Open access peer-reviewed chapter - ONLINE FIRST

Navigating the Terrain: Type 2 Cytokines and Biologic Intervention in Severe Eosinophilic Asthma

Written By

Brian W.P. Seymour, Ashley A. Seymour and Patricia M. Gilford

Submitted: 07 June 2024 Reviewed: 07 June 2024 Published: 23 August 2024

DOI: 10.5772/intechopen.1006274

Eosinophils and Their Role in Human Health and Disease IntechOpen
Eosinophils and Their Role in Human Health and Disease Edited by Seyyed Shamsadin Athari

From the Edited Volume

Eosinophils and Their Role in Human Health and Disease [Working Title]

Seyyed Shamsadin Athari, Entezar Mehrabi Nasab and Luis Rodrigo

Chapter metrics overview

30 Chapter Downloads

View Full Metrics

Abstract

Asthma is a chronic respiratory disease characterized by bronchial hyperreactivity. There are several endotypes of which allergic asthma is the most common. Severe eosinophilic asthma is prevalent in approximately 5% of asthmatics and its phenotype overlaps with allergic asthma and type 2 inflammation. Patients with refractiveness to corticosteroids underline the difficulty in controlling persistent inflammation in severe eosinophilic asthma. The focus of biological therapies is geared towards the understanding of the intricate interplay of the cytokines that drive the eosinophil’s ability to induce chronic inflammation with airway obstruction. This chapter takes the reader down a historical journey of initial studies that were performed using mouse helper T cell clones for reconstitution experiments to unravel the mechanism of the role T helper 2 cytokines play in allergic asthma. We then reviewed the classic in vivo experiments that demonstrated how antibodies to IL5 can down regulate eosinophils in the blood and their progenitors in the bone marrow of mice. We also delve into the complex interaction of the alarmins on the cytokines triggers of allergic inflammation with elevated eosinophils. Finally, we review the clinical literature on the beneficial effects of humanized monoclonal antibodies in use for treatment of patients suffering from severe eosinophilic asthma.

Keywords

  • cytokine
  • antibody
  • alarmin
  • asthma
  • eosinophil
  • inflammation

1. Introduction

Asthma is a chronic respiratory disease characterized by bronchial hyperreactivity (narrowing of the airways), wheezing and tightness of the chest [1, 2]. It affects more than 300 million people worldwide with approximately 25 million in the United States [3, 4, 5, 6]. It is one of the most chronic diseases in children with the morbidity and mortality rates highest among African American children in the United States [7]. They are at least 10 times more likely to die from asthma than their white counterparts [7, 8, 9]. It is a complex and heterogenous disease with varying severity that has a great spectrum of symptoms and wide differences in treatment efficacy [10]. The future in asthma management is to classify its many patterns known as endotypes, that connect recognizable characteristics with immunological mechanisms [5]. These endotypes can range from allergic bronchopulmonary aspergillosis, viral-exacerbated asthma, exercise-induced asthma to allergic and eosinophilic asthma [5, 10, 11].

Allergic asthma is the most common endotype as it affects approximately 66% of patients with asthma and more than half of the patients with severe asthma [12]. It is characterized by elevated levels of serum IgE (>0.35kU/L) with specificity for allergens in sensitized individuals [13]. These allergens are often innocuous and ubiquitous such as the antigens in house dust mites, pollen, dog and or cat dander. Atopic individuals diagnosed with allergic asthma show a positive skin prick test to allergens along with bronchial hyper-responsiveness and elevated levels of blood eosinophils [13].

Eosinophilia of the airways is observed in greater than 50% of all asthmatics. Those patients with sputum eosinophilia (≥3%) and blood eosinophil levels of ≥300cells/μl are designated as having severe eosinophilic asthma [13, 14]. High blood eosinophil count is a risk factor for asthma [15, 16]. Individuals exposed to tobacco smoke as well as ex-smokers have significantly higher levels of eosinophils when compared to non-smokers [17]. Indeed, in a murine model of asthma, mice exposed to environmental tobacco smoke had significantly higher levels of blood eosinophils and increases in bronchial hyperreactivity when compared to those exposed to filtered air [18]. Patients with prolonged eosinophilic asthma will have an accumulation of eosinophils not only in the blood and sputum but also in the bronchial tract. These patients will have a basement membrane zone of the airways that is thickened. Here, eosinophils will produce cytokines, chemokines and other mediators of inflammation [19] resulting in airway obstruction and airway remodeling [20]. This remodeling results in subepithelial fibrosis, thickening of the sub-basement membrane, increase airway smooth muscle mass and mucous gland hyperplasia with consequences of asthma exacerbation [19]. The Global Initiative for Asthma (GINA) describes asthma exacerbation as the inability of an asthmatic to respond to inhaled corticosteroids (ICS) due to high levels of inflammation in the airways with consequences of tightness of the chest, wheezing and decrease in lung function [21].

The European Academy of Allergy and Clinical Immunology Biological Guidelines from 2020 states that allergic asthma and eosinophilic asthma are subtypes of a type 2 (T2)—high inflammation [22]. A T2 high phenotype is characteristic of patients with high levels of blood eosinophil and IL5 in bronchial biopsies. A common biomarker for a T2-high inflammation is a fractional exhaled nitric oxide (FeNO) of ≥35 ppb [13]. Clinical studies show that many of these patients met the definition of both allergic and eosinophilic asthma demonstrating overlapping characteristics between both groups [13, 23]. Severe eosinophilic asthma patients will express a T2-high asthma phenotype with a biomarker of FeNO of ≥35 ppb [13]. These patients also exhibit a T helper (Th)2 cytokine profile (IL4, IL5, IL9, IL13) with IL5 significantly elevated in those with severe asthma [5, 24].

Asthma in many patients is controlled by treatment using standard protocol which includes ICS and beta2 adrenergic bronchodilators [25]. However, 5–10% of patients on corticosteroids do not respond to these medications resulting in persistent inflammation [14]. At center stage of patients with persistent inflammation is the eosinophil and the major cytokine-IL5. IL5 plays the most important role in the growth, differentiation, activation, recruitment and survival of eosinophils [26, 27, 28]. Excessive amount of IL5 in the bronchial region is believed to be responsible for the persistent inflammation and the ineffectiveness of corticosteroids in patients with severe eosinophilic asthma [29].

Severe eosinophilic asthma has now become the focus of many new therapies due to the eosinophil’s ability to induce chronic airway inflammation, leading to edema, mucus plugging and ultimately airway remodeling and its refractiveness to corticosteroids [30, 31]. The presence of IL5 has been implicated in the prolonged inflammation and remodeling of the airways as shown by several investigators [32, 33, 34]. The production of IL5 in this capacity involved a series of complex interactions involving type 2 innate lymphoid cells (ILC2), Th2 cells, the cytokines produced by them and other cells of the immune system. In this chapter, we delve into the early studies (in vitro and in vivo) that paved the way to the understanding of the cytokine functions particularly IL4 and IL5 and the series of intricate interactions that lead to severe eosinophilic asthma. We conclude by discussing the efficacy of biologics in use to treat severe eosinophilia in patients with T2-high asthma.

Advertisement

2. Early in vitro studies helped to unravel the functions of IL4 and IL5

In the early 1980s, investigators from the laboratories of Vitetta used supernatants from a specific T helper cell clone in culture which when added to liposaccharide (LPS) activated murine B cells caused enhancement of IgG1 while inhibiting the production of IgG3 and IgG2b [35]. When the supernatant was analyzed using biochemical procedures, it was found that it contained the B cell stimulatory factor-1 which today is known as IL4 [36]. Indeed, when anti- IL4 antibodies were added to these cultures, this enhancing effect was abrogated [36]. Later, Coffman et al. observed that LPS stimulated murine splenic B cells in culture produced elevated levels of IgG1, IgG2a, IgG2b, IgM but no IgE [37]. However, when IL4 was added to the cultures of splenic B cell, IgE was produced along with the other isotypes mentioned above showing that IL4 was responsible for the production of IgE [37]. When IL5 was added to the LPS stimulated B-cell cultures, there was a significant increase in the production of IgA and it was markedly increased when the combination of IL4 and IL5 were added.

Soon after the discovery by Mosmann et al. [38] that T helper cells can be of 2 types; namely Th1 and Th2, immunologists began the tedious process of functionally characterizing the roles of these cells. With the use of mouse T helper clones, scientists were able to decipher the functions of the cytokines secreted by these T cell clones. Coffman et al. performed in vitro reconstitution experiments in which they used Th1 and Th2 clones to show how T cell can help B cells produce various isotypes [39]. The major cytokines produced by Th1 clones are IL2 and IFN-γ while Th2 clones produced IL4 and IL5. Studies revealed that when Th2 clones were stimulated in 7-day culture with mouse B cells, they produced elevated levels of the IgE, IgG1, IgM and IgA. Removal of IL4 with the use of anti-IL4 antibodies, significantly blocked the IgE response while only slightly reducing the other isotype responses.

In these early studies, Coffman et al. were also able to reconstitute the Th2 response using the Th1 clones [39]. Here, an autoreactive mouse T cell clone called H66-61 Th1 cell line was used to reconstruct a Th2 type response. Irradiated H66-61 Th1 cells were placed in 7-day culture with mouse B-cells. Therefore, these irradiated T cells cannot proliferate but can provide help to B cells. Since Th1 clones secrete IFN-γ and this cytokine can inhibit the function of IL4 in culture, anti-IFN-γ was added to all cultures. Recombinant (r) IL4 and or IL5 cytokines were added to the cultures yielding the isotypes as shown in Table 1. The results demonstrated that Th1 clone was able to provide help to the B-cells for the secretion of the isotypes that are characteristics of a Th2 response. The similarity in isotype production was observed when supernatant from a Th2 clone (D9-sup) was added to B cell cultures. Furthermore, these results showed that IL4 triggered a significant increase in the isotypes IgM, IgG1 and especially IgE. The addition of IL5 to the cultures showed a significant increase in IgA but not IgE. Previously, Sanderson et al. in 1985 used a bone marrow culture system to show that a cytokine from a specific T cell clone that is not IL2 or IL3 is an eosinophil differentiating factor [40]. Molecular cloning and genetic characterization revealed that this factor is indeed IL5 which is a growth factor for eosinophils [41, 42]. These early in vitro studies revealed that Th2 cells and their cytokines (mainly IL4 and IL5) played a prominent role in allergic responses.

H66-61 Cells + B Cells + Anti-IFN-γ +IgEIgG1IgMIgA
Medium<1227159030
r IL4610416011,10041
r IL5<1138015,200208
r IL4 + r IL533312,30064,300281
D9-Sup57720,70091,900407

Table 1.

The effects of recombinant (r) IL4 and IL5 on H66-61-stimulated B cell cultures rIL4 was added at a concentration of 500 U/ml, rIL5 at 5 U/ml, D9 supernatant at 3%, anti-IFN- at 5 g/ml.

All additions were added on day 0 of the cell cultures.

Advertisement

3. Murine models of parasitic infection, allergy and asthma helped to demonstrate the in vivo functions of the allergic cytokines -IL4 and IL5

The role of IL4 and IL5 was further defined in experimental models using rodents infected with the nematode Nippostrongylus brasiliensis (Nb) larvae [43, 44]. Nb larvae, upon infection in mice pass through the lung where they molt during the first few days triggering an immune response with blood and lung eosinophilia and elevation of IgE antibodies. Coffman et al. performed an elegant study with the use of monoclonal antibodies (mAb) to IL4 and IL5 to functionally characterize the role of these cytokines in vivo [43]. BALB/c mice were injected subcutaneously (SC) with 750 third stage Nb larvae. On the same day, they were treated with 2 mg per mouse of an anti-IL5 (TRFK-5) mAb. Another infected group received 10 mg/mouse of an anti-IL4 (11B11) mAb instead of TRFK-5 mAb. Nb infected BALB/c mice showed a 25- to 100-fold increase in total IgE levels and a 4- to 8-fold increase in eosinophils. However, the group that was treated with anti-IL5 showed no increase in blood and lung tissue eosinophils but made the normal increases in serum IgE as expected from the infection. This inhibition of eosinophils by TRFK-5 also blocked the development of eosinophils from the progenitors in the bone marrow [44]. The infected group that was treated with 11B11 made significantly reduced levels of total serum IgE (Table 2) but made elevated levels of eosinophils as characteristic of an Nb-infected mouse. The inability of TRFK-5 to block the IgE response was consistent with previous in vitro work by Coffman et al. [39]. This was the first in vivo model to demonstrate the ability of antibodies to IL5 to block eosinophilia. This seminal study on the independent regulation of IgE and eosinophils was later confirmed by others using mouse models of allergy, asthma and allergic aspergillosis [45, 46].

TreatmentTotal serum IgE (μg/ml) on days
-71114
No antibody0.87 (0.43)32.4 (9.7)31.1 (11.2)
Anti-IL-50.53 (0.34)34.3 (19.9)34.9 (20)
Anti-IL-41.85 (0.83)2.5 (1.6)3.5 (2.4)
IgG1 Control1.15 (0.23)12.0 (6.7)24.8 (7.7)

Table 2.

Total serum IgE in BALB/c mice after subcutaneous injection of Nippostrongylus brasiliensis.

Savelkoul et al. further clarified the necessity of IL4 for an IgE response from the in vivo model of inflammation using Nb infected SJA/9 mice [47]. SJA/9 mice genetically have an SJL background (H-2s) and bear the BALB/c H chain allotype [Iga] [48]. They are unable to mount IgE responses to allergenic substances and helminthic parasites [48]. When the T cells of Nb- infected SJA/9 mice were placed in culture with the mitogen concanavalin A, IL4 was produced in the supernatant. Furthermore, when B cells from Nb infected SJA/9 mice were placed in culture with IL4 and LPS, IgE was produced. Similarly, the administration of rIL4 to Nb infected SJA/9 mice resulted in IgE production as they made significant levels of serum IgE which was comparable to strains that are normal IgE responders. These Nb infected mice made eosinophils that were blocked when these mice were treated with anti-IL5. Thus, IgE defect was restored by administration of IL4 to these SJA/9 mice. This study further confirmed that the contribution of IgE and eosinophil to immune responses are controlled independently by IL4 and IL5 respectively. Today, it is known that IL4 is a switch factor for the production of IgE [49] and IL5 does not act on human B cells but is the major cytokine responsible for eosinophil maturation, differentiation and survival [19].

Advertisement

4. How strain variations in mice influenced the allergic response to the allergenic substance ovalbumin (OVA)

A study was done to examine the allergic response from various strains of mice that were classified by the quantity of allergic antibodies (lgE) they produced after immunization with the allergenic substance- ovalbumin (OVA) [50]. These classifications were termed “high,” “low,” and “non” responders according to the levels of IgE mice produce upon stimulation with allergens. Therefore, experiments were performed on SJA/9, BALB/c, C57BL/6 and 129SvEv mice to examine their abilities to make an allergic response. Except for the IgE non responder strain SJA/9, all strains are capable of mounting IgE responses to allergenic substances. The aim of this study was to understand the genetics of these strains of mice with respect to IgE production to gain an understanding of the immunogenicity of individuals with a genetic predisposition to allergy and asthma.

Mice were immunized by intraperitoneal (IP) injection with 1O µg OVA in aluminum hydroxide (OVA/AL) on day O followed by exposure to a 1% aerosolized OVA which was done on day 27. Using this experimental protocol, OVA-specific lgE was elevated in all groups except in the SJA/9 strain in which it was undetectable (Figure 1). It was hypothesized that this lack of lgE secretion in SJA/9 mice is due to a mechanism which is similar to that which is responsible for the nonallergic state in some individuals. OVA specific IgE peaked in all IgE responsive groups at about day 8 after the challenge with aerosolized OVA. The largest amount of lgE was seen in the 129SvEv mice while the lowest detectable amount was from the C57BL/6. After the peak response, OVA specific-lgE remained detectable in the last collection of serum which was on day 100.

Figure 1.

IgE responses from various strains of mice after exposure to OVA.

Eosinophils in the blood were quantified after the first inhalation of aerosolized OVA (data not shown). All mice had elevated levels of blood eosinophils 5 days after this inhalation. Although, there were no lgE in the blood of SJA/9, this group had the highest level of blood eosinophils. Two weeks after the aerosol challenge, eosinophils remained significantly elevated in the 129/SvEv group when compared to the others.

At this point, it was necessary to examine the cytokine profile in the lung of each strain of mice. Thus, on day 150, all mice were rechallenged with a 1% aerosolized OVA and their lung cytokines assessed 4 days after this tertiary challenge. Table 3 shows that despite the high levels of OVA-specific lgE from the 129/SvEv group, IL4 was very low in this group. OVA-specific lgE and IL4 were undetectable in SJA/9 mice. The highest level of IL4 was seen in BALB/c mice with 888 ± 301 ng/ml in the lung of these mice. IFN-γ was not detected in any of the groups demonstrating that the immune response was not a Th1 type response. IL3 was detected in the BALB/c mice but not in the other groups.

StrainIL3 (ng/ml)IL4 (pg/ml)IL5 (ng/ml)IFN-γ (ng/ml)
SJA/9<0.156<1.561.11 ± 0.41<0.156
C57/BL9<0.15670 ± 243.09 ± 1.00<0.156
BALB/c0.556 ± 0.0830888 ± 3018.94 ± 2.09<0.156
129/SvEv<0.15634 ± 103.56 ± 1.24<0.156

Table 3.

Lung cytokines from OVA immunized strains of mice.

All mice received 10 ug OVA/AL IP on day 0 and 1% aerosolized OVA for 20 minutes on day 27 and day 150. In vitro restimulation of unseparated lung cells were done on day 154.

The result of this experiment confirms previous studies that IL4 is a necessary cytokine for the production of lgE [47]. That is, SJA/9 mice did not produce detectable levels of lgE and did not produce any IL4 after the OVA specific in vitro restimulation of homogenized lung cells. This study also shows that IL4 is necessary but not fully responsible for the production of lgE since the highest levels of lgE came from the 129SvEv mice even though IL4 in this group was very low compared to the BALB/c group. Presently, it is known that IL13 also plays a role in the production of IgE [51]. Low levels of IL5 were seen in the SJA/9 mice when compared to the BALB/c mice (1.11 ± 0.41 vs. 8.94 ± 2.09 respectively). However, the eosinophils in the SJA/9 strains were highest when compared to the other groups. Despite the absence of IL4 and lgE from the SJA/9 strain, a Th2 response (eosinophilia and Th2 cytokines) was seen in this group. The lack of IFN-γ from all the strains revealed that these animals were not responding with a Th 1 type response, instead they are capable of producing an allergic type of response even in the SJA/9 strain that lacked detectable lgE and IL4. The level of the response from each strain may represent the atopic state of a particular strain. The SJA/9 strain represents the low responders while the 129/SvEv with their high levels of lgE may be mice which can be comparable to atopic individuals.

Advertisement

5. The complex role of the cytokines and inflammatory mediators in severe eosinophilic asthma

5.1 Damaged epithelial cells release alarmins to initiate the innate immune response

Numerous studies have been performed which have recognized the major regulatory mechanisms in allergic airway diseases [4, 52, 53]. There are many multifactorial events that are necessary for a non-allergic individual to develop allergic airway disease such as severe eosinophilic asthma (Figure 2). It begins with damaged lung epithelial cells after an atopic individual is exposed to aeroallergens such as pollen and/or environmental irritants such as tobacco smoke [54]. The damaged epithelium secretes the alarmins -IL33, IL25 and TSLP [55, 56]. These cytokines can cause a shift in the immune response towards an allergic phenotype with pathological consequences related to airway allergic disease [55]. For example, in a mouse model of asthma, mice treated intranasally with IL25, developed epithelial cell hyperplasia, mucus hyper-secretion and bronchial hyperreactivity [57]. Indeed, severe asthmatic patients with fixed airflow limitation show an increase in IL25 [58]. Others have shown that the level of circulating fibrocytes (progenitor cells that enter the circulation and inflame the bronchial epithelium) bearing the receptor for IL25 correlates with the severity of asthma [59].

Figure 2.

The interaction of the alarmins with the T2 cytokines and the prolongation of eosinophilia from damaged epithelial cells of the airways (image created with BioRender.com).

IL33 is a member of the IL1 family which participates in the polarization of the immune response towards a T2 type inflammation. It is expressed not only by epithelial cells but also by vascular endothelia cells. Atopic patients chronically exposed to environmental pollutants or allergens containing serine proteases will show an early increase in IL33 from their damaged epithelial cells. This cytokine is present in the bronchoalveolar lavage and airway smooth muscles, and they activate dendritic cells - the major antigen presenting cells [60]. Thus, the expression of IL33 in humans can lead to atopic diseases with elevated levels of IgE, hyper eosinophilia and asthma.

The thymic stromal lymphopoietin (TSLP) is another cytokine that is secreted by the damaged epithelial cell in response to proteases [61, 62]. TSLP is a member of the IL2 cytokines family, and its gene can be found on the human chromosome at 5q22.1 which is close to the cluster of cytokines- IL4, 5 and 13 located at 5q3185 [63]. TSLP is also expressed in lung fibroblast, and smooth muscle. Its receptor is expressed on innate lymphoid cell type 2 (ILC2), dendritic cells, T cells, B cells, mast cells, basophils, monocytes and eosinophils [61]. It is involved in dendritic cells maturation and the skewing of T helper cells to a Th2 phenotype particularly during an allergic response [64]. Indeed, TSLP’s mRNA and protein are elevated in asthmatic patients and correlate with airway hyperresponsiveness regardless of the levels of eosinophilia and T2 inflammation [52, 65].

Collectively, the alarmins work together to activate the ILC2 cell during the innate phase of the allergic response by contributing to the initiation of the inflammatory response. ILC2 cells are the early producers of several T2 cytokines including IL4, IL5, IL9, IL13 [66]. The ILCs which arise from the common lymphoid progenitor are closely related to the Natural Killer (NK) cells. They reside in peripheral tissues and are categorized into 3 groups namely 1, 2 and 3 according to the cytokines they produce and their surface characteristics [67].

It was found that when ILC cells were stimulated with IL25 and IL33, the neuropeptide receptor Nmur1 was expressed preferentially by ILC2 cells. Furthermore, when neuromedin U (NMU), the ligand for Nmur1 was co administered with IL25, they amplified the allergic inflammation with increases in eosinophils in the lung and broncho alveolar lavage [68]. TSLP and to a lesser extent IL25 and IL33 from epithelial cells causes the release of IL5 from ILC2 cells. Also, studies have shown that when TSLP was added to IL33-stimulated ILC2 human cells, the production of IL4, IL5 and IL13 was enhanced along with increase expression of the transcription factor GATA3 [69]. This transcription is required for the expression of IL5 from Th2 cells [69]. IL13 produced by ILC2 cells is necessary for the migration of dendritic cells to the draining lymph nodes where they induce naïve T cells to become Th2 cells during the process of T cell activation. TSLP stimulated dendritic cells has been shown to lead to the production of chemokines CCL17 and CCL21 and ultimately to the priming of Th2 cells [64]. In chronic conditions, TSLP downregulates the development of Tregs while maintaining the Th2 type inflammation [64]. Indeed, TSLP is increased in the airway walls of patients with severe asthma. Thus, the ILC2 cell, due to its early secretion of cytokines, participates in the innate phase and is necessary for the adaptive phase of the allergic response as naïve T cells differentiate into effector Th2 cells [70, 71].

IL3, IL5 and GM-CSF participate in the development of eosinophils however, IL5 is the major cytokine for its development, and it is at center stage in patients with severe eosinophilic asthma [31]. Though the main cellular sources of IL5 are Th2 and ILC2 cells, it is also secreted by other cells such as mast cells, basophils, NKT cells and eosinophils themselves pointing to the increase in eosinophils as largely due to the many sources of IL5 [26, 72, 73, 74]. Meanwhile, the damaged epithelial cells also secrete chemokines CCL5 (RANTES) and CCL11 (eotaxin-1) that can bind to a receptor called CCR3. Also, IL4 and IL13 can stimulate epithelial cells in the lungs to produce eotaxins [75]. The CCR3 receptor is expressed on Th2 cells, macrophages, eosinophils, basophils and mast cells [31, 76]. While eotaxin-1 is required for the initial steps in the inflammatory response, eotaxin-2 and eotaxin-3 participate in the prolongation of eosinophil survival [31]. Thus, there will be an influx of these cells from the blood to the respiratory tract which contributes to the obstruction of the airways.

5.2 Th2 type allergic response is responsible for the adaptive phase and participates in the prolongation of the chronic inflammation in severe eosinophilic asthma

After the initial phase of the allergic response, Th2 cells with the series of cytokines they produce which are mainly IL4, IL5, and IL13 play a prominent role in propagating the chronic inflammation seen in severe eosinophilic asthma. IL4, a switch factor for IgE, is largely responsible for the increase in allergic antibodies. IgE attaches to mast cells via the high affinity Fc epsilon receptor. If crosslinking of antigen occurs via the fragment antigen-binding (FAB) region, then the mast cell becomes degranulated as shown in Figure 2 with the release of inflammatory mediators such as histamine, chemotactic factors, cytokines, metabolites of arachidonic acid. These mediators act on vasculature, goblet cells, smooth muscles and inflammatory cells in the airways to bring about bronchoconstriction [77]. IL4 also promotes the commitment for the differentiation of naïve T cells to Th2 cells. The IL4 receptor activates STAT6 from the naïve T cell which promotes the expression of the transcription factor -GATA3. The B lymphocyte-induced maturation protein-1 (Blimp-1) enhances the GATA3 expression in T helper cells causing them to differentiate to Th2 cells with amplification of the inflammatory allergic response in atopic individuals exposed to aeroallergens [78]. Therefore, IL4 with its ability to cause naïve T cells to commit to a Th2 pathway, plays a prominent role in the prolonged elevation of IL5 in severe eosinophilic asthma. Epithelial cell activation occurs due to the presence of IL-5, IL-13, and periostin, (a protein upregulated by IL-13) which also causes the biomarker FeNO for a Th2 inflammation to be expressed [79]. Together, IL13 and IL9 have a role in bronchial hyperreactivity and the remodeling of the airway epithelium by causing epithelial cells to differentiate into goblet cells [4]. The increase in goblet cells can lead to a hyper secretion of mucus in an asthmatic individual [4, 80, 81].

The high levels of IL5, particularly in the bronchial tracts, in patients with severe eosinophilic asthma can hinder the pro-apoptotic effect from the treatment with corticosteroids [82]. This refractory state is defined as severe T2-high eosinophilic asthma in which eosinophils accumulate and proliferate throughout the airways. The IL5 receptor alpha chain is expressed on eosinophils, airway epithelial cells and lung fibroblast [78, 83]. The elevated presence of IL5 and eosinophils can lead to the development of co-morbidities such as chronic rhinosinusitis and nasal polyps due to the continuous production and survival of eosinophils in the bronchial tracts [79]. These patients experience frequent exacerbation of their allergic phenotype and difficulty controlling their respiratory conditions. The unraveling of the function of Th2 cytokines, particularly IL5, in maintaining the elevated levels of eosinophils has aroused the interest of researchers to explore the use of biological markers for therapeutic intervention in severe eosinophilic asthma [84, 85]. This development of biologics can be the key to control severe eosinophilic asthma when it is uncontrollable with the use of corticosteroids due to the high levels of IL-5.

Advertisement

6. The use of biologics for the treatment of eosinophilic asthma

The first published report that antibodies to IL5 can inhibit eosinophils in vivo was done using a mouse model of Nippostrongylus brasiliensis infected parasitized mice in 1989 [43]. Later scientists used a monkey model of asthma and a mouse model of pulmonary inflammation to also demonstrate the inhibitory effect of anti-IL5 on eosinophils [86, 87]. These preclinical studies paved the way for the use of anti-IL5 biologicals in the treatment of severe eosinophilic asthma. Over the last decade, biologics that target cytokines and cytokine receptors, have been developed to be used as an adjuvant therapy in the management of severe eosinophilic asthma [88, 89, 90, 91]. Several clinical trials have evaluated the efficacy of anti-IL5 or anti-IL5 receptor antibodies in patients with asthma [92, 93]. These biologics are: Mepolizumab, Reslizumab and Benralizumab.

Mepolizumab is a humanized mAb (IgG1 k type), originated by binding anti human IL5 antigen recognition sites from murine origins into a human IgG1 heavy chain [94]. It has a high affinity in binding free IL-5 (a 134 amino acid dimeric glycoprotein with 4 helix bundle mortif that has a 52-KDa homodimer) resulting in IL-5 inability to bind to the receptor IL5Rα (Figure 3) [94, 95]. This binding causes a reduction of eosinophils in both the blood and airways [96]. Its very specific binding ability may explain the lack of relatively significant side effects of Mepolizumab as it does not seem to interfere with the biological activities of other cytokines [94]. However, early studies with Mepolizumab showed mixed response from clinical trials conducted in several asthmatic patients’ population where there was documented reduction in circulating eosinophils without much significant clinical response in the severity of the disease [92, 97]. These early investigations included a small population study that was conducted on difficult to treat asthmatic patients whose treatment included high dose ICS and/or oral corticosteroid (OCS). This study also showed a similar outcome in reduction of blood eosinophils with little effect on the clinical outcome, except for a small improvement in lung FEV1 (forced expiratory volume in 1 second) function [98]. Many of these early studies were done in which a specific participant group consisted of various asthma phenotypes ranging from mild to moderate chronic asthma. Despite the reduction of eosinophils, the quality of life for these patients was not improved with respect to asthma exacerbation rates [93].

Figure 3.

The mechanisms of action of reslizumab, mepolizumab and benralizumab on eosinophils. Reslizumab and mepolizumab use different idiotypic regions to bind to the IL5 molecule. The FAB region of Benralizumab attaches to the IL5 receptor on the eosinophil and the Benralizumab’s Fc region attaches to the Fcγ receptor as shown on the NK cell to induce ADCC. The sources of IL5 are also shown (image created with BioRender.com).

Eventually, investigators examined patients suffering solely from severe chronic asthma [99]. This small study involved patients with bronchial eosinophilia who were not responsive to treatment with corticosteroids. The result of this study showed reduction not only in eosinophil levels but also in asthma exacerbations after treatment with Mepolizumab. Later, a 52-weeks study termed; the DREAM (Dose Ranging Efficacy And safety with Mepolizumab in severe asthma) study was conducted as one of the largest asthma studies (621 participants) to examine the effect of Mepolizumab in participants with severe asthma [100]. The study had strict criteria that required at least 2 exacerbation which included the use of OCS or a visit to an emergency room or hospitalization and signs of eosinophilic inflammation (either sputum eosinophils >3%, peripheral blood eosinophils >300 x 106/L, FeNO>50 ppb or loss of asthma control after <25% reduction in either ICS or OCS dose). Participants received 13 infusions of either 75, 250 or 750 mg of intravenous (IV) mepolizumab. Results showed all three doses significantly reduced asthma exacerbation equally and reduced both blood and sputum eosinophils [100].

Mepolizumab as Adjunctive Therapy in Patients with Severe Asthma (MENSA) was a study of 576 asthmatic patients over 52 weeks who were treated with ICS with or without OCS [101]. They were randomized to receive either 75 or 100 mg mepolizumab SC every 4 weeks or a placebo. The inclusion criteria were asthmatic with at least 2 exacerbations requiring systemic corticosteroids the previous year with evidence of eosinophilic inflammation (eosinophil count 150 cells/uL at screening or above 300 cells/μL at some point in the previous year). Results showed a 47 and 53% respectively in IV and SC mepolizumab associated reduction in asthma exacerbations [101].

Mechanistically, investigators have shown that after treatment with Mepolizumab, there is a decrease in PD-1 expression on Tregs hereby allowing this subset of cells to perform its immunomodulatory duty [102]. TGF-β is also reduced after severe asthmatics with eosinophilia are treated with Mepolizumab. This cytokine is secreted by many cells in the respiratory tract but is mainly liberated by eosinophils [103]. It is believed to play a role in the differentiation of fibroblast to myofibroblasts which participates in airway remodeling. Thus, with the treatment of severe asthmatics with Mepolizumab, there was a reduced expression of some extracellular matrix proteins and consequently the reduction of airway remodeling [31, 102]. Corticosteroids have been shown to be ineffective in reducing airway remodeling in patients with chronic asthma particularly those with severe eosinophilic asthma. Therefore, Mepolizumab is a necessary add-on therapeutic for the inhibition of fibrosis and airway remodeling of individuals with severe eosinophilic asthma which accounted for approximately 5% of all asthmatics [14, 31]. This treatment also reduces the severity of exacerbation even in patients with comorbidity such as bronchiectasis [104]. The FDA and the European Medicines Agency both eventually approved Mepolizumab as an adjuvant treatment for severe eosinophilic asthma [93].

Reslizumab, another anti-IL5 mAb showed significant improvement in lung function in a study (P = 0.002 vs. placebo) with trend towards improved asthma score and reduction in sputum eosinophils [96]. It is an IL-5 neutralizing IgG4K mAb, that is currently approved for adjunct therapy for adults with severe eosinophilic asthma [91]. Data from the BREATH phase 111 clinical trial which involved three double -blinded studies in patients 12–75 year with eosinophilic asthma not controlled on an ICS used reslizumab 3 mg/kg once every 4 weeks for 52 weeks. The results in various subgroups showed a significant reduction in asthma exacerbation frequencies, lung function and improved quality of life [105]. Ibrahim et al., explored the clinical efficacy of Reslizumab in patients with inadequately controlled asthma, elevated blood eosinophils, taking high dose ICS, a second controller with at least 4 exacerbations and one hospitalization [106]. The results were statistically significant through the use of a validated asthma control questionnaire. Of note, patients had a decrease in maintenance steroid usage while taking Reslizumab [106]. In comparison of severity, patients with late-onset eosinophilic asthma were noted to have a greater response to Reslizumab in reductions of exacerbation and improvement to lung function than those with early-onset asthma [91, 107].

Benralizumab, is an anti-IL5-receptor cytolytic mAb that binds directly to the IL-5 receptors on eosinophils thus enabling the immune system to remove them [108]. It is a humanized IgG1k, afucosylated mAb, (lacking oligosaccharides in the Fc region) which works by inducing apoptosis in target cells via antibody-dependent cellular cytotoxicity (ADCC) (Figure 3). In comparison to the other anti -IL-5 biologics, it has the most efficiency in the depletion of eosinophil in peripheral blood [96]. Others have observed clinically and statistically significant reduction in asthma exacerbations with the addition of Benralizumab with current treatment protocols. Some patients were able to discontinue systemic steroid therapy and became exacerbation-free by this adjunct therapy [108, 109]. However, studies have shown bronchial eosinophils remained after treatment with Benralizumab [85]. Investigators have shown that there exists a subset of lung-resident eosinophils with regulatory functions which are different from the inflammatory eosinophils that participate in the Th2 responses [110, 111, 112]. These lung resident homeostatic eosinophils are IL5 independent while the inflammatory eosinophils are IL5 dependent [107]. Indeed, IL5 deficient mice have reduced level of basal eosinophils and are unable to produce an eosinophilic inflammation as in the context of the Th2 response [110]. Therefore, the eosinophils in the bronchial tree that remained after treatment with anti-IL5 biologics may be the protective homeostasis eosinophils as those remaining may be responsible for the lack of increased risk of infection after treatment [113]. Overall, the therapeutic effect of anti-IL5 demonstrates that IL5 is the major cytokine responsible for the activation and survival of eosinophils [114]. Presently, the GINA recommendations for anti IL5 therapy are on the final Stepwise (step 5) approach to the management of severe asthma [21].

Beside targeting IL5 and its receptor for the treatment of severe eosinophilic asthma, other T2 cytokines have been targeted successfully and are currently approved. Dupilumab is a fully human mAb that blocks the alpha receptor subunit of IL-4, blocking both IL-4 and IL-13 since they share a common subunit [115]. The first clinical trial involved 52 patients with severe eosinophil asthma who were given dupilumab at a dose of 300 mg SC while 52 with similar baseline characteristic were given a placebo. The results showed that only 6% of participants from the dupilumab group continued to have asthma exacerbation versus 44% in the placebo group. This study was a 12-week provocative method where long-acting β2 agonist (LABA) was discontinued at week 4 and ICS at weeks 6–9. The results showed an 87% reduction in asthma exacerbation, improved lung function and Th2 inflammatory markers when compared to the placebo [116]. Several other studies have concluded that Dupilumab was effective in decreasing asthma exacerbation, improving lung function and quality of life in patients with asthma [117, 118].

Omalizumab, an anti-IgE mAb, downregulates IgE receptors by inhibiting IgE binding to mast cells, dendritic cells and basophils which results in the inhibition of IgE mediated inflammation. Consequently, Omalizumab treatment decreased eosinophils and multiple inflammatory mediators. It is one of the biologics currently being utilized for the treatment of moderate to severe allergic asthma with a positive treatment response [119].

Tezepelumab is a human mAb that binds to TSLP. In a randomized, double blind, placebo-controlled 52 weeks trial in patients with uncontrolled asthma who were on LABA and medium to high dose ICS, 3 doses of Tezepelumab were compared to placebo [31]. Asthma exacerbation rates were significantly reduced irrespective of the baseline eosinophil blood count or despite the dose (low, medium, or high) given. However, health related quality of life was only noted to be improved in the high dose group [31]. It has been FDA approved for children ages 12 years and older and adults with severe eosinophilic asthma.

Advertisement

7. Concluding remarks

The use of mAb to treat severe eosinophilic asthma has been effective as observed in clinical trials and real-world clinical efficacy studies [101, 102, 103, 120, 121]. Biologic therapy as an adjunct treatment has decreased the number of asthma exacerbations, reported symptom days, and improved lung function. Across the studies these improvements have been particularly notable in those with the highest blood eosinophil counts when anti-IL5 biologics were compared to omalizumab and dupilumab [118, 122, 123, 124]. This treatment works for asthmatics with severe eosinophilia [118]. However, the benefits of biologics in moderate asthma are still an ongoing area of study.

The main limitations of biologics in asthma treatments are their high cost and the tedious ongoing research into the pathogenesis of the subtypes of severe asthma [107]. The variations in endotypes pose challenges to its application in various patient populations. The utilization of biomarkers such as IgE, eosinophil count and FeNO have been shown to be beneficial for identification of endotypes especially for severe eosinophil asthmatic patients at increased risk of exacerbations [89, 119, 125, 126]. However, their identification and treatment can also be challenging due to the varying amounts of overlap of eosinophilic asthma with other endotypes particularly T2 and allergic asthma. Biomarkers such as eosinophil counts can be misleading because environmental factors can cause natural fluctuation in baseline count as observed in some healthy individuals. For example, healthy individuals exposed to tobacco smoke have a significantly higher median eosinophil count compared to nonsmokers making it difficult to determine the necessity for the use of biologics as add on in treatment [17]. Also, race should be considered as a factor when implementing treatment for vulnerable demographics. African Americans are at a higher risk for morbidity and mortality from severe asthma and studies show that those on ICS exhibit higher levels of eosinophilic inflammation than their white counterparts [127]. Finally, early identification of at-risk individuals can be instrumental in the prevention and treatment of severe eosinophilic asthma as it would potentially reduce the need for healthcare resources as medicine becomes more personalized.

Advertisement

Acknowledgments

This research was supported in part by HCA Healthcare and/or an HCA Healthcare affiliated entity.

The views expressed in this publication represent those of the author(s) and do not necessarily represent the official views of HCA Healthcare or any of its affiliated entities. This project has received funding from the US National Science Foundation (NSF) Research and Innovation program under the Grant Agreement no. 2000433. The authors wish to acknowledge the contributions of Annik T. Segree and Bethnay M. Simmons in the preparation of this chapter.

References

  1. 1. Sinyor B, Concepcion Perez L. Pathophysiology of asthma. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2023. [Copyright in 2024]
  2. 2. Holgate ST, Wenzel S, Postma DS, et al. Asthma. Nature Reviews. Disease Primers. 2015;1:15025. DOI: 10.1038/nrdp.2015.25
  3. 3. McCracken JL, Veeranki SP, Ameredes BT, Calhoun WJ. Diagnosis and management of asthma in adults: A review. Journal of the American Medical Association. 2017;318(3):279-290. DOI: 10.1001/jama.2017.8372
  4. 4. Gour N, Wills-Karp M. IL-4 and IL-13 signaling in allergic airway disease. Cytokine. 2015;75:68-78. DOI: 10.1016/j.cyto.2015.05.014
  5. 5. Gans MD, Gavrilova T. Understanding the immunology of asthma: Pathophysiology, biomarkers, and treatments for asthma endotypes. Paediatric Respiratory Reviews. 2020;36:118-127. DOI: 10.1016/j.prrv.2019.08.002
  6. 6. Nunes C, Pereira AM, Morais-Almeida M. Asthma costs and social impact. Asthma Research and Practice. 2017;3:1. DOI: 10.1186/s40733-016-0029-3
  7. 7. Daya M, Barnes KC. African American ancestry contribution to asthma and atopic dermatitis. Annals of Allergy, Asthma and Immunology. 2019;122:456-462. DOI: 10.1016/j.anai.2019.02.009
  8. 8. Bateman ED, Hurd SS, Barnes PJ, et al. Global strategy for asthma management and prevention: GINA executive summary. The European Respiratory Journal. 2008;31(1):143-178. DOI: 10.1183/09031936.00138707
  9. 9. Serebrisky D, Wiznia A. Pediatric asthma: A global epidemic. Annals of Global Health. 2019;85(1):6. DOI: 10.5334/aogh.2416
  10. 10. Wenzel SE. Asthma phenotypes: The evolution from clinical to molecular approaches. Nature Medicine. 2012;18:716-725. DOI: 10.1038/nm.2678
  11. 11. Lötvall J, Akdis CA, Bacharier LB, et al. Asthma endotypes: A new approach to classification of disease entities within the asthma syndrome. The Journal of Allergy and Clinical Immunology. 2011;127(2):355-360. DOI: 10.1016/j.jaci.2010.11.037
  12. 12. Backman H, Räisänen P, Hedman L, et al. Increased prevalence of allergic asthma from 1996 to 2006 and further to 2016-results from three population surveys. Clinical and Experimental Allergy. 2017;47:1426-1435. DOI: 10.1111/cea.12963
  13. 13. Chen M, Shepard K 2nd, Ming Yang M, et al. Overlap of allergic, eosinophilic and type 2 inflammatory subtypes in moderate-to-severe asthma. Clinical and Experimental Allergy. 2021;51:546-555. DOI: 10.1111/cea.13790
  14. 14. Tan LD, Bratt JM, Godor D, et al. Benralizumab: A unique IL-5 inhibitor for severe asthma. Journal of Asthma and Allergy. 2016;9:71-81. DOI: 10.2147/JAA.S78049
  15. 15. Adamiec A, Cieślik M, Maczka K, et al. A systematic review and meta-analysis on absolute eosinophil counts and the risk of asthma in preschool children with wheezing: An EAACI task force report. Pediatric Allergy and Immunology. 2024;35:e14078. DOI: 10.1111/pai.14078
  16. 16. Li H, Li X. Genetic relationships between high blood eosinophil count, asthma susceptibility, and asthma severity. The Journal of Asthma. 2024;61:119-131. DOI: 10.1080/02770903.2023.2247490
  17. 17. Lommatzsch M, Nair P, Virchow JC. Normal blood eosinophil counts in humans. Respiration. 2024;103(4):214-216. DOI: 10.1159/000537833
  18. 18. Seymour BW, Schelegle ES, Pinkerton KE, et al. Second-hand smoke increases bronchial hyperreactivity and eosinophilia in a murine model of allergic aspergillosis. Clinical and Developmental Immunology. 2003;10:35-42. DOI: 10.1080/10446670310001598483
  19. 19. Bochner BS, Gleich GJ. What targeting eosinophils has taught us about their role in diseases. The Journal of Allergy and Clinical Immunology. 2010;126:16-25. DOI: 10.1016/j.jaci.2010.02.026
  20. 20. Bousquet J, Chanez P, Lacoste JY, et al. Eosinophilic inflammation in asthma. The New England Journal of Medicine. 1990;323:1033-1039. DOI: 10.1056/NEJM199010113231505
  21. 21. Levy ML, Bacharier LB, Bateman E, et al. Key recommendations for primary care from the 2022 global initiative for asthma (GINA) update. NPJ Primary Care Respiratory Medicine. 2023;33(1):7. DOI: 10.1038/s41533-023-00330-1
  22. 22. Agache I, Akdis CA, Akdis M, et al. EAACI biologicals guidelines-recommendations for severe asthma. Allergy. 2021;76:14-44. DOI: 10.1111/all.14425
  23. 23. Tran TN, Zeiger RS, Peters SP, et al. Overlap of atopic, eosinophilic, and TH2-high asthma phenotypes in a general population with current asthma. Annals of Allergy, Asthma and Immunology. 2016;116:37-42. DOI: 10.1016/j.anai.2015.10.027
  24. 24. Pavlidis S, Takahashi K, Kwong FNK, et al. “T2-high” in severe asthma related to blood eosinophil, exhaled nitric oxide and serum periostin. The European Respiratory Journal. 2019;53(1):1800938. DOI: 10.1183/13993003.00938-2018
  25. 25. Del Pozo V, Bobolea I, Rial MJ, et al. Expert consensus on the use of systemic glucocorticoids for managing eosinophil-related diseases. Frontiers in Immunology. 2024;14:1310211. DOI: 10.3389/fimmu.2023.1310211
  26. 26. Pelaia C, Paoletti G, Puggioni F, et al. Interleukin-5 in the pathophysiology of severe asthma. Frontiers in Physiology. 2019;10:1514. DOI: 10.3389/fphys.2019.01514
  27. 27. Stirling RG, van Rensen EL, Barnes PJ, Chung KF. Interleukin-5 induces CD34(+) eosinophil progenitor mobilization and eosinophil CCR3 expression in asthma. American Journal of Respiratory and Critical Care Medicine. 2001;164(8 Pt. 1):1403-1409. DOI: 10.1164/ajrccm.164.8.2010002
  28. 28. Fulkerson PC, Rothenberg ME. Targeting eosinophils in allergy, inflammation and beyond. Nature Reviews. Drug Discovery. 2013;12:117-129. DOI: 10.1038/nrd3838
  29. 29. Dunican EM, Fahy JV. Asthma and corticosteroids: Time for a more precise approach to treatment. The European Respiratory Journal. 2017;49:1701167. DOI: 10.1183/13993003.01167-2017
  30. 30. Agache I, Beltran J, Akdis C, et al. Efficacy and safety of treatment with biologicals (benralizumab, dupilumab, mepolizumab, omalizumab and reslizumab) for severe eosinophilic asthma. A systematic review for the EAACI guidelines—Recommendations on the use of biologicals in severe asthma. Allergy. 2020;75(5):1023-1042. DOI: 10.1111/all.14221
  31. 31. Bakakos A, Loukides S, Bakakos P. Severe eosinophilic asthma. Journal of Clinical Medicine. 2019;8:1375. DOI: 10.3390/jcm8091375
  32. 32. Kay AB, Phipps S, Robinson DS. A role for eosinophils in airway remodelling in asthma. Trends in Immunology. 2004;25:477-482. DOI: 10.1016/j.it.2004.07.006
  33. 33. Cho JY, Miller M, Baek KJ, et al. Inhibition of airway remodeling in IL-5-deficient mice. The Journal of Clinical Investigation. 2004;113:551-560. DOI: 10.1172/JCI1913
  34. 34. Tanaka H, Komai M, Nagao K, et al. Role of interleukin-5 and eosinophils in allergen-induced airway remodeling in mice. American Journal of Respiratory Cell and Molecular Biology. 2004;31:62-68. DOI: 10.1165/rcmb.2003-0305OC
  35. 35. Isakson PC, Puré E, Vitetta ES, Krammer PH. T cell-derived B cell differentiation factor(s). Effect on the isotype switch of murine B cells. The Journal of Experimental Medicine. 1982;155:734-748. DOI: 10.1084/jem.155.3.734
  36. 36. Vitetta ES, Ohara J, Myers CD, et al. Serological, biochemical, and functional identity of B cell-stimulatory factor 1 and B cell differentiation factor for IgG1. The Journal of Experimental Medicine. 1985;162:1726-1731. DOI: 10.1084/jem.162.5.1726
  37. 37. Coffman RL, Carty J. A T cell activity that enhances polyclonal IgE production and its inhibition by interferon-gamma. Journal of Immunology. 1986;136:949-954
  38. 38. Mosmann TR, Cherwinski H, Bond MW, et al. Two types of murine helper T cell clone. I. Definition according to profiles of lymphokine activities and secreted proteins. Journal of Immunology. 1986;136(7):2348-2357
  39. 39. Coffman RL, Seymour BWP, Lebman D, et al. The role of helper T cells product in mouse B cells differentiation and isotype regulation. Immunological Reviews. 1988;102:5-28. DOI: 10.1111/j.1600-065x.1988.tb00739.x
  40. 40. Sanderson CJ, Warren DJ, Strath M. Identification of a lymphokine that stimulates eosinophil differentiation in vitro. Its relationship to interleukin 3, and functional properties of eosinophils produced in cultures. The Journal of Experimental Medicine. 1985;162(1):60-74. DOI: 10.1084/jem.162.1.60
  41. 41. Campbell HD, Tucker WQ , Hort Y, et al. Molecular cloning, nucleotide sequence, and expression of the gene encoding human eosinophil differentiation factor (interleukin 5). Proceedings of the National Academy of Sciences of the United States of America. 1987;84(19):6629-6633. DOI: 10.1073/pnas.84.19.6629
  42. 42. Clutterbuck E, Shields JG, Gordon J, et al. Recombinant human interleukin 5 is an eosinophil differentiation factor but has no activity in standard human B cell growth factor assay. European Journal of Immunology. 1987;17:1743-1750. DOI: 10.1002/eji.1830171210
  43. 43. Coffman RL, Seymour BW, Hudak S, et al. Antibody to interleukin-5 inhibits helminth-induced eosinophilia in mice. Science. 1989;245:308-310. DOI: 10.1126/science.2787531
  44. 44. Rennick DM, Thompson-Snipes L, Coffman RL, et al. In vivo administration of antibody to interleukin-5 inhibits increased generation of eosinophils and their progenitors in bone marrow of parasitized mice. Blood. 1990;76:312-316
  45. 45. Foster PS, Hogan SP, Ramsay AJ, et al. Interleukin 5 deficiency abolishes eosinophilia, airways hyperreactivity, and lung damage in a mouse asthma model. The Journal of Experimental Medicine. 1996;183(1):195-201. DOI: 10.1084/jem.183.1.195
  46. 46. Kurup VP, Choi H, Murali PS, Coffman RL. IgE and eosinophil regulation in a murine model of allergic aspergillosis. Journal of Leukocyte Biology. 1994;56(5):593-598. DOI: 10.1002/jlb.56.5.593
  47. 47. Savelkoul HFJ, Seymour BWP, Sullivan L, Coffman RL. IL-4 can correct the defective Ig-E production of SJA/9 mice. Journal of Immunology. 1991;146:1801-1805
  48. 48. Herzenberg LA, Okumura K, Cantor H, et al. T-cell regulation of antibody responses: Demonstration of allotype-specific helper T cells and their specific removal by suppressor T cells. The Journal of Experimental Medicine. 1976;144(2):330-344. DOI: 10.1084/jem.144.2.330
  49. 49. Severinson E. Identification of the IgG1 induction factor (interleukin 4). Frontiers in Immunology. 2014;5:628. DOI: 10.3389/fimmu.2014.00628
  50. 50. Seymour BWP. The effect of environmental tobacco smoke on murine models of allergy [thesis]. Davis: University of California; 1996
  51. 51. Imraish A, Abu-Thiab T, Zihlif M. IL-13 and FOXO3 genes polymorphisms regulate IgE levels in asthmatic patients. Biomedical Reports. 2021;14(6):55. DOI: 10.3892/br.2021.1431
  52. 52. Soumelis V, Reche PA, Kanzler H, et al. Human epithelial cells trigger dendritic cell mediated allergic inflammation by producing TSLP. Nature Immunology. 2002;3(7):673-680. DOI: 10.1038/ni805
  53. 53. Oboki K, Nakae S, Matsumoto K, Saito H. IL-33 and airway inflammation. Allergy, Asthma and Immunology Research. 2011;3(2):81-88. DOI: 10.4168/aair.2011.3.2.81
  54. 54. Shrestha SK, Katelaris C, Dharmage SC, et al. High ambient levels of grass, weed and other pollen are associated with asthma admissions in children and adolescents: A large 5-year case-crossover study. Clinical and Experimental Allergy. 2018;48:1421-1428. DOI: 10.1111/cea.13225
  55. 55. Gauvreau GM, Bergeron C, Boulet L-P, et al. Sounding the alarmins-the role of alarmin cytokines in asthma. Allergy. 2023;78(2):402-417. DOI: 10.1111/all.15609
  56. 56. Bradding P, Porsbjerg C, Côté A, et al. Airway hyperresponsiveness in asthma: The role of the epithelium. The Journal of Allergy and Clinical Immunology. 2024;153(5):1181-1193. DOI: 10.1016/j.jaci.2024.02.011
  57. 57. Hurst SD, Muchamuel T, Gorman DM, et al. New IL-17 family members promote Th1 or Th2 responses in the lung: In vivo function of the novel cytokine IL-25. Journal of Immunology. 2002;169:443-453. DOI: 10.4049/jimmunol.169.1.443
  58. 58. Yao X, Liu X, Wang X, Zhang L. IL-25R+ circulating fibrocytes are increased in asthma and correlate with fixed airflow limitation. The Clinical Respiratory Journal. 2021;15(11):1248-1256. DOI: 10.1111/crj.13433
  59. 59. Yao X, Chen Q , Wang X, et al. IL-25 induces airway remodeling in asthma by orchestrating the phenotypic changes of epithelial cell and fibrocyte. Respiratory Research. 2023;24(1):212. DOI: 10.1186/s12931-023-02509-z
  60. 60. Besnard A-G, Togbe D, Guillou N, et al. IL-33-activated dendritic cells are critical for allergic airway inflammation. European Journal of Immunology. 2011;41(6):1675-1686. DOI: 10.1002/eji.201041033
  61. 61. Kouzaki H, O'Grady SM, Lawrence CB, Kita H. Proteases induce production of thymic stromal lymphopoietin by airway epithelial cells through protease-activated receptor-2. Journal of Immunology. 2009;183(2):1427-1434. DOI: 10.4049/jimmunol.0900904
  62. 62. Theofani E, Tsitsopoulou A, Morianos I, Semitekolou M. Severe asthmatic responses: The impact of TSLP. International Journal of Molecular Sciences. 2023;24(8):7581. DOI: 10.3390/ijms24087581
  63. 63. He R, Geha RS. Thymic stromal lymphopoietin. Annals of the New York Academy of Sciences. 2010;1183:13-24. DOI: 10.1111/j.1749-6632.2009.05128.x
  64. 64. Cianferoni A, Spergel J. The importance of TSLP in allergic disease and its role as a potential therapeutic target. Expert Review of Clinical Immunology. 2014;10(11):1463-1474. DOI: 10.1586/1744666X.2014.967684
  65. 65. Andreasson LM, Dyhre-Petersen N, Hvidtfeldt M, et al. Airway hyperresponsiveness correlates with airway TSLP in asthma independent of eosinophilic inflammation. The Journal of Allergy and Clinical Immunology. 2024;153(4):988-997.e11. DOI: 10.1016/j.jaci.2023.11.915
  66. 66. Salter BM, Aw M, Sehmi R. The role of type 2 innate lymphoid cells in eosinophilic asthma. Journal of Leukocyte Biology. 2019;106:889-901. DOI: 10.1002/JLB.3MR1217-497R
  67. 67. Kato A. Group 2 innate lymphoid cells in airway diseases. Chest. 2019;156(1):141-149. DOI: 10.1016/j.chest.2019.04.101
  68. 68. Wallrapp A, Riesenfeld SJ, Burkett PR, et al. The neuropeptide NMU amplifies ILC2-driven allergic lung inflammation. Nature. 2017;549(7672):351-356. DOI: 10.1038/nature24029
  69. 69. Mjösberg J, Bernink J, Golebski K, et al. The transcription factor GATA3 is essential for the function of human type 2 innate lymphoid cells. Immunity. 2012;37(4):649-659. DOI: 10.1016/j.immuni.2012.08.015
  70. 70. Halim TYF, Steer CA, Mathä L, et al. Group 2 innate lymphoid cells are critical for the initiation of adaptive T helper 2 cell-mediated allergic lung inflammation. Immunity. 2014;40(3):425-435. DOI: 10.1016/j.immuni.2014.01.011
  71. 71. Smith SG, Chen R, Kjarsgaard M, et al. Increased numbers of activated group 2 innate lymphoid cells in the airways of patients with severe asthma and persistent airway eosinophilia. The Journal of Allergy and Clinical Immunology. 2016;137(1):75-86.e8. DOI: 10.1016/j.jaci.2015.05.037
  72. 72. Shakoory B, Fitzgerald SM, Lee SA, et al. The role of human mast cell-derived cytokines in eosinophil biology. Journal of Interferon and Cytokine Research. 2004;24(5):271-281. DOI: 10.1089/107999004323065057
  73. 73. Sakuishi K, Oki S, Araki M. Invariant NKT cells biased for IL-5 production act as crucial regulators of inflammation. Journal of Immunology. 2007;179(6):3452-3462. DOI: 10.4049/jimmunol.179.6.3452
  74. 74. Yanagibashi T, Satoh M, Nagai Y, et al. Allergic diseases: From bench to clinic—Contribution of the discovery of interleukin-5. Cytokine. 2017;98:59-70. DOI: 10.1016/j.cyto.2016.11.011
  75. 75. Gutierrez-Ramos JC, Lloyd C, Gonzalo JA. Eotaxin: From an eosinophilic chemokine to a major regulator of allergic reactions. Immunology Today. 1999;20(11):500-504. DOI: 10.1016/s0167-5699(99)01522-4
  76. 76. Joubert P, Lajoie-Kadoch S, Labonté I, et al. CCR3 expression and function in asthmatic airway smooth muscle cells. Journal of Immunology. 2005;175(4):2702-2708. DOI: 10.4049/jimmunol.175.4.2702
  77. 77. Galdiero MR, Varricchi G, Seaf M, et al. Bidirectional mast cell-eosinophil interactions in inflammatory disorders and cancer. Frontiers in Medicine. 2017;4:103. DOI: 10.3389/fmed.2017.00103
  78. 78. He K, Hettinga A, Kale SL, et al. Blimp-1 is essential for allergen-induced asthma and Th2 cell development in the lung. The Journal of Experimental Medicine. 2020;217:e20190742. DOI: 10.1084/jem.20190742
  79. 79. Petsky HL, Cates CJ, Kew KM, Chang AB. Tailoring asthma treatment on eosinophilic markers (exhaled nitric oxide or sputum eosinophils): A systematic review and meta-analysis. Thorax. 2018;73(12):1110-1119. DOI: 10.1136/thoraxjnl-2018-211540
  80. 80. Vermeer PD, Harson R, Einwalter LA, et al. Interleukin-9 induces goblet cell hyperplasia during repair of human airway epithelia. American Journal of Respiratory Cell and Molecular Biology. 2003;28(3):286-295. DOI: 10.1165/rcmb.4887
  81. 81. Venegas Garrido C, Mukherjee M, Svenningsen S, Nair P. Eosinophil-mucus interplay in severe asthma: Implications for treatment with biologicals. Allergology International. 2024;73(3):351-361. DOI: 10.1016/j.alit.2024.03.001
  82. 82. Zustakova M, Kratochvilova L, Slama P. Apoptosis of eosinophil granulocytes. Biology (Basel). 2020;9:457. DOI: 10.3390/biology9120457
  83. 83. Bajbouj K, AbuJabal R, Sahnoon L, et al. IL-5 receptor expression in lung fibroblasts: Potential role in airway remodeling in asthma. Allergy. 2023;78:882-885. DOI: 10.1111/all.15627
  84. 84. Maglio A, Vitale C, Pelaia C, et al. Severe asthma remissions induced by biologics targeting IL5/IL5r: Results from a multicenter real-life study. International Journal of Molecular Sciences. 2023;24:2455. DOI: 10.3390/ijms24032455
  85. 85. Koranteng J, Chung KF, Michaeloudes C, Bhavsar P. The role of mitochondria in eosinophil function: Implications for severe asthma pathogenesis. Frontiers in Cell and Development Biology. 2024;12:1360079. DOI: 10.3389/fcell.2024.1360079
  86. 86. Mauser PJ, Pitman AM, Fernandez X, et al. Effects of an antibody to interleukin-5 in a monkey model of asthma. American Journal of Respiratory and Critical Care Medicine. 1995;152(2):467-472. DOI: 10.1164/ajrccm.152.2.7633694
  87. 87. Garlisi CG, Kung TT, Wang P, et al. Effects of chronic anti-interleukin-5 monoclonal antibody treatment in a murine model of pulmonary inflammation. American Journal of Respiratory Cell and Molecular Biology. 1999;20(2):248-255. DOI: 10.1165/ajrcmb.20.2.3327
  88. 88. Pavord ID, Bel EH, Bourdin A, et al. From DREAM to REALITI-A and beyond: Mepolizumab for the treatment of eosinophil-driven diseases. Allergy. 2022;77(3):778-797. DOI: 10.1111/all.15056
  89. 89. Yancey SW, Keene ON, Albers FC, et al. Biomarkers for severe eosinophilic asthma. The Journal of Allergy and Clinical Immunology. 2017;140:1509-1518. DOI: 10.1016/j.jaci.2017.10.005
  90. 90. Pelaia C, Calabrese C, Vatrella A, et al. Benralizumab: From the basic mechanism of action to the potential use in the biological therapy of severe eosinophilic asthma. BioMed Research International. 2018;2018:4839230. DOI: 10.1155/2018/4839230
  91. 91. Walsh GM. Reslizumab in the treatment of severe eosinophilic asthma: An update. Immunotherapy. 2018;10(8):695-698. DOI: 10.2217/imt-2017-0176
  92. 92. Flood-Page PT, Menzies-Gow AN, Kay AB, Robinson DS. Eosinophil role remains uncertain as anti-interleukin-5 only partially depletes numbers in asthmatic airways. American Journal of Respiratory and Critical Care Medicine. 2003;167:199-204. DOI: 10.1164/rccm.200208-789OC
  93. 93. Pelaia C, Vatrella A, Busceti MT, et al. Severe eosinophilic asthma: From the pathogenic role of interleukin-5 to the therapeutic action of mepolizumab. Drug Design, Development and Therapy. 2017;11:3137-3144. DOI: 10.2147/DDDT.S150656
  94. 94. Abonia JP, Putnam PE. Mepolizumab in eosinophilic disorders. Expert Review of Clinical Immunology. 2011;7(4):411-417. DOI: 10.1586/eci.11.27
  95. 95. Kusano S, Kukimoto-Niino M, Hino N, et al. Structural basis of interleukin-5 dimer recognition by its α receptor. Protein Science. 2012;21(6):850-864. DOI: 10.1002/pro.2072
  96. 96. Mukherjee M, Sehmi R, Nair P. Anti-IL5 therapy for asthma and beyond. World Allergy Organization Journal. 2014;7(1):32. DOI: 10.1186/1939-4551-7-32. eCollection 2014
  97. 97. Leckie MJ, Brinke AT, Khan J, et al. Effects of an interleukin-5 blocking monoclonal antibody on eosinophils, airway hyper-responsiveness, and the late asthmatic response. Lancet. 2000;356(9248):2144-2148. DOI: 10.1016/s0140-6736(00)03496-6
  98. 98. Kips JC, O'Connor BJ, Langley SJ, et al. Effect of SCH55700, a humanized anti-human interleukin-5 antibody, in severe persistent asthma: A pilot study. American Journal of Respiratory and Critical Care Medicine. 2003;167(12):1655-1659. DOI: 10.1164/rccm.200206-525OC
  99. 99. Haldar P, Brightling CE, Hargadon B, et al. Mepolizumab and exacerbations of refractory eosinophilic asthma. The New England Journal of Medicine. 2009;360(10):973-984. DOI: 10.1056/NEJMoa0808991
  100. 100. Pavord ID, Korn S, Howarth P, et al. Mepolizumab for severe eosinophilic asthma (DREAM): A multicentre, double-blind, placebo-controlled trial. Lancet. 2012;380(9842):651-659. DOI: 10.1016/S0140-6736(12)60988-X
  101. 101. Ortega HG, Liu MC, Pavord ID, et al. Mepolizumab treatment in patients with severe eosinophilic asthma. The New England Journal of Medicine. 2014;371(13):1198-1207. DOI: 10.1056/NEJMoa1403290
  102. 102. Bergantini L, Pianigiani T, d'Alessandro M, et al. The effect of anti-IL5 monoclonal antibodies on regulatory and effector T cells in severe eosinophilic asthma. Biomedicine and Pharmacotherapy. 2023;166:115385. DOI: 10.1016/j.biopha.2023.115385
  103. 103. Halwani R, Al-Muhsen S, Al-Jahdali H, Hamid Q. Role of transforming growth factor-β in airway remodeling in asthma. American Journal of Respiratory Cell and Molecular Biology. 2011;44(2):127-133. DOI: 10.1165/rcmb.2010-0027TR
  104. 104. Crimi C, Campisi R, Nolasco S, et al. Mepolizumab effectiveness in patients with severe eosinophilic asthma and co-presence of bronchiectasis: A real-world retrospective pilot study. Respiratory Medicine. 2021;185:106491. DOI: 10.1016/j.rmed.2021.106491
  105. 105. Deeks ED, Brusselle G. Reslizumab in eosinophilic asthma: A review. Drugs. 2017;77(7):777-784. DOI: 10.1007/s40265-017-0740-2
  106. 106. Ibrahim H, O'Sullivan R, Casey D, et al. The effectiveness of reslizumab in severe asthma treatment: A real-world experience. Respiratory Research. 2019;20(1):289. DOI: 10.1186/s12931-019-1251-3
  107. 107. Lam J, Hay JW, Salcedo J, Kenyon NJ. A cost-effectiveness analysis of reslizumab in the treatment of poorly controlled eosinophilic asthma. The Journal of Asthma. 2019;56(8):872-881. DOI: 10.1080/02770903.2018.1500584
  108. 108. Kavanagh JE, Hearn AP, Dhariwal J, et al. Real-world effectiveness of benralizumab in severe eosinophilic asthma. Chest. 2021;159(2):496-506. DOI: 10.1016/j.chest.2020.08.2083
  109. 109. Pham TH, Damera G, Newbold P, Ranade K. Reductions in eosinophil biomarkers by benralizumab in patients with asthma. Respiratory Medicine. 2016;111:21-29. DOI: 10.1016/j.rmed.2016.01.003
  110. 110. Marichal T, Mesnil C, Bureau F. Homeostatic eosinophils: Characteristics and functions. Frontiers in Medicine (Lausanne). 2017;4:101. DOI: 10.3389/fmed.2017.00101
  111. 111. Van Hulst G, Jorssen J, Jacobs N, et al. Anti-IL5 mepolizumab minimally influences residual blood eosinophils in severe asthma. The European Respiratory Journal. 2022;59(3):2100935. DOI: 10.1183/13993003.00935-2021
  112. 112. Mesnil C, Raulier S, Paulissen G, et al. Lung-resident eosinophils represent a distinct regulatory eosinophil subset. The Journal of Clinical Investigation. 2016;126(9):3279-3295. DOI: 10.1172/JCI85664
  113. 113. Jackson DJ, Korn S, Mathur SK, et al. Safety of eosinophil-depleting therapy for severe, eosinophilic asthma: Focus on benralizumab. Drug Safety. 2020;43(5):409-425. DOI: 10.1007/s40264-020-00926-3
  114. 114. Luo J, Chen W, Liu W, et al. IL-5 antagonism reverses priming and activation of eosinophils in severe eosinophilic asthma. Mucosal Immunology. 2024;S1933-0219(24):00024-00022. DOI: 10.1016/j.mucimm.2024.03.005
  115. 115. McCormick SM, Heller NM. Commentary: IL-4 and IL-13 receptors and signaling. Cytokine. 2015;75(1):38-50. DOI: 10.1016/j.cyto.2015.05.023
  116. 116. Wenzel S, Ford L, Pearlman D, et al. Dupilumab in persistent asthma with elevated eosinophil levels. The New England Journal of Medicine. 2013;368:2455-2466. DOI: 10.1056/NEJMoa1304048
  117. 117. Xiong XF, Zhu M, Wu HX, et al. Efficacy and safety of dupilumab for the treatment of uncontrolled asthma: A meta-analysis of randomized clinical trials. Respiratory Research. 2019;20(1):108. DOI: 10.1186/s12931-019-1065-3
  118. 118. Bacharier LB, Guilbert TW, Katelaris CH, et al. Dupilumab improves lung function parameters in pediatric type 2 asthma: VOYAGE study. The Journal of Allergy and Clinical Immunology. In Practice. 2024;12(4):948-959. DOI: 10.1016/j.jaip.2023.12.006
  119. 119. Casale TB, Luskin AT, Busse W, et al. Omalizumab effectiveness by biomarker status in patients with asthma: Evidence from PROSPERO, a prospective real-world study. The Journal of Allergy and Clinical Immunology. In Practice. 2019;7(1):156-164.e1. DOI: 10.1016/j.jaip.2018.04.043
  120. 120. Wechsler ME, Munitz A, Ackerman SJ, et al. Eosinophils in health and disease: A state-of-the-art review. Mayo Clinic Proceedings. 2021;96(10):2694-2707. DOI: 10.1016/j.mayocp.2021.04.025
  121. 121. Rojo-Tolosa S, González-Gutiérrez MV, Jiménez-Gálvez G, et al. Impact of anti-IL5 therapies on patients with severe uncontrolled asthma and possible predictive biomarkers of response: A real-life study. International Journal of Molecular Sciences. 2023;24:2011. DOI: 10.3390/ijms24032011
  122. 122. Kimura Y, Suzukawa M, Inoue N, et al. Differences in asthma-related outcomes by anti-IL-5 biologics, omalizumab, and dupilumab based on blood eosinophil counts. Asian Pacific Journal of Allergy and Immunology. 2024. DOI: 10.12932/AP-290623-1645 [Epub ahead of print]
  123. 123. Pfeffer PE, Ali N, Murray R, et al. Comparative effectiveness of anti-IL5 and anti-IgE biologic classes in patients with severe asthma eligible for both. Allergy. 2023;78:1934-1948. DOI: 10.1111/all.15711
  124. 124. Charles D, Shanley J, Temple SN, et al. Real-world efficacy of treatment with benralizumab, dupilumab, mepolizumab and reslizumab for severe asthma: A systematic review and meta-analysis. Clinical and Experimental Allergy. 2022;52:616-627. DOI: 10.1111/cea.14112
  125. 125. Bourdin A, Brusselle G, Couillard S, et al. Phenotyping of severe asthma in the era of broad-acting anti-asthma biologics. The Journal of Allergy and Clinical Immunology. In Practice. 2024;12(4):809-823. DOI: 10.1016/j.jaip.2024.01.023
  126. 126. Oppenheimer J, Hoyte FCL, Phipatanakul W, et al. Allergic and eosinophilic asthma in the era of biomarkers and biologics: Similarities, differences and misconceptions. Annals of Allergy, Asthma and Immunology. 2022;129(2):169-180. DOI: 10.1016/j.anai.2022.02.021
  127. 127. Nyenhuis SM, Krishnan JA, Berry A, et al. Race is associated with differences in airway inflammation in patients with asthma. The Journal of Allergy and Clinical Immunology. 2017;140(1):257-265.e11. DOI: 10.1016/j.jaci.2016.10.024

Written By

Brian W.P. Seymour, Ashley A. Seymour and Patricia M. Gilford

Submitted: 07 June 2024 Reviewed: 07 June 2024 Published: 23 August 2024