Eosinophils as Drivers of Severe Eosinophilic Asthma: Endotypes or Plasticity?

Asthma is now recognized as a heterogeneous disease, encompassing different phenotypes driven by distinct pathophysiological mechanisms called endotypes. Common phenotypes of asthma, referred to as eosinophilic asthma, are characterized by the presence of eosinophilia. Eosinophils are usually considered invariant, terminally differentiated effector cells and have become a primary therapeutic target in severe eosinophilic asthma (SEA) and other eosinophil-associated diseases (EADs). Biological treatments that target eosinophils reveal an unexpectedly complex role of eosinophils in asthma, including in SEA, suggesting that “not all eosinophils are equal”. In this review, we address our current understanding of the role of eosinophils in asthma with regard to asthma phenotypes and endotypes. We further address the possibility that different SEA phenotypes may involve differences in eosinophil biology. We discuss how these differences could arise through eosinophil “endotyping”, viz. adaptations of eosinophil function imprinted during their development, or through tissue-induced plasticity, viz. local adaptations of eosinophil function through interaction with their lung tissue niches. In doing so, we also discuss opportunities, technical challenges, and open questions that, if addressed, might provide considerable benefits in guiding the choice of the most efficient precision therapies of SEA and, by extension, other EADs.

[1]  R. Louis,et al.  Anti-IL5 mepolizumab minimally influences residual blood eosinophils in severe asthma , 2021, European Respiratory Journal.

[2]  D. Barber,et al.  Eosinophils are part of the granulocyte response in tuberculosis and promote host resistance in mice , 2021, The Journal of experimental medicine.

[3]  H. Simon,et al.  The Enigma of Eosinophil Degranulation , 2021, International journal of molecular sciences.

[4]  I. Pavord,et al.  Patient characteristics, biomarkers and exacerbation risk in severe, uncontrolled asthma , 2021, European Respiratory Journal.

[5]  N. Hacohen,et al.  Plasma from patients with bacterial sepsis or severe COVID-19 induces suppressive myeloid cell production from hematopoietic progenitors in vitro , 2021, Science Translational Medicine.

[6]  M. Idzko,et al.  Response to Mepolizumab Treatment in Patients with Severe Eosinophilic Asthma and Atopic Phenotypes , 2021, Journal of asthma and allergy.

[7]  S. Berdnikovs The twilight zone: plasticity and mixed ontogeny of neutrophil and eosinophil granulocyte subsets , 2021, Seminars in Immunopathology.

[8]  I. Pavord,et al.  The inflammatory profile of exacerbations in patients with severe refractory eosinophilic asthma receiving mepolizumab (the MEX study): a prospective observational study. , 2021, The Lancet. Respiratory medicine.

[9]  Y. Politanska,et al.  More than neutrophils: Lin(+)Ly6G(+)IL‐5Rα(+) multipotent myeloid cells (MMCs) are dominant in normal murine bone marrow and retain capacity to differentiate into eosinophils and monocytes , 2021, Journal of leukocyte biology.

[10]  J. L. Dunn,et al.  Bidirectional Crosstalk Between Eosinophils and Esophageal Epithelial Cells Regulates Inflammatory and Remodeling Processes , 2021, Mucosal Immunology.

[11]  H. Hammad,et al.  The basic immunology of asthma , 2021, Cell.

[12]  A. Nanzer,et al.  Real world effectiveness of anti-IL5/5R therapy in severe atopic eosinophilic asthma with fungal sensitisation. , 2021, The journal of allergy and clinical immunology. In practice.

[13]  I. Pavord,et al.  Eosinophil Knockout Humans: Uncovering the Role of Eosinophils Through Eosinophil-Directed Biological Therapies. , 2021, Annual review of immunology.

[14]  H. Dweep,et al.  Analysis of classical neutrophils and polymorphonuclear myeloid-derived suppressor cells in cancer patients and tumor-bearing mice , 2021, The Journal of experimental medicine.

[15]  M. Guilliams,et al.  Does tissue imprinting restrict macrophage plasticity? , 2021, Nature Immunology.

[16]  S. Ackerman,et al.  A tribute to eosinophils from a comparative and evolutionary perspective. , 2021, The Journal of allergy and clinical immunology.

[17]  G. Canonica,et al.  Onset of effect and impact on health-related quality of life, exacerbation rate, lung function, and nasal polyposis symptoms for patients with severe eosinophilic asthma treated with benralizumab (ANDHI): a randomised, controlled, phase 3b trial. , 2020, The Lancet. Respiratory medicine.

[18]  I. Pavord,et al.  Clinical Development of Mepolizumab for the Treatment of Severe Eosinophilic Asthma: On The Path to Personalized Medicine. , 2020, The journal of allergy and clinical immunology. In practice.

[19]  P. Nair,et al.  Asthma exacerbations on benralizumab are largely non‐eosinophilic , 2020, Allergy.

[20]  L. Boulet,et al.  The Management of Severe Asthma in 2020. , 2020, Biochemical pharmacology.

[21]  Yin-fang Wu,et al.  Homeostatic and early-recruited CD101− eosinophils suppress endotoxin-induced acute lung injury , 2020, European Respiratory Journal.

[22]  J. Mikes,et al.  Systems-Level Immunomonitoring from Acute to Recovery Phase of Severe COVID-19 , 2020, Cell Reports Medicine.

[23]  R. Louis,et al.  Advances toward precision medicine for asthma. , 2020, Biochemical pharmacology.

[24]  E. V. van Roon,et al.  Prediction of response to biological treatment with monoclonal antibodies in severe asthma. , 2020, Biochemical pharmacology.

[25]  R. Louis,et al.  Eosinophil diversity in asthma. , 2020, Biochemical pharmacology.

[26]  K. McCoy,et al.  The emerging roles of eosinophils in mucosal homeostasis , 2020, Mucosal Immunology.

[27]  M. Humbert,et al.  Efficacy of intravenous reslizumab in oral corticosteroid-dependent asthma. , 2020, The journal of allergy and clinical immunology. In practice.

[28]  S. Ackerman,et al.  Contributions of Eosinophils to Human Health and Disease. , 2020, Annual review of pathology.

[29]  A. Fryer,et al.  IL-5 Exposure in utero Increases Lung Nerve Density and Causes Airway Reactivity in Adult Offspring. , 2019, American journal of respiratory cell and molecular biology.

[30]  I. Pavord,et al.  Prognostic and predictive value of blood eosinophil count, fractional exhaled nitric oxide and their combination in severe asthma: a post-hoc analysis. , 2019, American journal of respiratory and critical care medicine.

[31]  F. Albers,et al.  Baseline blood eosinophil count as a predictor of treatment response to the licensed dose of mepolizumab in severe eosinophilic asthma. , 2019, Respiratory medicine.

[32]  F. Albers,et al.  Mepolizumab reduces exacerbations in patients with severe eosinophilic asthma, irrespective of body weight/body mass index: meta-analysis of MENSA and MUSCA , 2019, Respiratory Research.

[33]  G. Chupp,et al.  Phenotypes and endotypes of adult asthma: Moving toward precision medicine. , 2019, The Journal of allergy and clinical immunology.

[34]  F. Albers,et al.  Effect of mepolizumab in severe eosinophilic asthma according to omalizumab eligibility. , 2019, Respiratory medicine.

[35]  H. Hammad,et al.  Protein crystallization promotes type 2 immunity and is reversible by antibody treatment , 2019, Science.

[36]  Supat Thongjuea,et al.  Identification of two distinct pathways of human myelopoiesis , 2019, Science Immunology.

[37]  H. Ortega,et al.  Assessment of the long‐term safety of mepolizumab and durability of clinical response in patients with severe eosinophilic asthma , 2019, The Journal of allergy and clinical immunology.

[38]  C. Nerlov,et al.  Haematopoiesis in the era of advanced single-cell technologies , 2019, Nature Cell Biology.

[39]  Allon M. Klein,et al.  Lineage tracing on transcriptional landscapes links state to fate during differentiation , 2018, Science.

[40]  V. Witko-Sarsat,et al.  Expanding Neutrophil Horizons: New Concepts in Inflammation , 2018, Journal of Innate Immunity.

[41]  A. Fryer,et al.  Eosinophils increase airway sensory nerve density in mice and in human asthma , 2018, Science Translational Medicine.

[42]  E. Bleecker,et al.  Baseline patient factors impact on the clinical efficacy of benralizumab for severe asthma , 2018, European Respiratory Journal.

[43]  A. Fryer,et al.  Eosinophil and airway nerve interactions in asthma , 2018, Journal of leukocyte biology.

[44]  James J. Lee,et al.  Shaping eosinophil identity in the tissue contexts of development, homeostasis, and disease , 2018, Journal of leukocyte biology.

[45]  J. Lasky,et al.  A critical role for IL-18 in transformation and maturation of naive eosinophils to pathogenic eosinophils. , 2018, The Journal of allergy and clinical immunology.

[46]  Jessica E. Bolden,et al.  Transcriptional profiling of eosinophil subsets in interleukin‐5 transgenic mice , 2018, Journal of leukocyte biology.

[47]  B. Chipps,et al.  Benralizumab efficacy by atopy status and serum immunoglobulin E for patients with severe, uncontrolled asthma. , 2018, Annals of allergy, asthma & immunology : official publication of the American College of Allergy, Asthma, & Immunology.

[48]  F. Roufosse Targeting the Interleukin-5 Pathway for Treatment of Eosinophilic Conditions Other than Asthma , 2018, Front. Med..

[49]  Samuel L. Wolock,et al.  Population Snapshots Predict Early Hematopoietic and Erythroid Hierarchies , 2018, Nature.

[50]  Lin Ying Liu,et al.  Mepolizumab Attenuates Airway Eosinophil Numbers, but Not Their Functional Phenotype, in Asthma , 2017, American journal of respiratory and critical care medicine.

[51]  B. Lambrecht,et al.  Personalized medicine with biologics for severe type 2 asthma: current status and future prospects , 2017, mAbs.

[52]  T. Marichal,et al.  Homeostatic Eosinophils: Characteristics and Functions , 2017, Front. Med..

[53]  W. Busse,et al.  Asthma Exacerbations: Pathogenesis, Prevention, and Treatment , 2017, The Journal of Allergy and Clinical Immunology: In Practice.

[54]  G. Brusselle,et al.  Reslizumab in patients with inadequately controlled late-onset asthma and elevated blood eosinophils. , 2017, Pulmonary pharmacology & therapeutics.

[55]  W. Busse,et al.  Benralizumab, an anti-interleukin-5 receptor α monoclonal antibody, as add-on treatment for patients with severe, uncontrolled, eosinophilic asthma (CALIMA): a randomised, double-blind, placebo-controlled phase 3 trial , 2016, The Lancet.

[56]  E. Bleecker,et al.  Efficacy and safety of benralizumab for patients with severe asthma uncontrolled with high-dosage inhaled corticosteroids and long-acting β2-agonists (SIROCCO): a randomised, multicentre, placebo-controlled phase 3 trial , 2016, The Lancet.

[57]  J. Corren,et al.  Phase 3 Study of Reslizumab in Patients With Poorly Controlled Asthma: Effects Across a Broad Range of Eosinophil Counts. , 2016, Chest.

[58]  C. Lemière,et al.  Reslizumab for Inadequately Controlled Asthma With Elevated Blood Eosinophil Levels: A Randomized Phase 3 Study. , 2016, Chest.

[59]  M. Thiry,et al.  Lung-resident eosinophils represent a distinct regulatory eosinophil subset. , 2016, The Journal of clinical investigation.

[60]  Christopher E Brightling,et al.  Severe eosinophilic asthma treated with mepolizumab stratified by baseline eosinophil thresholds: a secondary analysis of the DREAM and MENSA studies. , 2016, The Lancet. Respiratory medicine.

[61]  R. Sakalauskas,et al.  Eosinophils enhance WNT-5a and TGF-β1 genes expression in airway smooth muscle cells and promote their proliferation by increased extracellular matrix proteins production in asthma , 2016, BMC Pulmonary Medicine.

[62]  Alice Giustacchini,et al.  Distinct myeloid progenitor differentiation pathways identified through single cell RNA sequencing , 2016, Nature Immunology.

[63]  A. Misharin,et al.  Phenotypic plasticity and targeting of Siglec‐FhighCD11clow eosinophils to the airway in a murine model of asthma , 2016, Allergy.

[64]  N. Paul,et al.  Circulating Human Eosinophils Share a Similar Transcriptional Profile in Asthma and Other Hypereosinophilic Disorders , 2015, PloS one.

[65]  Helen K. Reddel,et al.  A summary of the new GINA strategy: a roadmap to asthma control , 2015, European Respiratory Journal.

[66]  E. Bateman,et al.  Reslizumab for inadequately controlled asthma with elevated blood eosinophil counts: results from two multicentre, parallel, double-blind, randomised, placebo-controlled, phase 3 trials. , 2015, The Lancet. Respiratory medicine.

[67]  H. Hammad,et al.  The immunology of asthma , 2014, Nature Immunology.

[68]  Paige Lacy,et al.  Eosinophil Cytokines, Chemokines, and Growth Factors: Emerging Roles in Immunity , 2014, Front. Immunol..

[69]  M. Rothenberg,et al.  IL-5 Triggers a Cooperative Cytokine Network That Promotes Eosinophil Precursor Maturation , 2014, The Journal of Immunology.

[70]  R. Louis,et al.  Importance of concomitant local and systemic eosinophilia in uncontrolled asthma , 2014, European Respiratory Journal.

[71]  E. Bleecker,et al.  International ERS/ATS guidelines on definition, evaluation and treatment of severe asthma , 2013, European Respiratory Journal.

[72]  W. Busse,et al.  Effects of benralizumab on airway eosinophils in asthmatic patients with sputum eosinophilia. , 2013, The Journal of allergy and clinical immunology.

[73]  R. Flavell,et al.  TH2, allergy and group 2 innate lymphoid cells , 2013, Nature Immunology.

[74]  M. Henket,et al.  Distribution of sputum cellular phenotype in a large asthma cohort: predicting factors for eosinophilic vs neutrophilic inflammation , 2013, BMC Pulmonary Medicine.

[75]  James J. Lee,et al.  Human versus mouse eosinophils: "that which we call an eosinophil, by any other name would stain as red". , 2012, The Journal of allergy and clinical immunology.

[76]  Ian D Pavord,et al.  Mepolizumab for severe eosinophilic asthma (DREAM): a multicentre, double-blind, placebo-controlled trial , 2012, The Lancet.

[77]  M. Bivas-Benita,et al.  CCL11 elicits secretion of RNases from mouse eosinophils and their cell‐free granules , 2012, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[78]  W. Crawford,et al.  Elevated exhaled nitric oxide is a clinical indicator of future uncontrolled asthma in asthmatic patients on inhaled corticosteroids. , 2011, The Journal of allergy and clinical immunology.

[79]  J. Connolly,et al.  Netting Neutrophils Are Major Inducers of Type I IFN Production in Pediatric Systemic Lupus Erythematosus , 2011, Science Translational Medicine.

[80]  James J. Lee,et al.  Eosinophils in health and disease: the LIAR hypothesis , 2010, Clinical and experimental allergy : journal of the British Society for Allergy and Clinical Immunology.

[81]  Barmak Modrek,et al.  T-helper type 2-driven inflammation defines major subphenotypes of asthma. , 2009, American journal of respiratory and critical care medicine.

[82]  K. Akashi,et al.  Identification of the human eosinophil lineage-committed progenitor: revision of phenotypic definition of the human common myeloid progenitor , 2009, The Journal of experimental medicine.

[83]  A. Dvorak,et al.  Mechanisms of eosinophil secretion: large vesiculotubular carriers mediate transport and release of granule‐derived cytokines and other proteins , 2008, Journal of leukocyte biology.

[84]  D. Robinson,et al.  The role of eosinophils in airway tissue remodelling in asthma. , 2007, Current opinion in immunology.

[85]  W. Busse,et al.  A study to evaluate safety and efficacy of mepolizumab in patients with moderate persistent asthma. , 2007, American journal of respiratory and critical care medicine.

[86]  R. Scott,et al.  Inflammatory subtypes in asthma: Assessment and identification using induced sputum , 2006, Respirology.

[87]  S. Peters,et al.  Airway remodeling contributes to the progressive loss of lung function in asthma: an overview. , 2005, The Journal of allergy and clinical immunology.

[88]  K. Akashi,et al.  Identification of eosinophil lineage–committed progenitors in the murine bone marrow , 2005, The Journal of experimental medicine.

[89]  E. Israel,et al.  Sputum eosinophil counts predict asthma control after discontinuation of inhaled corticosteroids. , 2005, The Journal of allergy and clinical immunology.

[90]  J. Denburg,et al.  Anti-IL-5 (mepolizumab) therapy induces bone marrow eosinophil maturational arrest and decreases eosinophil progenitors in the bronchial mucosa of atopic asthmatics. , 2003, The Journal of allergy and clinical immunology.

[91]  A. Kay,et al.  Eosinophil's role remains uncertain as anti-interleukin-5 only partially depletes numbers in asthmatic airway. , 2003, American journal of respiratory and critical care medicine.

[92]  I. Pavord,et al.  Asthma exacerbations and sputum eosinophil counts: a randomised controlled trial , 2002, The Lancet.

[93]  C. Jenkins,et al.  Predictive markers of asthma exacerbation during stepwise dose reduction of inhaled corticosteroids. , 2001, American journal of respiratory and critical care medicine.

[94]  K. Chung,et al.  Effects of an interleukin-5 blocking monoclonal antibody on eosinophils, airway hyper-responsìveness, and the late asthmatic response , 2000, The Lancet.

[95]  S. Wenzel,et al.  Evidence that severe asthma can be divided pathologically into two inflammatory subtypes with distinct physiologic and clinical characteristics. , 1999, American journal of respiratory and critical care medicine.

[96]  P. Howarth,et al.  Transforming growth factor-beta 1 in asthma. Measurement in bronchoalveolar lavage fluid. , 1997, American journal of respiratory and critical care medicine.

[97]  E. Gelfand,et al.  Interleukin-5 Expression in the Lung Epithelium of Transgenic Mice Leads to Pulmonary Changes Pathognomonic of Asthma , 1997, The Journal of experimental medicine.

[98]  L. Lichtenstein,et al.  Blood and bronchoalveolar eosinophils in allergic subjects after segmental antigen challenge: surface phenotype, density heterogeneity, and prostanoid production. , 1994, The Journal of allergy and clinical immunology.

[99]  D. Wong,et al.  Eosinophils from patients with blood eosinophilia express transforming growth factor beta 1. , 1991, Blood.

[100]  T. Fukuda,et al.  Heterogeneity of human eosinophils. , 1989, The Journal of allergy and clinical immunology.

[101]  M. Peters,et al.  Ultrastructural study of eosinophils from patients with the hypereosinophilic syndrome: a morphological basis of hypodense eosinophils. , 1988, Blood.

[102]  M. Peters,et al.  Increased numbers of hypodense eosinophils in the blood of patients with bronchial asthma. , 1985, The American review of respiratory disease.

[103]  A. Capron,et al.  Heterogeneity of human eosinophils. II. Variability of respiratory burst activity related to cell density. , 1984, Clinical and experimental immunology.

[104]  B. Chipps,et al.  Benralizumab Efficacy for Patients with Fixed Airflow Obstruction and Severe, Uncontrolled Eosinophilic Asthma. , 2019, Annals of allergy, asthma & immunology : official publication of the American College of Allergy, Asthma, & Immunology.

[105]  E. Bleecker,et al.  Predictors of enhanced response with benralizumab for patients with severe asthma: pooled analysis of the SIROCCO and CALIMA studies. , 2018, The Lancet. Respiratory medicine.

[106]  M. Rothenberg,et al.  Eosinophil Development, Disease Involvement, and Therapeutic Suppression. , 2018, Advances in immunology.

[107]  K. Chung,et al.  Precision medicine in asthma: linking phenotypes to targeted treatments , 2018, Current opinion in pulmonary medicine.

[108]  L. Swedin,et al.  Patient stratification and the unmet need in asthma. , 2017, Pharmacology & therapeutics.

[109]  Shiori Yamamoto,et al.  Eosinophil extracellular trap cell death-derived DNA traps: Their presence in secretions and functional attributes. , 2016, The Journal of allergy and clinical immunology.

[110]  S. Willsie Mepolizumab and Exacerbations of Refractory Eosinophilic Asthma , 2010 .

[111]  P. Hodgkin,et al.  IL-5-deficient mice have a developmental defect in CD5+ B-1 cells and lack eosinophilia but have normal antibody and cytotoxic T cell responses. , 1996, Immunity.