Severe eosinophilic asthma: a roadmap to consensus

Similar documents
Biologics in Asthma: Present and Future

Biologics in asthma Are we turning the corner? Roland Buhl Pulmonary Department Mainz University Hospital

Biologic Therapy in the Management of Asthma. Nabeel Farooqui, MD

Cigna Drug and Biologic Coverage Policy

Personalised medicine in asthma: time for action

Precision medicine in asthma: linking phenotypes to targeted treatments

Predicting response to triamcinolone in severe asthma by machine learning: solving the Enigma

Traiter l asthme sévère par le phénotype. Dr. Alain Michils CUB-Hôpital Erasme

Do We Need Biologics in Pediatric Asthma Management?

SEVERE ASTHMA NUCALA 100MG SUBCUTANEOUS INJECTION THE FIRST TARGETED ANTI IL-5 ADD-ON TREATMENT FOR ADULTS WITH SEVERE REFRACTORY EOSINOPHILIC ASTHMA

Disclosures. Learning Objective. Biological therapies. Biologics with action against 11/30/2011. Biologic Asthma Therapies and Individualized Medicine

Study Investigators/Centers: GSK sponsored studies MEA112997, MEA115588, and MEA and a proof of concept investigator sponsored study CRT110184

Single Technology Appraisal (STA) Benralizumab for treating severe asthma

Asma e BPCO: le strategie terapeutiche

Severe Asthma & Exacerbations: Dawn of a New Era?

Targeting Interleukin-5 in Patients With Severe Eosinophilic Asthma: A Clinical Review

Searching for Targets to Control Asthma

Pharmacy Management Drug Policy

ERJ Express. Published on December 12, 2013 as doi: /

Biologic Therapies for Treatment of Asthma Associated with Type 2 Inflammation: Effectiveness, Value, and Value-Based Price Benchmarks

Is reslizumab effective in improving quality of life and asthma control in adolescent and adult patients with poorly controlled eosinophilic asthma?

Severe Asthma(s): Can THEY be prevented or reversed?

10/24/2016. Precision Management of Severe Asthma How, When and Where. The Goals of Today s Presentation

Review. Key words : asthma, benralizumab, interleukin-5, mepolizumab, reslizumab. Introduction

Care pathways for the selection of a biologic in severe asthma

Biomarker-based corticosteroid adjustment in severe asthma: a modified Delphi consensus

Phenotypes of asthma; implications for treatment. Medical Grand Rounds Feb 2018 Jim Martin MD DSc

Asthma Hetereogeneity, Phenotypes and Endotypes Choosing the Right Biologic for your Patient

Uncontrolled Moderate-to-Severe-Asthma: Latest Data from the Floor of CHEST 2018

Jamie Lee Memorial Lecture ( ) Targets and Outcomes: Mepolizumab, Benralizumab, Reslizumab

Identifying Biologic Targets to Attenuate or Eliminate Asthma Exacerbations

Clinical Implications of Asthma Phenotypes. Michael Schatz, MD, MS Department of Allergy

HCT Medical Policy. Bronchial Thermoplasty. Policy # HCT113 Current Effective Date: 05/24/2016. Policy Statement. Overview

Monoclonal antibody therapy for severe asthma

Biologicals in the management of bronchial asthma. Deepa Shrestha

Potential new targets for drug development in severe asthma

Clinical profile of patients with adult-onset eosinophilic asthma

Add-on Therapy for Symptomatic Asthma despite Long-Acting Beta-Agonists/Inhaled Corticosteroid

Blood eosinophil count: a biomarker of an important treatable trait in patients with airway disease

Cinqair (reslizumab injection for intravenous use)

Ioana Agache Transylvania University Brasov. Precision medicine in allergy and asthma

Precision Asthma Therapy: Picking the Right Biologic for the Right Patient. The Asthma Umbrella. Asthma. Late onset. Early onset.

Biologic Agents in the treatment of Severe Asthma

Are emerging PGD2 antagonists a promising therapy class for treating asthma?

Update on Biologicals for ABPA and Asthma

LINEE GUIDA DELL ASMA: UP TO DATE

WEBINAR. Difficult-to-treat and severe asthma: changing the paradigm

Immunotherapy in Asthma Management

NG80. Asthma: diagnosis, monitoring and chronic asthma management (NG80)

SEVERE ASTHMA - EVIDENCE TABLES APPENDIX 1

Improving the diagnosis of eosinophilic asthma

What s new in Asthma? Dr Alexandra Nanzer-Kelly Consultant Respiratory Physician Royal Brompton and Harefield Hospitals

Review Article Efficacy and safety of mepolizumab in patients with severe eosinophilic asthma: a meta-analysis

SEVERE ASTHMA: not for distribution CHARACTERIZATION FOR INDIVIDUALIZED THERAPY RESPIROLOGY

Novel Biologics for Asthma: Steps on the Long Road to Therapies for Uncontrolled Asthma

Distinguishing Type-2 Asthma

Personalized medicine in childhood asthma. Dr Mariëlle Pijnenburg, Erasmus MC Sophia, Rotterdam, NL

ERJ Express. Published on August 23, 2018 as doi: /

Treatment of Severe Asthma: Biologics to Bronchial Thermoplasty. Disclosures

Managing severe asthma in adults: lessons from the ERS/ATS guidelines

Different kinds of asthma, different kinds of therapies

Prof Neil Barnes. Respiratory and General Medicine London Chest Hospital and The Royal London Hospital

Cynthia S. Kelly, M.D. Professor of Pediatrics Eastern Virginia Medical School Division Director Allergy Children s Hospital of The King s Daughters

Professor Richard Beasley

Indirect Comparison of Dupilumab and Mepolizumab Treatments for Uncontrolled Eosinophilic Asthma Bayesian Analysis of Randomized Controlled Trials

General Comments to the Author: Reviewer #1 Well done!!

Management of Severe Asthma Including Biologics and Bronchial Thermoplasty. Karen L. Gregory, DNP, APRN, CNS, RRT, AE-C, FAARC

The U.S. asthma population poses a significant burden

Asthma Upate 2018: What s New Since the 2007 Asthma Guidelines of NAEPP?

Steps to better understand severe asthma in adults

Current Asthma Therapy: Little Need to Phenotype. Phenotypes of Severe Asthma. Cellular Phenotypes 12/7/2012

Difficult Asthma Assessment: A systematic approach

5/1/18. Emerging Challenges in Primary Care: The Role of Type 2 Inflammation in Severe Asthma: Integrating Biologic Therapy to Optimize Outcomes

Rapid and Consistent Improvements in Morning PEF in Patients with Severe Eosinophilic Asthma Treated with Mepolizumab

The Pharmacist s Role in Managing Severe Asthma. This activity is supported by an educational grant from Genentech. Educational Objectives

Emerging Challenges in Primary Care: 2018 The Role of Type 2 Inflammation in Severe Asthma: Integrating Biologic Therapy to Optimize Outcomes

10801 Sixth St, Rancho Cucamonga, CA Tel (909) Fax (909) Visit our web site at:

Addressing Unmet Medical Needs in Severe Asthma: Where Do We Stand?

Technology appraisal guidance Published: 25 January 2017 nice.org.uk/guidance/ta431

Cost-effectiveness of mepolizumab (Nucala ) as an add-on treatment for severe refractory eosinophilic asthma in adult patients.

Emerging Challenges in Primary Care: The Role of Type 2 Inflammation in Severe Asthma: Integrating Biologic Therapy to Optimize Outcomes

Treatment Options for Complicated/Severe Asthma. Henry J. Kanarek, MD Kanarek Allergy Asthma Immunology

Mepolizumab: a new drug programme for patients with severe eosinophilic asthma

Omalizumab (Xolair ) ( Genentech, Inc., Novartis Pharmaceuticals Corp.) September Indication

Summary A LOOK AT BIOLOGICS FOR ASTHMA DECEMBER 2018 KEY REPORT FINDINGS WHAT IS ASTHMA? TREATMENT OPTIONS KEY POLICY RECOMMENDATIONS

Severe Asthma Reference Guide Phenotypes, Endotypes, Biomarkers, and Treatment

Subclinical phenotypes of asthma

ABCs of Immune Modifiers. Relevant Disclosures

Outcome, classification and management of wheezing in preschool children Paul L.P. Brand

Improving Outcomes in the Management & Treatment of Asthma. April 21, Spring Managed Care Forum

and one and and not any and and and

EFFECTIVE ASTHMA MANAGEMENT IN PRIMARY CARE Severity Assessment, Guidelines, and New Therapeutic Options

Which asthma patient has allergic airway inflammation?

Asthma biomarkers in the age of biologics

Rising Incidence of Asthma

New Therapies for Asthma

Asthma Therapy 2017 JOSHUA S. JACOBS, M.D.

Transcription:

EDITORIAL EOSINOPHILIC ASTHMA Severe eosinophilic asthma: a roadmap to consensus Roland Buhl 1, Marc Humbert 2, Leif Bjermer 3, Pascal Chanez 4, Liam G. Heaney 5, Ian Pavord 6, Santiago Quirce 7, Johann C. Virchow 8, Stephen Holgate 9 and the expert group of the European Consensus Meeting for Severe Eosinophilic Asthma 10 Affiliations: 1 Dept of Pulmonary Medicine, Mainz University Hospital, Mainz, Germany. 2 Dept of Respiratory Medicine, Université Paris-Sud, Paris, France. 3 Dept of Respiratory Medicine and Allergology, Skåne University Hospital, Lund, Sweden. 4 Dept of Respiratory Medicine, Aix-Marseille University, Marseille, France. 5 Dept of Respiratory Medicine, Queen s University Belfast, Belfast, UK. 6 Nuffield Dept of Medicine, University of Oxford, Oxford, UK. 7 Dept of Allergy Medicine, La Paz University Hospital, Madrid, Spain. 8 Dept of Pulmonary Medicine, University of Rostock, Rostock, Germany. 9 Dept of Clinical and Experimental Sciences, University of Southampton, Southampton, UK. 10 Details of the contributing members of the expert group of the European Consensus Meeting for Severe Eosinophilic Asthma and their affiliations can be found in the Acknowledgements section. Correspondence: S. Holgate, Southampton General Hospital (Mail Point 810, Level F, South Block), Tremona Road, Southampton, SO16 6YD, UK. E-mail: S.Holgate@soton.ac.uk @ERSpublications A task force has identified key consensus issues for defining, diagnosing and treating severe eosinophilic asthma http://ow.ly/zrvo30axywx Cite this article as: Buhl R, Humbert M, Bjermer L, et al. Severe eosinophilic asthma: a roadmap to consensus. EurRespirJ2017; 49: 1700634 [https://doi.org/10.1183/13993003.00634-2017]. Introduction Asthma is widely accepted as a complex heterogeneous condition with diverse pathophysiological mechanisms, clinical presentations, comorbidities, physiological characteristics, pathology and outcomes that is typically best managed by a multidisciplinary team [1 4]. Severe asthma is recognised as a major unmet need with a high personal and social impact, as well as a high burden on healthcare resources [4]. As a consequence of advances in the development of precision medicines for patients with severe asthma, the need to identify asthma subtypes by phenotype based on clinical, functional or inflammatory parameters is becoming a mandatory part of management [4 6]. Severe eosinophilic asthma (SEA), also referred to as severe asthma with eosinophilia, is yet to be clearly defined and warrants further discussion. It is a well-recognised but still imprecisely characterised sub-classification of severe asthma, driven by a distinct pathophysiological process involving the abnormal production of type 2 cytokines from T-helper 2 and innate lymphoid cells [3, 4, 7]. Sputum eosinophilia is found in slightly under half of all patients with asthma and both blood and sputum eosinophilia are associated with more severe disease, worse control, and worse prognosis [7]. The consequences of this persistent airway inflammation include air trapping, worsening of symptoms, frequent exacerbations and impact on quality of life [7]. Received: March 27 2017 Accepted: March 27 2017 Support statement: TEVA Pharmaceuticals Europe B.V. provided financial and logistical support for the European Consensus Meeting for Severe Eosinophilic Asthma (November 24, 2016; Amsterdam, The Netherlands) from which discussions this article is derived. In addition, TEVA provided administrative and medical writing support to the authors to prepare this manuscript through their appointed agency (Sciterion London Ltd, London, UK). Funding information for this article has been deposited with the Crossref Funder Registry Conflict of interest: Disclosures can be found alongside this article at erj.ersjournals.com Copyright ERS 2017 https://doi.org/10.1183/13993003.00634-2017 Eur Respir J 2017; 49: 1700634

Following recent advances in the mechanistic understanding of eosinophilic disease, which has allowed the introduction of a number of targeted biologic add-on treatments for SEA (licensed and in development), there is a need for guidance aimed at the treating clinician to both define SEA and to indicate by whom, when and how patients should be managed. Guidelines and clinical recommendations have yet to be updated to reflect the latest evidence on these therapies and their potential benefits for patients with SEA. As such, a robust evidence-based consensus on defining SEA in adults, as well as diagnosis, treatment, long-term management and follow-up, is now necessary. In addition, it may eventually be possible to address management of other groups with SEA, such as adolescents, children and the elderly, where there is currently less clinical experience and less trial data [8 15]. Towards a consensus on SEA In November 2016, an expert task force of clinicians with an interest in and experience of SEA met to identify the key issues and to set out a roadmap for an independent evidence-based consensus statement intended to be of practical help to colleagues in the respiratory clinic. Figure 1 describes the development process for the meeting and this roadmap editorial. Diagnosis: what does a patient with SEA look like? Early identification of patients with SEA in clinical practice is important; however, identifying these patients in day-to-day practice is not always straightforward [5, 16, 17]. A key initial step, as described by the European Respiratory Society (ERS)/American Thoracic Society (ATS) severe asthma guidelines, is to confirm the diagnosis of asthma and address comorbidities. Secondly, adherence to therapy and inhaler technique must be assessed before a diagnosis of severe uncontrolled asthma refractory to treatment can be confirmed [4]. This phenotype has been variously described in the literature and cluster analysis suggests a phenotype of patients with late-onset, eosinophilic, inflammation-predominant asthma [18]. Adult-onset asthma patients with a high blood eosinophil count ( 0.3 10 9 per L) have been found to have a distinct phenotype of severe asthma with frequent exacerbations and poor prognosis [19]. Persistent airflow limitation and distal inflammation with air trapping are common in these patients, as is upper airway pathology such as chronic rhinosinusitis with nasal polyposis [17]. The characteristics described in these and other studies are not ubiquitous in all patients (some are found in almost all patients and some in a minority). As such, classification using a set of major and minor diagnostic criteria may help to identify this pattern (table 1). Asthma patients with atopy will not necessarily have an allergic aetiology to their asthma [20]. Furthermore, it remains to be validated in atopic patients with blood eosinophilia, in the context of a thorough patient history, whether or not age at asthma onset, association between allergen exposure and asthma symptoms/ exacerbations, and presence of fixed airflow obstruction and/or upper airway comorbidities can help to differentiate between severe allergic and severe eosinophilic asthma. Late 2016 Initial discussions and formation of the initial group Late 2016 Initial review of the literature conducted Nov 2016 Kick-off meeting Diagnosis: what does a patient with SEA look like? Treatment: how do we treat patients with SEA? Monitoring and long-term follow-up: what monitoring is necessary, how do we follow up and how do we determine who has responded? Late 2017 ERS research seminar Publication of a position paper Proposal for an ERS task force Proposal for a session at a future ERS Congress FIGURE 1 The biologics consensus roadmap 2018 ERS consensus statement or guideline update for management of adults/adolescents with severe refractory asthma https://doi.org/10.1183/13993003.00634-2017 2

TABLE 1 Possible diagnostic scheme for severe eosinophilic asthma (SEA) Major criteria Diagnosis of severe asthma Evidence of high-load eosinophilic disease (persistent blood or sputum eosinophilia detected on 2 occasions) Frequent exacerbations ( 2 per year) Dependence (continuous or intermittent) on oral corticosteroids to achieve asthma control Minor criteria Late onset of disease Upper airway disease (i.e. chronic rhinosinusitis, often with nasal polyposis) Role of other biomarkers (e.g. FeNO, periostin and DPP-4) Fixed airflow obstruction Air trapping/presence of mucus plugs DPP-4: dipeptidyl peptidase-4; FeNO: exhaled nitric oxide fraction. Biomarkers have been widely studied in SEA and, in clinical practice, blood eosinophil testing is readily available. There is a consistent relationship between blood and sputum eosinophilia [21]. Evidence exists that blood eosinophilia is a superior biomarker and that those patients with raised eosinophil counts have a higher frequency of severe exacerbation, poorer disease control and show a greater response to anti-eosinophilic therapies [10, 19, 22]. There is evidence to support the predictive value of eosinophils in response to these therapies, with a range of responses at different blood eosinophil levels, and patients with higher blood eosinophil levels (e.g. 300 cells µl 1 ) tending to have a better response to treatment [14, 22]. Overall, there remains a degree of debate about the optimal use and interpretation of biomarkers for SEA in clinical practice, and this revolves around two main concerns in particular. 1. The magnitude and persistence of eosinophilic disease (the level of eosinophilia which may indicate SEA in different patients, the number of test results needed to confirm SEA, and the management of the complications arising from oral corticosteroid use masking systemic eosinophilia and delaying diagnosis). 2. The role of other biomarkers such as exhaled nitric oxide fraction (FeNO), serum periostin and dipeptidyl peptidase-4 (DPP-4). A key aim is to develop a clinically useful aid to the identification and diagnosis of patients with severe asthma where eosinophils probably play an important role in the pathophysiology of disease. Based on a system of major and minor criteria to accurately phenotype these patients, each subject will need to meet a specific number or combination of criteria in order to reach a diagnosis of SEA. Robust validation of any tools will be required to ensure applicability. Treatment: how do we treat patients with SEA? Patients with severe asthma who have an eosinophilic-driven phenotype may benefit from newly developed precision medicines. One group clearly eligible for treatment with biologics is those patients with severe asthma adherent to high-dose inhaled corticosteroids who nevertheless have frequent exacerbations with or without symptoms, and who have persistently elevated levels of circulating blood eosinophils. There are number of targeted therapies, both licensed and in development, that are appropriate for consideration in this patient population. Three biologic therapies are now licensed for severe asthma: omalizumab (anti-immunoglobulin E (IgE)), mepolizumab (anti-interleukin-5 (IL-5)) and reslizumab (anti-il-5). Four more are currently in the development process: benralizumab (anti-il-5rα) has recently reported successful phase 3 results; tralokinumab (anti-il-13) and dupilumab (anti-il-4rα) are in phase 3 trials; and pitrakinra (IL-4 and IL-13 antagonist) has completed phase 2 trials (all as of April 2017). Guidance is needed to understand how best to use these different treatments in practice, tailoring treatment choice to the clinical picture and selecting patients likely to benefit. Anti-IgE therapy may reduce airway and blood eosinophils and result in less frequent exacerbations for patients with severe persistent allergic asthma. However, the drug is approved only for patients with severe allergic asthma due to perennial allergens and not all patients with uncontrolled asthma respond to anti-ige therapy. It is not yet clear if persistent eosinophilic inflammation predicts a response to omalizumab, in particular in non-atopic asthma [5], and there is no real predictive factor available to manage this treatment [23]. However, methods such as basophil allergen threshold sensitivity may be effective at monitoring and evaluating response in different populations of patients taking omalizumab. This tool has been studied in patients with severe nut allergy to evaluate treatment efficacy, and has been explored as a biomarker in a small cohort of children with different manifestations of persistent allergic asthma [24, 25]. Interleukin-5 (IL-5) plays a critical role in eosinophil differentiation, maturation, recruitment, survival and activation in tissues. Anti-IL-5 therapies (such as mepolizumab, reslizumab and benralizumab) have all established efficacy in this patient population, have no serious safety concerns in https://doi.org/10.1183/13993003.00634-2017 3

clinical trials and have demonstrated favourable benefit risk profiles [8 12, 15, 26]. In addition, IL-4 and IL-13 play a key role in the pathogenesis of asthma. Therapies that inhibit both IL-4 and IL-13 (including dupilumab) have shown some promising results [5]. All of these biologic treatments target the lower airways and associated symptoms but also have potential benefits in terms of comorbidities. These include potentially avoiding the adverse events associated with frequent oral steroid use, the rhinosinusitis and nasal polyps associated with upper airway eosinophilia, and atopic dermatitis associated with IL-4/IL-13 activity. Could the ultimate goal of anti-eosinophilic therapy be to minimise and perhaps even eliminate oral corticosteroid use, thereby consigning maintenance oral corticosteroids to history, at least in a majority of SEA patients? There remains a need to look at how anti-eosinophilic therapies can be used to treat patients with uncontrolled disease, including children, adolescents and the elderly. In addition, patients with significant involvement of the upper airways, such as chronic rhinosinusitis with polyposis, will require a multidisciplinary team approach. Recommendations for other treatment options, including bronchial thermoplasty, macrolides and prostaglandin PGD 2 -CRTH2 antagonists, also need to be evaluated. Monitoring and follow-up When patients are prescribed a biologic therapy for SEA, a number of questions are raised. Which sort of monitoring is necessary, how should patient follow-up be conducted, and how can we determine which patients have responded to treatment? Also, under which clinical circumstances should patients stop or switch therapy [26]? Identifying therapeutic responders and non-responders is not easy, as it may take a year or more for a reduction in clinical exacerbations to emerge. Treatment goals vary and, most of the time, a combination of clinical signals is involved, including: symptom reduction (as captured by treatment effects on the individual asthma symptom load and the Asthma Control Test (ACT) or Asthma Control Questionnaire (ACQ)); reduced exacerbation rates; restoration of the sense of smell; and overall health-status restoration. This last point includes the ability to get a good night s sleep and feeling more alert (captured by Health-Related Quality of Life (HRQoL) questionnaires such as the Asthma Quality of Life Questionnaire). However, these asthma questionnaires do not always reflect this variety in treatment goals in clinical trials involving SEA. A number of elements contribute to response and individual responses will differ. Monitoring of lung function, symptoms, quality-of-life and exacerbation history may support early clinical decisions on treatment response. Improvements in comorbidities, rather than in asthma itself, may result in higher quality-of-life scores and data should be collected from both the upper and lower airways to evaluate treatment effect. Thus, it is important to conduct a careful characterisation of the symptom profile in order to have objective measures to follow when response is evaluated. Anti-eosinophilic therapies for SEA are not curative, so guidance on the switching and stopping of these therapies, along with other systemic (e.g. oral corticosteroids) and inhaled treatments, is important. The initial suggestion of the expert task force is to develop a traffic-light system to determine response. Similar to the management approach for omalizumab, it has been agreed that patients with SEA require treatment for at least 4 months before an initial assessment of response can be determined. Following this, patients who are intermediate responders should either continue treatment for a year to assess response, or be considered for a switch to an alternative anti-eosinophilic therapy if response is low. The initial proposal, which will require further development, is described in figure 2. Discussion and next steps The high clinical need in severe asthma and the introduction of effective biological therapies for severe asthma related to eosinophilia make the development of a consensus to support clinicians treating these patients an important undertaking. The ERS has expressed an interest in taking this subject forward, potentially through an ERS research seminar and through discussion in sessions at national and international congresses. The intention is to develop a position statement supported by evidence to reflect a broader expert opinion around the management of patients with severe corticosteroid-refractory asthma, and to ensure that the fast-moving scientific developments in this field are applied to clinical practice for the benefit of patients. https://doi.org/10.1183/13993003.00634-2017 4

4 months 12 months Stop treatment Non-responders Continue treatment for a year to assess response, or consider switching if response is low Intermediate responders Continue treatment Superresponders FIGURE 2 The traffic-light system for response and non-response following at least 4 months of treatment with an anti-eosinophilic therapy Acknowledgements The contents of this paper reflect the discussions between attending participants at the European Consensus Meeting for Severe Eosinophilic Asthma (November 24 2016, Amsterdam, The Netherlands) where medical attendees from TEVA Pharmaceuticals Europe B.V. were present as observers. The authors have been given full editorial control and independence and TEVA has had no influence on the contents. The following members of the expert group made important contributions at the initial meeting and/or made useful comments on earlier drafts of this editorial: Ratko Djukanovic (University of Southampton, Southampton, UK), Kian Fan Chung (Imperial College, London, UK), Walter Canonica (Humanitas University, Milan, Italy), Alberto Papi (Ferrara University, Ferrara, Italy), Chris Brightling (University of Leicester, Leicester, UK), Ildiko Horváth (Semmelweis University, Budapest, Hungary), Samantha Walker (Asthma UK, London, UK), Guy Brusselle (Ghent University Hospital, Ghent, Belgium), Vibeke Backer (Bispebjerg Hospital, Copenhagen, Denmark), Piotr Kuna (University of Lodz, Lodz, Poland), Elisabeth Bel (University of Amsterdam, Amsterdam, The Netherlands), Luis Perez de Llano (Lucus Agusti University Hospital, Lugo, Spain), and Klaus Rabe (University of Kiel, Kiel, Germany). References 1 Corren J. Asthma phenotypes and endotypes: an evolving paradigm for classification. Discov Med 2013; 15: 243 249. 2 Agusti A, Bel E, Thomas M, et al. Treatable traits: toward precision medicine of chronic airway diseases. Eur Respir J 2016; 47: 410 419. 3 Chung KF. Targeting the interleukin pathway in the treatment of asthma. Lancet 2015; 386: 1086 1096. 4 Chung KF, Wenzel SE, Brozek JL, et al. International ERS/ATS guidelines on definition, evaluation and treatment of severe asthma. Eur Respir J 2014; 43: 343 373. 5 de Groot JC, ten Brinke A, Bel EH. Management of the patient with eosinophilic asthma: a new era begins. ERJ Open Res 2015; 1: 00024-2015. 6 Castro M, Bacharier LB. Treatment for severe eosinophilic asthma-consistent effect of anti-interleukin-5 antibodies? Lancet 2016; 388: 2059 2060. 7 Schleich FN, Chevremont A, Paulus V, et al. Importance of concomitant local and systemic eosinophilia in uncontrolled asthma. Eur Respir J 2014; 44: 97 108. 8 Bleecker ER, FitzGerald JM, Chanez P, 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. Lancet 2016; 388: 2115 2127. 9 Castro M, Zangrilli J, Wechsler ME, 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. Lancet Respir Med 2015; 3: 355 366. 10 Ortega HG, Yancey SW, Mayer B, et al. Severe eosinophilic asthma treated with mepolizumab stratified by baseline eosinophil thresholds: a secondary analysis of the DREAM and MENSA studies. Lancet Respir Med 2016; 4: 549 556. 11 Pavord ID, Korn S, Howarth P, et al. Mepolizumab for severe eosinophilic asthma (DREAM): a multicentre, double-blind, placebo-controlled trial. Lancet 2012; 380: 651 659. https://doi.org/10.1183/13993003.00634-2017 5

12 FitzGerald JM, Bleecker ER, Nair P, 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. Lancet 2016; 388: 2128 2141. 13 Bel EH, Ortega HG, Pavord ID. Glucocorticoids and mepolizumab in eosinophilic asthma. N Engl J Med 2014; 371: 2433 2434. 14 Corren J, Weinstein S, Janka L, et al. Phase 3 study of reslizumab in patients with poorly controlled asthma: effects across a broad range of eosinophil counts. Chest 2016; 150: 799 810. 15 Bjermer L, Lemiere C, Maspero J, et al. Reslizumab for inadequately controlled asthma with elevated blood eosinophil levels: a randomized phase 3 study. Chest 2016; 150: 789 798. 16 Hekking PP, Wener RR, Amelink M, et al. The prevalence of severe refractory asthma. J Allergy Clin Immunol 2015; 135: 896 902. 17 de Groot JC, Storm H, Amelink M, et al. Clinical profile of patients with adult-onset eosinophilic asthma. ERJ Open Res 2016; 2: 00100-2015. 18 Haldar P, Pavord ID, Shaw DE, et al. Cluster analysis and clinical asthma phenotypes. Am J Respir Crit Care Med 2008; 178: 218 224. 19 Price DB, Rigazio A, Campbell JD, et al. Blood eosinophil count and prospective annual asthma disease burden: a UK cohort study. Lancet Respir Med 2015; 3: 849 858. 20 Arbes SJ Jr. Do all asthmatics with atopy have atopic asthma? J Allergy Clin Immunol 2012; 130: 1202 1204. 21 Wagener AH, de Nijs SB, Lutter R, et al. External validation of blood eosinophils, FE(NO) and serum periostin as surrogates for sputum eosinophils in asthma. Thorax 2015; 70: 115 120. 22 Castro M, Wenzel SE, Bleecker ER, et al. Benralizumab, an anti-interleukin 5 receptor α monoclonal antibody, versus placebo for uncontrolled eosinophilic asthma: a phase 2b randomised dose-ranging study. Lancet Respir Med 2014; 2: 879 890. 23 Froidure A, Mouthuy J, Durham SR, et al. Asthma phenotypes and IgE responses. Eur Respir J 2016; 47: 304 319. 24 Brandström J, Vetander M, Lilja G, et al. Individually dosed omalizumab: an effective treatment for severe peanut allergy. Clin Exp Allergy 2017; 47: 540 550. 25 Konradsen JR, Nordlund B, Nilsson OB, et al. High basophil allergen sensitivity (CD-sens) is associated with severe allergic asthma in children. Pediatr Allergy Immunol 2012; 23: 376 384. 26 Cabon Y, Molinari N, Marin G, et al. Comparison of anti-interleukin-5 therapies in patients with severe asthma: global and indirect meta-analyses of randomized placebo-controlled trials. Clin Exp Allergy 2017; 47: 129 138. https://doi.org/10.1183/13993003.00634-2017 6