H.F.; Dyer, K.D.; Foster, P.S. Eosinophils: Changing perspectives in health and disease. Nat. Rev. Immunol. 2013, 13, 9–22. [Google Scholar] [CrossRef] [PubMed]
  124. Ortega, H.G.; Liu, M.C.; Pavord, I.D.; Brusselle, G.G.; Fitzgerald, J.M.; Chetta, A.; Humbert, M.; Katz, L.E.; Keene, O.N.; Yancey, S.W.; et al. Mepolizumab treatment in patients with severe eosinophilic asthma. N. Engl. J. Med. 2014, 371, 1198–1207. [Google Scholar] [CrossRef]
  125. Casale, T.B.; Burnette, A.; Bourdin, A.; Howarth, P.; Hahn, B.; Stach-Klysh, A.; Khurana, S. Oral corticosteroid-sparing effects of mepolizumab in severe eosinophilic asthma: Evidence from randomized controlled trials and real-world studies. Ther. Adv. Respir. Dis. 2022, 16, 17534666221107313. [Google Scholar] [CrossRef]
  126. Chupp, G.L.; Bradford, E.S.; Albers, F.C.; Bratton, D.J.; Wang-Jairaj, J.; Nelsen, L.M.; Trevor, J.L.; Magnan, A.; Brinke, A.T. Efficacy of mepolizumab add-on therapy on health-related quality of life and markers of asthma control in severe eosinophilic asthma (MUSCA): A randomised, double-blind, placebo-controlled, parallel-group, multicentre, phase 3b trial. Lancet Respir. Med. 2017, 5, 390–400. [Google Scholar] [CrossRef]
  127. Principe, S.; Porsbjerg, C.; Bolm Ditlev, S.; Kjaersgaard Klein, D.; Golebski, K.; Dyhre-Petersen, N.; van Dijk, Y.E.; van Bragt, J.J.M.H.; Dankelman, L.L.H.; Dahlen, S.E.; et al. Treating severe asthma: Targeting the IL-5 pathway. Clin. Exp. Allergy 2021, 51, 992–1005. [Google Scholar] [CrossRef]
  128. Pelaia, C.; Crimi, C.; Pelaia, G.; Nolasco, S.; Campisi, R.; Heffler, E.; Valenti, G.; Crimi, N. Real-life evaluation of mepolizumab efficacy in patients with severe eosinophilic asthma, according to atopic trait and allergic phenotype. Clin. Exp. Allergy 2020, 50, 780–788. [Google Scholar] [CrossRef]
  129. Carpagnano, G.E.; Pelaia, C.; D’Amato, M.; Crimi, N.; Scichilone, N.; Scioscia, G.; Resta, O.; Calabrese, C.; Pelaia, G.; Quarato, C.M.I.; et al. Switching from omalizumab to mepolizumab: Real-life experience from Southern Italy. Ther. Adv. Respir. Dis. 2020, 14, 1753466620929231. [Google Scholar] [CrossRef]
  130. Lombardi, C.; Comberiati, P.; Ridolo, E.; Cottini, M.; Yacoub, M.R.; Casagrande, S.; Riccò, M.; Bottazzoli, M.; Berti, A. Anti-IL-5 Pathway Agents in Eosinophilic-Associated Disorders Across the Lifespan. Drugs 2024, 84, 661–684. [Google Scholar] [CrossRef]
  131. Kroes, J.A.; Zielhuis, S.W.; van der Meer, A.-N.; de Jong, K.; van Roon, E.N.; ten Brinke, A. Optimizing Omalizumab Dosing in Severe Asthma—The Exploration of Therapeutic Drug Monitoring. J. Allergy Clin. Immunol. Pract. 2021, 9, 1408–1410. [Google Scholar] [CrossRef] [PubMed]
  132. Iribarren, C.; Rahmaoui, A.; Long, A.A.; Szefler, S.J.; Bradley, M.S.; Carrigan, G.; Eisner, M.D.; Chen, H.; Omachi, T.A.; Farkouh, M.E.; et al. Cardiovascular and Cerebrovascular Events among Patients Receiving Omalizumab: Results from Excels, a Prospective Cohort Study in Moderate to Severe Asthma. J. Allergy Clin. Immunol. 2017, 139, 1489–1495. [Google Scholar] [CrossRef] [PubMed]
  133. CINQAIR. Prescribing Information. Teva Respiratory. 2020. Available online: https://www.cinqair.com/globalassets/cinqair/prescribinginformation.pdf (accessed on 1 June 2025).
  134. Hom, S.; Pisano, M. Reslizumab (Cinqair): An Interleukin-5 Antagonist for Severe Asthma of the Eosinophilic Phenotype. Pharm. Ther. 2017, 42, 564–568. Available online: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5565129/ (accessed on 1 June 2025).
  135. Bjermer, L.; Lemiere, C.; Maspero, J.; Weiss, S.; Zangrilli, J.; Germinaro, M. Reslizumab for Inadequately Controlled Asthma with Elevated Blood Eosinophil Levels. Chest 2016, 150, 789–798. [Google Scholar] [CrossRef] [PubMed]
  136. CTG Labs—NCBI. A Study to Evaluate the Efficacy and Safety of Reslizumab in Patients with Eosinophilic Asthma; Teva Branded Pharmaceutical Products R&D, Inc.: West Chester, PA, USA, 2021. Available online: https://beta.clinicaltrials.gov/study/NCT01285323?tab=results#publications (accessed on 1 June 2025).
  137. Pelaia, C.; Calabrese, C.; Vatrella, A.; Busceti, M.T.; Garofalo, E.; Lombardo, N.; Terracciano, R.; Pelaia, G. Benralizumab: From the basic mechanism of action to the potential use in the biological therapy of severe eosinophilic asthma. BioMed Res. Int. 2018, 2018, 4839230. [Google Scholar] [CrossRef]
  138. Farne, H.A.; Wilson, A.; Powell, C.; Bax, L.; Milan, S.J. Anti-IL5 therapies forasthma. Cochrane Database Syst. Rev. 2017, 9, CD010834. [Google Scholar]
  139. Nanzer, A.M.; Dhariwal, J.; Thomson, L.; Green, L.; Fernandes, M.; Roxas, C.; Haris, F.; Cefaloni, F.; Gates, J.; Jackson, D.J. Benralizumab in Eosinophilic Granulomatosis with Polyangiitis: Real-World Experience. J. Allergy Clin. Immunol. Pract. 2024, 12, 724–732. [Google Scholar] [CrossRef]
  140. Hanania, N.A.; Alpan, O.; Hamilos, D.L.; Condemi, J.J.; Reyes-Rivera, I.; Zhu, J.; Rosen, K.E.; Eisner, M.D.; Wong, D.A.; Busse, W. Omalizumab in Severe Allergic Asthma Inadequately Controlled with Standard Therapy. Ann. Intern. Med. 2011, 154, 573–582. [Google Scholar] [CrossRef]
  141. CTG Labs—NCBI. A Study to Evaluate the Efficacy and Safety of Reslizumab (0.3 or 3.0 Mg/Kg) as Treatment for Patients (12–75 Years of Age) with Eosinophilic Asthma; Teva Branded Pharmaceutical Products R&D, Inc.: West Chester, PA, USA, 2016. Available online: https://beta.clinicaltrials.gov/study/NCT01270464 (accessed on 1 June 2025).
  142. Corren, J.; Weinstein, S.; Janka, L.; Zangrilli, J.; Garin, M. Phase 3 Study of Reslizumab in Patients with Poorly Controlled Asthma. Chest 2016, 150, 799–810. [Google Scholar] [CrossRef]
  143. Bleecker, E.R.; FitzGerald, J.M.; Chanez, P.; Papi, A.; Weinstein, S.F.; Barker, P.; Sproule, S.; Gilmartin, G.; Aurivillius, M.; Werkström, V.; et al. Efficacy and Safety of Benralizumab for Patients with Severe Asthma Uncontrolled with High-Dosage Inhaled Corticosteroids and Long-Acting 2-Agonists (SIROCCO): A Randomised, Multicentre, Placebo-Controlled Phase 3 Trial. Lancet 2016, 388, 2115–2127. [Google Scholar] [CrossRef]
  144. FitzGerald, J.M.; Bleecker, E.R.; Nair, P.; Korn, S.; Ohta, K.; Lommatzsch, M.; Ferguson, G.T.; Busse, W.W.; Barker, P.; Sproule, S.; et al. Benralizumab, an Anti-Interleukin-5 Receptor Monoclonal Antibody, as Add-on Treatment for Patients with Severe, Uncontrolled, Eosinophilic Asthma (Calima): A Randomised, Double-Blind, Placebo-Controlled Phase 3 Trial. Lancet 2016, 388, 2128–2141. [Google Scholar] [CrossRef] [PubMed]
  145. Gandhi, N.A.; Bennett, B.L.; Graham, N.M.H.; Pirozzi, G.; Stahl, N.; Yancopoulos, G.D. Targeting key proximal drivers of type 2 in-flammation in disease. Nat. Rev. Drug Discov. 2016, 15, 35–50. [Google Scholar] [CrossRef] [PubMed]
  146. Le Floc’h, A.; Allinne, J.; Nagashima, K.; Scott, G.; Birchard, D.; Asrat, S.; Bai, Y.; Lim, W.K.; Martin, J.; Huang, T.; et al. Dual blockade of IL-4 and IL-13 with dupilumab, an IL-4Ralpha antibody, is required to broadly inhibit type 2 inflammation. Allergy 2020, 75, 1188–1204. [Google Scholar] [CrossRef]
  147. Wechsler, M.E.; Klion, A.D.; Paggiaro, P.; Nair, P.; Staumont-Salle, D.; Radwan, A.; Johnson, R.R.; Kapoor, U.; Khokhar, F.A.; Daizadeh, N.; et al. Effect of dupilumab on blood eosinophil counts in patients with asthma, chronic rhinosinusitis with nasal polyps, atopic dermatitis, or eosinophilic esophagitis. J. Allergy Clin. Immunol. Pract. 2022, 10, 2695–2709. [Google Scholar] [CrossRef]
  148. Gates, L.E.; Park, H.; Vasu, T.; Vandeplas, G.; White, M.; Brightling, C.E.; Bel, E.H.; Bleecker, E.R.; Canonica, G.W.; Menzies-Gow, A.; et al. Long-Term Effectiveness of Anti-IL-4R Therapy Following Suboptimal Response to Anti-IL-5/5R Therapy in Severe Eosinophilic Asthma. J. Allergy Clin. Immunol. Pract. 2024, 12, 1794–1800. [Google Scholar] [CrossRef]
  149. Silverberg, J.I.; Yosipovitch, G.; Simpson, E.L.; Kim, B.S.; Wu, J.J.; Eckert, L.; Guillemin, I.; Chen, Z.; Ardeleanu, M.; Bansal, A.; et al. Dupilumab Treatment Results in Early and Sustained Improvements in Itch in Adolescents and Adults with Moderate to Severe Atopic Dermatitis: Analysis of the Randomized Phase 3 Studies SOLO 1 and SOLO 2, AD ADOL, and CHRONOS. J. Am. Acad. Dermatol. 2020, 82, 1328–1336. [Google Scholar] [CrossRef]
  150. Paller, A.S.; Siegfried, E.C.; Yosipovitch, G.; Silverberg, J.I.; Wu, J.J.; Chen, Z.; Prescilla, R.; Rossi, A.B.; Delevry, D. Rapid and Sustained Improvement in Itch in Children Aged 6–11 Years with Severe Atopic Dermatitis (AD) Treated with Dupilumab: Analysis from the LIBERTY AD PEDS Phase 3 Trial. J. Am. Acad. Dermatol. 2021, 85, AB140. [Google Scholar] [CrossRef]
  151. Busse, W.W.; Maspero, J.F.; Rabe, K.F.; Papi, A.; Wenzel, S.E.; Ford, L.B.; Pavord, I.D.; Zhang, B.; Staudinger, H.; Pirozzi, G.; et al. Liberty Asthma QUEST: Phase 3 Randomized, Double-Blind, Placebo-Controlled, Parallel-Group Study to Evaluate Dupilumab Efficacy/Safety in Patients with Uncontrolled, Moderate-to-Severe Asthma. Adv. Ther. 2018, 35, 737–748. [Google Scholar] [CrossRef] [PubMed]
  152. Bacharier, L.B.; Maspero, J.F.; Katelaris, C.H.; Fiocchi, A.G.; Gagnon, R.; de Mir, I.; Jain, N.; Sher, L.D.; Mao, X.; Liu, D.; et al. Dupilumab in Children with Uncontrolled Moderate-to-Severe Asthma. N. Engl. J. Med. 2021, 385, 2230–2240. [Google Scholar] [CrossRef]
  153. Han, J.K.; Bachert, C.; Desrosiers, M.; Laidlaw, T.M.; Hopkins, C.; Fokkens, W.J.; Paggiaro, P.; Ho Cho, S.; Olze, H.; Greos, L.S.; et al. Efficacy and Safety of Dupilumab in Patients with Chronic Rhinosinusitis with Nasal Polyps: Results from the Randomized Phase 3 SINUS-24 Study. J. Allergy Clin. Immunol. 2019, 143, AB422. [Google Scholar] [CrossRef]
  154. Spergel, J.; Sher, L.; Durrani, S.; Dellon, E.; Rothenberg, M.; Sun, X.; Beazley, B.; Patel, K.; Shabbir, A. Dupilumab Improves Type 2 Comorbidity Outcomes in Patients with Eosinophilic Esophagitis and Comorbid Disease at Baseline: From Parts A and B of LIBERTY-EoE-TREET. J. Allergy Clin. Immunol. 2023, 151, AB199. [Google Scholar] [CrossRef]
  155. Chehade, M.; Dellon, E.S.; Spergel, J.M.; Collins, M.H.; Rothenberg, M.E.; Pesek, R.D.; Hirano, I.; Liu, R.; Laws, E.; Mortensen, E.; et al. Dupilumab for Eosinophilic Esophagitis in Patients 1 to 11 Years of Age. N. Engl. J. Med. 2024, 390, 2239–2251. [Google Scholar] [CrossRef] [PubMed]
  156. Verstraete, K.; Peelman, F.; Braun, H.; Lopez, J.; Van Rompaey, D.; Dansercoer, A.; Vandenberghe, I.; Pauwels, K.; Tavernier, J.; Lambrecht, B.N.; et al. Structure and antagonism of the receptor complex mediated by human TSLP in allergy and asthma. Nat. Commun. 2017, 8, 14937. [Google Scholar] [CrossRef] [PubMed]
  157. Wong, C.K.; Hu, S.; Cheung, P.F.; Lam, C.W. Thymic stromal lymphopoietin induces chemotactic and prosurvival effects in eosinophils: Implications in allergic inflammation. Am. J. Respir. Cell Mol. Biol. 2010, 43, 305–315. [Google Scholar] [CrossRef]
  158. Cook, E.B.; Stahl, J.L.; Schwantes, E.A.; Fox, K.E.; Mathur, S.K. IL-3 and TNFa increase thymic stromal lymphopoietin receptor (TSLPR) expression on eosinophils and enhance TSLP-stimulated degranulation. Clin. Mol. Allergy 2012, 10, 8. [Google Scholar] [CrossRef]
  159. Pelaia, C.; Pelaia, G.; Crimi, C.; Maglio, A.; Gallelli, L.; Terracciano, R.; Vatrella, A. Tezepelumab: A potential new biological therapy for severe refractory asthma. Int. J. Mol. Sci. 2021, 22, 4369. [Google Scholar] [CrossRef]
  160. Gates, J.; Haris, F.; Cefaloni, F.; Khooshemehri, P.; Green, L.; Fernandes, M.; Thomson, L.; Roxas, C.; Lam, J.; d’Ancona, G.; et al. Clinical and Biological Remission With Tezepelumab: The Real-World Response in Severe Uncontrolled Asthma. Allergy 2025, 80, 1669–1676. [Google Scholar] [CrossRef]
  161. Available online: https://www.fdanews.com/articles/188355-fda-awards-astrazeneca-and-amgens-tezepelumab-breakthrough-designation (accessed on 2 June 2025).
  162. Lipworth, B.J.; Caruso, M.; Corren, J.; Klimek, L.; Canonica, G.W.; Han, J.K.; Naclerio, R.M.; Hellings, P.W.; Bachert, C. Tezepelumab in Patients with Severe Chronic Rhinosinusitis with Nasal Polyps. N. Engl. J. Med. 2025, 392, 1178–1188. [Google Scholar] [CrossRef]
  163. Gelhorn, H.L.; Balantac, Z.; Ambrose, C.S.; Chung, Y.N.; Stone, B. Patient and physician preferences for attributes of biologic medications for severe asthma. Patient Prefer. Adherence 2019, 13, 1253–1268. [Google Scholar] [CrossRef]
  164. Jackson, D.J.; Wechsler, M.E.; Jackson, D.J.; Bernstein, D.; Korn, S.; Pfeffer, P.E.; Chen, R.; Saito, J.; de Luíz Martinez, G.; Dymek, L.; et al. SWIFT 1 and SWIFT-2 Investigators; SWIFT-1 Investigators; SWIFT-2 Investigators. Twice-yearly depemokimab in severe asthma with an eosinophilic phenotype. N. Engl. J. Med. 2024, 391, 2337–2349. [Google Scholar] [CrossRef]
  165. Antalfy, A.; Berman, K.; Everitt, C.; Alten, R.; Latymer, M.; Godfrey, C.M. The adherence and outcomes ben efits of using a connected, reusable auto-injector for self-injecting biologics: A narrative review. Adv. Ther. 2023, 40, 4758–4776. [Google Scholar] [CrossRef] [PubMed]
  166. van den Bemt, B.J.F.; Gettings, L.; Domańska, B.; Bruggraber, R.; Mountian, I.; Kristensen, L.E. A portfolio of biologic self-injection devices in rheumatology: How patient involvement in device design can improve treatment experience. Drug Deliv. 2019, 26, 384–392. [Google Scholar] [CrossRef]
  167. Stauffer, V.L.; Sides, R.; Lanteri-Minet, M.; Kielbasa, W.; Jin, Y.; Selzler, K.J.; Tepper, S.J. Com parison between prefilled syringe and autoinjector devices on patient-reported experiences and pharma cokinetics in galcanezumab studies. Patient Prefer. Adherence 2018, 12, 1785–1795. [Google Scholar] [CrossRef] [PubMed]
  168. Vermeire, S.; D’HEygere, F.; Nakad, A.; Franchimont, D.; Fontaine, F.; Louis, E.; Van Hootegem, P.; Dewit, O.; Lambrecht, G.; Strubbe, B.; et al. Preference for a prefilled syringe or an auto-injection device for de livering golimumab in patients with moderate-to-severe ulcerative colitis: A randomized crossover study. Patient Prefer. Adherence 2018, 12, 1193–1202. [Google Scholar] [CrossRef]
  169. Demary, W.; Schwenke, H.; Rockwitz, K.; Kästner, P.; Liebhaber, A.; Schoo, U.; Hübner, G.; Pichlmeier, U.; Guimbal-Schmolck, C.; Müller-Ladner, U. Subcu taneously administered methotrexate for rheumatoid arthritis, by prefilled syringes versus prefilled pens: Patient preference and comparison of the self-injection experience. Patient Prefer. Adherence 2014, 8, 1061–1071. [Google Scholar]
  170. Shabbir, S.; Pouliquen, I.J.; Bentley, J.H.; Bradford, E.S.; Kaisermann, M.C.; Albayaty, M. The pharmacokinetics and relative bioavailability of mepolizumab 100 mg liquid formu lation administered subcutaneously to healthy partici pants: A randomized trial. Clin. Pharmacol. Drug Dev. 2020, 9, 375–385. [Google Scholar] [CrossRef]
  171. Martin, U.J.; Fuhr, R.; Forte, P.; Barker, P.; Axley, M.J.; Aurivillius, M.; Yan, L.; Roskos, L. Comparison of au toinjector with accessorized prefilled syringe for benral izumab pharmacokinetic exposure: AMES trial results. J Asthma 2021, 58, 93–101. [Google Scholar] [CrossRef] [PubMed]
  172. Martin, N.T.; Martin, M.U. Interleukin 33 is a guardian of barriers and a local alarmin. Nat. Immunol. 2016, 17, 122–131. [Google Scholar] [CrossRef]
  173. Cayrol, C.; Girard, J.P. Interleukin-33 (IL-33): A nuclear cytokine from the IL-1 family. Immunol. Rev. 2018, 281, 154–168. [Google Scholar] [CrossRef]
  174. Hong, H.; Liao, S.; Chen, F.; Yang, Q.; Wang, D.Y. Role of IL-25, IL-33, and TSLP in triggering united airway diseases toward type2 inflammation. Allergy 2020, 75, 2794–2804. [Google Scholar] [CrossRef]
  175. Macdonald, L.E.; Karow, M.; Stevens, S.; Auerbach, W.; Poueymirou, W.T.; Yasenchak, J.; Frendewey, D.; Valenzuela, D.M.; Giallourakis, C.C.; Alt, F.W.; et al. Precise and in situ genetic humanization of 6 Mb of mouse immunoglobulin genes. Proc. Natl. Acad. Sci. USA 2014, 111, 5147–5152. [Google Scholar] [CrossRef] [PubMed]
  176. Murphy, A.J.; Macdonald, L.E.; Stevens, S.; Karow, M.; Dore, A.T.; Pobursky, K.; Huang, T.T.; Poueymirou, W.T.; Esau, L.; Meola, M.; et al. Mice with megabase humanization of their immunoglobulin genes generate antibodies as efficiently as normal mice. Proc. Natl. Acad. Sci. USA 2014, 111, 5153–5158. [Google Scholar] [CrossRef] [PubMed]
  177. Yang, X.; Ambrogelly, A. Enlarging the repertoire of therapeutic monoclonal antibodies platforms: Domesticating half molecule exchange to produce stable IgG4 and IgG1 bispecific antibodies. Curr. Opin. Biotechnol. 2014, 30, 225–229. [Google Scholar] [CrossRef]
  178. Allinne, J.; Scott, G.; Lim, W.K.; Birchard, D.; Erjefält, J.S.; Sandén, C.; Ben, L.-H.; Agrawal, A.; Kaur, N.; Kim, J.H.; et al. IL-33 blockade affects mediators of persistence and exacerbation in a model of chronic airway inflammation. J. Allergy Clin. Immunol. 2019, 144, 1624–1637.e10. [Google Scholar] [CrossRef]
  179. Global Initiative for Asthma (GINA). Global Strategy for Asthma Management and Prevention. GINA. 2015. Available online: https://ginasthma.org/wp-content/uploads/2016/01/GINA_Report_2015_Aug11-1.pdf (accessed on 1 June 2025).
  180. .Kim, S.W.; Rhee, C.K.; Kim, K.U.; Lee, S.H.; Hwang, H.G.; Kim, Y.I.; Kim, D.K.; Oh, Y.M.; Yoon, H.K. Factors associated with plasma IL-33 levels in patients with chronic obstructive pulmonary disease. Int. J. Chronic Obstr. Pulmon. Dis. 2017, 12, 395–402. [Google Scholar] [CrossRef]
  181. Zhang, Y.; Yan, B.; Shen, S.; Song, X.; Jiang, Y.; Shi, L.; Zhao, C.; Yang, Y.; Jiang, L.; Li, J.; et al. Efficacy and safety of CM310 in severe eosinophilic chronic rhinosinusitis with nasal polyps (CROWNS-1): A multicentre, randomised, double-blind, placebo-controlled phase 2 clinical trial. eClinicalMedicine 2023, 61, 102076. [Google Scholar] [CrossRef] [PubMed]
  182. Zhao, Y.; Zhang, L.; Zhang, J.; CM310AD005 Study Investigators. Efficacy and safety of stapokibart (CM310) in adults with moderate-to-severe atopic dermatitis: A multicenter, randomized, double-blind, placebo-controlled phase 3 trial. J. Am. Acad. Dermatol. 2024, 190, 2498. [Google Scholar] [CrossRef] [PubMed]
  183. Bachert, C.; Han, J.K.; Desrosiers, M.; Hellings, P.W.; Amin, N.; Lee, S.E.; Mullol, J.; Greos, L.S.; Bosso, J.V.; Laidlaw, T.M.; et al. Efficacy and safety of dupilumab in patients with severe chronic rhinosinusitis with nasal polyps (LIBERTY NP SINUS-24 and LIBERTY NP SINUS-52): Results from two multicentre, randomised, double-blind,placebo-controlled, parallel-group phase 3 trials. Lancet 2019, 394, 1638–1650. [Google Scholar] [CrossRef]
  184. Zhao, Y.; Wu, L.; Lu, Q.; Gao, X.; Zhu, X.; Yao, X.; Li, L.; Li, W.; Ding, Y.; Song, Z.; et al. The efficacy and safety of dupilumab in Chinese patients with moderate-to-severe atopic dermatitis: A randomized, double-blind, placebo-controlled study. Br. J. Dermatol. 2022, 186, 633–641. [Google Scholar] [CrossRef]
  185. Piper, E.; Brightling, C.; Niven, R.; Oh, C.; Faggioni, R.; Poon, K.; She, D.; Kell, C.; May, R.D.; Geba, G.P.; et al. A Phase II placebo-controlled study of tralokinumab in moderate-to-severe asthma. Eur. Respir. J. 2013, 41, 330–338. [Google Scholar] [CrossRef]
  186. Brightling, C.E.; Chanez, P.; Leigh, R.; O’BYrne, P.M.; Korn, S.; She, D.; May, R.D.; Streicher, K.; Ranade, K.; Piper, E. Efficacy and safety of tralokinumab in patients with severe uncontrolled asthma: A randomised, double-blind, placebo-controlled, Phase 2b trial. Lancet Respir. Med. 2015, 3, 692–701. [Google Scholar] [CrossRef] [PubMed]
  187. Baverel, P.G.; White, N.; Vicini, P.; Karlsson, M.O.; Agoram, B. Dose-exposure–response relationship of the investigational anti-IL-13 monoclonal antibody tralokinumab in patients with severe, uncontrolled asthma. Clin. Pharmacol. Ther. 2017, 103, 826–835. [Google Scholar] [CrossRef] [PubMed]
  188. Corren, J.; Szefler, S.J.; Sher, E.; Korenblat, P.; Soong, W.; Hanania, N.A.; Berman, G.; Brusselle, G.; Zitnik, R.; Natalie, C.R.; et al. Lebrikizumab in Uncontrolled Asthma: Reanalysis in a Well-Defined Type 2 Population. J. Allergy Clin. Immunol. Pract. 2024, 12, 1215–1224.e3. [Google Scholar] [CrossRef]
  189. Wenzel, S.; Castro, M.; Corren, J.; Maspero, J.; Wang, L.; Zhang, B.; Pirozzi, G.; Sutherland, E.R.; Evans, R.R.; Joish, V.N.; et al. Dupilumab efficacy and safety in adults with uncontrolled persistent asthma despite use of medium-to-high-dose inhaled corticosteroids plus a long-acting β2 agonist: A randomised double-blind placebo controlled pivotal Phase 2b dose-ranging trial. Lancet 2016, 388, 31–44. [Google Scholar] [CrossRef]
  190. Baverel, P.; She, D.; Piper, E.; Ueda, S.; Yoshioka, T.; Faggioni, R.; Gevorkyan, H. A Randomized, Placebo-Controlled, Single Ascending-Dose Study to Assess the Safety, Tolerability, Pharmacokinetics, and Immunogenicity of Subcutaneous Tralokinumab in Japanese Healthy Volunteers. Drug Metab. Pharmacokinet. 2018, 33, 150–158. [Google Scholar] [CrossRef] [PubMed]
  191. Panettieri, R.A., Jr.; Wang, M.; Braddock, M.; Bowen, K.; Colice, G. Tralokinumab for the Treatment of Severe, Uncontrolled Asthma: The ATMOSPHERE Clinical Development Program. Immunotherapy 2018, 10, 473–490. [Google Scholar] [CrossRef]
  192. Panettieri, R.A., Jr.; Sjöbring, U.; Péterffy, A.; Wessman, P.; Bowen, K.; Piper, E.; Colice, G.; Brightling, C.E. Tralokinumab for Severe, Uncontrolled Asthma (STRATOS 1 and STRATOS 2): Two Randomised, Double-Blind, Placebo-Controlled, Phase 3 Clinical Trials. Lancet Respir. Med. 2018, 6, 511–525. [Google Scholar] [CrossRef]
  193. Corren, J.; Lemanske, R.F., Jr.; Hanania, N.A.; Korenblat, P.E.; Parsey, M.V.; Arron, J.R.; Harris, J.M.; Scheerens, H.; Wu, L.C.; Su, Z.; et al. Lebrikizumab treatment in adults with asthma. N. Engl. J. Med. 2011, 365, 1088–1098. [Google Scholar] [CrossRef]
  194. Ultsch, M.; Bevers, J.; Nakamura, G.; Vandlen, R.; Kelley, R.F.; Wu, L.C.; Eigenbrot, C. Structural basis of signaling blockade by anti-IL-13 antibody lebrikizumab. J. Mol. Biol. 2013, 425, 1330–1339. [Google Scholar] [CrossRef]
  195. Scheerens, H.; Arron, J.R.; Zheng, Y.; Putnam, W.S.; Erickson, R.W.; Choy, D.F.; Harris, J.M.; Lee, J.; Jarjour, N.N.; Matthews, J.G. The effects of lebrikizumab in patients with mild asthma following whole lungallergen challenge. Clin. Exp. Allergy 2014, 44, 38–46. [Google Scholar] [CrossRef]
  196. Hanania, N.A.; Korenblat, P.; Chapman, K.R.; Bateman, E.D.; Kopecky, P.; Paggiaro, P.; Yokoyama, A.; Olsson, J.; Gray, S.; Holweg, C.T.J.; et al. Efficacy and safety of lebrikizumab in patients with uncontrolled asthma(LAVOLTA I and LAVOLTA II): Replicate, phase 3, randomised, double-blind, placebo-controlled trials. Lancet Respir. Med. 2016, 4, 781–796. [Google Scholar] [CrossRef] [PubMed]
  197. Szefler, S.; Roberts, G.; Rubin, A.S.; Zielen, S.; Kuna, P.; Alpan, O.; Anzures-Cabrera, J.; Chen, Q.; Holweg, C.; Kaminski, J.; et al. Efficacy, safety, and tolerability of lebrikizumab in adolescent patients with uncontrolled asthma (ACOUSTICS). Clin. Transl. Allergy 2022, 12, e12176. [Google Scholar] [CrossRef] [PubMed]
  198. Harrison, T.; Canonica, G.W.; Chupp, G.; Lee, J.; Schleich, F.; Welte, T.; Valero, A.; Gemzoe, K.; Maxwell, A.; Joksaite, S.; et al. Real-world mepolizumab in the prospective severe asthma REALITI—A study: Initial analysis. Eur. Respir. J. 2020, 56, 2000151. [Google Scholar] [CrossRef] [PubMed]
  199. Gyawali, B.; Georas, S.N.; Khurana, S. Biologics in severe asthma: A state-of-the-art review. Eur. Respir. Rev. 2025, 34, 240088. [Google Scholar] [CrossRef]
Figure 1. Immune cell differentiation and IgE-mediated responses in allergic asthma pathogenesis; Treg—regulatory T cells; Th1 cells—Type 1 helper T cells; IL-4—interleukin 4; IL-5—interleukin 5; IL-6—interleukin 6; IL-13—interleukin 13; IFN-γ —interferon gamma; GM-CSF—granulocyte-macrophage colony-stimulating factor. https://app.biorender.com/illustrations/6846a3f3ff2e9e10c41e8f5d (accessed on 9 June 2025).
Figure 1. Immune cell differentiation and IgE-mediated responses in allergic asthma pathogenesis; Treg—regulatory T cells; Th1 cells—Type 1 helper T cells; IL-4—interleukin 4; IL-5—interleukin 5; IL-6—interleukin 6; IL-13—interleukin 13; IFN-γ —interferon gamma; GM-CSF—granulocyte-macrophage colony-stimulating factor. https://app.biorender.com/illustrations/6846a3f3ff2e9e10c41e8f5d (accessed on 9 June 2025).
Pharmaceuticals 18 01021 g001
Figure 2. Molecular targets of biologic therapies in type 2-high asthma: cytokine pathways and immune cell interactions. TSLP—thymic stromal lymphopoietin; ILC2—group 2 innate lymphoid cells; Th2 cell—T helper 2 cell; IgE—immunoglobulin E; https://app.biorender.com/illustrations/68503e3f1998ef5af7e0af7a (accessed on 9 June 2025).
Figure 2. Molecular targets of biologic therapies in type 2-high asthma: cytokine pathways and immune cell interactions. TSLP—thymic stromal lymphopoietin; ILC2—group 2 innate lymphoid cells; Th2 cell—T helper 2 cell; IgE—immunoglobulin E; https://app.biorender.com/illustrations/68503e3f1998ef5af7e0af7a (accessed on 9 June 2025).
Pharmaceuticals 18 01021 g002
Figure 3. PRISMA flow diagram of study selection process.
Figure 3. PRISMA flow diagram of study selection process.
Pharmaceuticals 18 01021 g003
Table 1. The cytokines in asthma.
Table 1. The cytokines in asthma.
CytokinePrimary TargetsRole in Asthma PathogenesisRef.
IL-4Th2 lymphocytes, B cellsOrchestrates Th2 polarization and drives IgE class switching, fueling allergic sensitization.[47]
IL-5EosinophilsPromotes eosinophil maturation, survival and trafficking to the airways, a hallmark of eosinophilic asthma.[48]
IL-6T cells, B cells, macrophagesActs as an acute-phase mediator; elevated in severe exacerbations and linked to systemic inflammation.[49]
IL-9Mast cellsEnhances mast cell proliferation and stimulates mucus gland secretion, contributing to airway obstruction.[50]
IL-10Regulatory T cells, macrophagesDampens excessive inflammation by suppressing pro-inflammatory cytokines, helping maintain immune balance.[51]
IL-13Airway epithelial cells, goblet cellsTriggers goblet-cell hyperplasia and mucus overproduction; increases airway hyperresponsiveness and contributes to airway remodeling.[52]
IL-17NeutrophilsRecruits and activates neutrophils, driving neutrophilic inflammation seen in severe, steroid-resistant asthma.[53,54]
IL-25Dendritic cells, ILC2Potentiates innate type-2 responses by activating ILC2s and priming DCs for Th2 cytokine release.[55]
IL-33ILC2, mast cells, eosinophilsFunctions as an alarmin; amplifies type-2 inflammation and remodels airway tissue via multiple effector cells.[52]
TGF-βFibroblasts, airway smooth muscleDrives subepithelial fibrosis and smooth muscle proliferation, key processes in airway remodeling.[56]
TNF-αNeutrophils, eosinophils, macrophagesPotent pro-inflammatory mediator that recruits innate cells and exacerbates airway inflammation.[57]
IFN-γTh1 lymphocytes, macrophagesExerts a modulatory role by counterbalancing Th2 activity but may also contribute to chronic inflammation.[58]
TSLPDendritic cells, Group 2 innate lymphoid cells (ILC2), eosinophils, basophilsFunctions as an epithelial cell-derived alarmin; activates dendritic cells and ILC2s, promoting type 2 inflammation and bridging innate and adaptive immune responses; key initiator of airway inflammation and a therapeutic target for tezepelumab[59]
Table 2. Clinical studies evaluating the efficacy of omalizumab in asthma.
Table 2. Clinical studies evaluating the efficacy of omalizumab in asthma.
Study/Trial NameStudy TypeKey FindingsReference
Systematic Review25 Randomized Controlled TrialsOmalizumab led to ~25% reduction in asthma exacerbations and reduced need for ICS use.[105]
INNOVATE StudyRandomized Controlled TrialIn patients with severe asthma on high-dose ICS + LABA, omalizumab significantly reduced exacerbations.[106]
EXTRA TrialRandomized Controlled TrialOmalizumab treatment resulted in a ~25% decrease in asthma exacerbation rates.[92]
Prospective StudyObservational (806 patients)87% of patients showed improvement in asthma symptoms and lung function.[107]
Table 3. Anti-IL-5 and IL-5Rα biologics approved for severe eosinophilic asthma.
Table 3. Anti-IL-5 and IL-5Rα biologics approved for severe eosinophilic asthma.
BiologicMechanism of ActionApproved AgeEosinophil ThresholdDosageKey Efficacy ResultsNotable Trials
Mepolizumab (Nucala)Anti-IL-5; blocks IL-5 from binding to IL-5Rα on eosinophils≥6 years≥150 cells/μL100 mg SC every 4 weeks53% reduction in exacerbations (MENSA); OCS reduction (SIRIUS); QoL improvement (MUSCA)MENSA, SIRIUS, MUSCA [117]
Reslizumab (Cinqair)Anti-IL-5; binds circulating IL-5 to prevent receptor engagement≥18 years>400 cells/μL for best responseIV infusion (dose vary by weight) every 4 weeks92% eosinophil reduction; 50% and 41% reduction in exacerbations; 0.160 L FEV1 improvementStudy 3082 (Phase 3) Study 3083 (Phase 3) 3084 (Phase 3); Study 3085 (Phase 3) [118]; REALITI-A real-world cohort [119]
Benralizumab (Fasenra)Anti-IL-5Rα; induces ADCC leading to eosinophil apoptosis≥12 years>300 cells/μL30 mg SC every 4 weeks (3 doses), then every 8 weeks51% reduction in exacerbations (SIROCCO, CALIMA); 75% reduction in oral corticosteroid use (ZONDA); FEV1 improvementSIROCCO, CALIMA, ZEPHYR 2, ZONDA [120]
Table 5. Key clinical studies evaluating reslizumab in severe eosinophilic asthma.
Table 5. Key clinical studies evaluating reslizumab in severe eosinophilic asthma.
EffectStudy TypeKey FindingsReference
Eosinophil Reduction Study (Phase 3)Clinical TrialReslizumab reduced eosinophil levels by 92%, compared to 21% in placebo group.[132]
Exacerbation Reduction Trial 1 (Phase 3)RCT, Double-Blind, Placebo-ControlledDemonstrated a 50% decrease in asthma exacerbations in reslizumab-treated patients.[133]
Exacerbation Reduction Trial 2 (Phase 3)RCT, Double-Blind, Placebo-ControlledShowed a 41% reduction in asthma exacerbation rate with reslizumab use.[134]
Lung Function Improvement Study (Phase 3)RCTFound a 0.160 L greater improvement in FEV1 in the treatment group vs. placebo.[135]
Regulatory SummaryClinical OverviewApproved as add-on therapy for adults (≥18 years) with severe eosinophilic asthma.[131]
Stratified Eosinophil Response Study
(Phase 3)
Clinical TrialEnhanced lung function seen only in patients with eosinophil counts > 400 cells/μL; no FEV1 improvement in lower counts.[136]
Table 7. Clinical trials evaluating dupilumab across multiple type 2 inflammatory diseases.
Table 7. Clinical trials evaluating dupilumab across multiple type 2 inflammatory diseases.
Disease AreaStudy Name (Clinical Trial ID)Participant CriteriaDosage SchemeStudy DurationPrimary Outcome
Atopic DermatitisSOLO-1 (Phase 3; NCT02277743)Adults (≥18 yrs), moderate to severe cases, n = 671300 mg weekly/biweekly; placebo16 weeksInvestigator’s Global Assessment (IGA) score 0/1 with ≥2-point drop [149]
SOLO-2 (Phase 3; NCT02277769)Adults (≥18 yrs), moderate to severe, n = 708300 mg weekly/biweekly; placebo16 weeksIGA score 0/1 and ≥2-point drop [149]
CHRONOS (Phase 3; (NCT02260986)Adults (≥18 yrs), moderate to severe, n = 740300 mg weekly/biweekly; placebo52 weeksIGA score 0/1 and ≥2-point drop [149]
LIBERTY AD ADOL (NCT03054428)Adolescents 12–17 yrs, moderate to severe<60 kg: 200 mg biweekly; ≥60 kg: 300 mg biweekly; placebo16 weeksIGA score 0/1 and ≥2-point drop [149]
LIBERTY AD PEDS
(Phase 3; NCT03345914)
Children 6–11 yrs, severe<30 kg: 100 mg biweekly; ≥30 kg: 200 mg biweekly or 300 mg every 4 weeks; placebo16 weeksIGA score 0/1 [150]
LIBERTY AD PRESCHOOL (Phase 3; NCT03346434)Children 6 months–5 years, moderate to severe5–15 kg: 200 mg every 4 weeks; 15–30 kg: 300 mg every 4 weeks; placebo16 weeksIGA score 0/1 [150]
AsthmaLIBERTY ASTHMA QUEST (Phase 3; NCT02414854)Patients ≥ 12 yrs, moderate to severe, n = 1902200 mg or 300 mg biweekly; placebo52 weeksExacerbation frequency; FEV1 change [151]
LIBERTY ASTHMA VOYAGE (Phase 3; NCT02948959)Children 6–11 yrs, moderate to severe, n = 408<30 kg: 100 mg biweekly; ≥30 kg: 200 mg biweekly; placebo52 weeksAnnualized exacerbation rate [152].
hronic Rhinosinusitis with Nasal PolypsNP SINUS-24 (Phase 3; NCT02912468)Adults (≥18 yrs), severe symptoms, n = 276300 mg biweekly; placebo24 weeksChange in nasal congestion, polyp score, and CT findings [153]
NP SINUS-52 (Phase 3; NCT02898454)Adults (≥18 yrs), severe symptoms, n = 448300 mg biweekly for 24 weeks, then monthly; placebo52 weeksChange in nasal polyps, congestion, CT at 24 weeks [153]
Eosinophilic EsophagitisParts A, B, C (Phase 3; NCT03633617)Adults ≥ 12 yrs300 mg weekly or biweekly; placebo24–28 weeksHistological remission (≤6 eos/hpf), symptom reduction [154].
EoE KIDS (Phase 3; NCT04394351)Children 1–11 yrs<30 kg: 200 mg biweekly; 30–60 kg: 300 mg biweekly; placebo16–52 weeksHistologic response defined as ≤6 eosinophils per hpf [155].
Table 8. Summary of clinical trials evaluating tralokinumab in patients with moderate-to-severe asthma.
Table 8. Summary of clinical trials evaluating tralokinumab in patients with moderate-to-severe asthma.
Study (ID)Japanese LTS
Phase 3
MESOS
Phase 2
TROPOS
Phase 3
STRATOS 2
Phase 3
STRATOS 1
Phase 3
Subjects Enrolled28791408561207
Lead-in PhaseUp to 2 weeks2 weeks4–6 weeks4–6 weeks
Active Treatment Span52 weeks12 weeks40 weeks52 weeks52 weeks
Post-treatment Follow-up14 weeks14 weeks14 weeks20 weeks20 weeks
Enrollment CriteriaJapanese adults with asthma not controlled on ICS + LABAAsthmatics uncontrolled on ICS aloneAsthmatics requiring maintenance OCS + ICS/LABAAdults with asthma inadequately managed on ICS/LABAAdults with asthma inadequately managed on ICS/LABA
Investigational Dose300 mg SC every 2 weeks300 mg SC every 2 weeks300 mg SC every 2 weeks300 mg SC every 2 weeks300 mg SC every 2 weeks or 300 mg SC every 4 weeks
Comparator ArmNone (open-label)Placebo SC every 2 weeksPlacebo SC every 2 weeksPlacebo SC every 2 weeksPlacebo SC every 4 weeks
Primary AimEvaluate long-term tolerability and safety in Japanese patientsExamine impact on eosinophil-driven airway inflammation and remodelingDetermine whether tralokinumab allows reduction in maintenance OCS doseConfirm efficacy/safety in biomarker-positive subgroupAssess efficacy/safety in overall population and identify biomarker-positive subgroup
Primary EndpointRate and severity of adverse events (AEs) [190]Change in submucosal eosinophil density (cells/mm2) after 12 weeks [190]Percent change in average daily OCS dosage at week 40 [191]Annualized asthma exacerbation rate (AAER) through week 52 [192]AAER through week 52 [192]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Văruț, R.M.; Dalia, D.; Radivojevic, K.; Trasca, D.M.; Stoica, G.-A.; Adrian, N.S.; Carmen, N.E.; Singer, C.E. Targeted Biologic Therapies in Severe Asthma: Mechanisms, Biomarkers, and Clinical Applications. Pharmaceuticals 2025, 18, 1021. https://doi.org/10.3390/ph18071021

AMA Style

Văruț RM, Dalia D, Radivojevic K, Trasca DM, Stoica G-A, Adrian NS, Carmen NE, Singer CE. Targeted Biologic Therapies in Severe Asthma: Mechanisms, Biomarkers, and Clinical Applications. Pharmaceuticals. 2025; 18(7):1021. https://doi.org/10.3390/ph18071021

Chicago/Turabian Style

Văruț, Renata Maria, Dop Dalia, Kristina Radivojevic, Diana Maria Trasca, George-Alin Stoica, Niculescu Stefan Adrian, Niculescu Elena Carmen, and Cristina Elena Singer. 2025. "Targeted Biologic Therapies in Severe Asthma: Mechanisms, Biomarkers, and Clinical Applications" Pharmaceuticals 18, no. 7: 1021. https://doi.org/10.3390/ph18071021

APA Style

Văruț, R. M., Dalia, D., Radivojevic, K., Trasca, D. M., Stoica, G.-A., Adrian, N. S., Carmen, N. E., & Singer, C. E. (2025). Targeted Biologic Therapies in Severe Asthma: Mechanisms, Biomarkers, and Clinical Applications. Pharmaceuticals, 18(7), 1021. https://doi.org/10.3390/ph18071021

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

Article Metrics

Back to TopTop